Compositions, kits, and methods for identification, assessment, prevention and therapy of cancer

Abstract
The invention relates to compositions, kits, and methods for detecting, characterizing, preventing, and treating human cancer. A variety of chromosomal regions (MCRs) and markers corresponding thereto, are provided, wherein alterations in the copy number of one or more of the MCRs and/or alterations in the amount, structure, and/or activity of one or more of the markers is correlated with the presence of cancer.
Description
BACKGROUND OF THE INVENTION

Cancer represents the phenotypic end-point of multiple genetic lesions that endow cells with a full range of biological properties required for tumorigenesis. Indeed, a hallmark genomic feature of many cancers, including, for example, pancreatic cancer, breast cancer, ovarian cancer, and colon cancer, is the presence of numerous complex chromosome structural aberrations—including non-reciprocal translocations, amplifications and deletions.


Karyotype analyses (Johansson, B., et al. (1992) Cancer 69, 1674-81; Bardi, G., et al. (1993) Br J Cancer 67, 1106-12; Griffin, C. A., et al. (1994) Genes Chromosomes Cancer 9, 93-100; Griffin, C. A., et al. (1995) Cancer Res 55, 2394-9; Gorunova, L., et al. (1995) Genes Chromosomes Cancer 14, 259-66; Gorunova, L., et al. (1998) Genes Chromosomes Cancer 23, 81-99), chromosomal CGH and array CGH (Wolf M et al. (2004) Neoplasia 6 (3) 240; Kimura Y, et al. (2004) Mod. Pathol. 21 May (epub); Pinkel, et al. (1998) Nature Genetics 20:211; Solinas-Toldo, S., et al. (1996) Cancer Res 56, 3803-7; Mahlamaki, E. H., et al. (1997) Genes Chromosomes Cancer 20, 383-91; Mahlamaki, E. H., et al. (2002) Genes Chromosomes Cancer 35, 353-8; Fukushige, S., et al. (1997) Genes Chromosomes Cancer 19:161-9; Curtis, L. J., et al. (1998) Genomics 53, 42-55; Ghadimi, B. M., et al. (1999) Am J Pathol 154, 525-36; Armengol, G., et al. (2000) Cancer Genet Cytogenet 116, 133-41), fluorescence in situ hybridization (FISH) analysis (Nilsson M et al. (2004) Int J Cancer 109(3):363-9; Kawasaki K et al. (2003) Int J Mol. Med. 12(5):727-31) and loss of heterozygosity (LOH) mapping (Wang Z C et al. (2004) Cancer Res 64(1):64-71; Seymour, A. B., et al. (1994) Cancer Res 54, 2761-4; Hahn, S. A., et al. (1995) Cancer Res 55, 4670-5; Kimura, M., et al. (1996) Genes Chromosomes Cancer 17, 88-93) have identified recurrent regions of copy number change or allelic loss in various cancers. For example, in pancreatic cancer, frequent gains have been mapped to 3q, 5p, 7p, 8q, 11q, 12p, 17q and 20q and losses to 3p, 4q, 6q, 8p, 9p, 10q, 12q, 13q, 17p, 18q and 21q and 22q. In some instances, validated oncogenes and tumor suppressor genes residing within these loci have been identified, including MYC (8q24), pINK4A (9p21), p53 (17p13), SMAD4 (18q21) and AKT2 (19q13). However, for the majority of amplified and deleted loci and resident genes, the presumed cancer-relevant targets remain to be discovered.


SUMMARY OF THE INVENTION

The present invention is based, at least in part, on the identification of specific regions of the genome (referred to herein as minimal common regions (MCRs)), of recurrent copy number change which are contained within certain chromosomal regions (loci) and are associated with cancer. These MCRs were identified using a novel cDNA or oligomer-based platform and bioinformatics tools which allowed for the high-resolution characterization of copy-number alterations in the pancreatic adenocarcinoma genome (see Example 1). The present invention is based, also in part, on the identification of markers residing within the MCRs of the invention, which are also associated with cancer.


Accordingly, in one aspect, the present invention provides methods of assessing whether a subject is afflicted with cancer or at risk for developing cancer, comprising comparing the copy number of an MCR in a subject sample to the normal copy number of the MCR, wherein the MCR is selected from the group consisting of the MCRs listed in Table 1, and wherein an altered copy number of the MCR in the sample indicates that the subject is afflicted with cancer or at risk for developing cancer. In one embodiment, the copy number is assessed by fluorescent in situ hybridization (FISH). In another embodiment, the copy number is assessed by quantitative PCR (qPCR). In still another embodiment, the normal copy number is obtained from a control sample. In yet another embodiment, the sample is selected from the group consisting of tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue.


In another aspect, the invention provides methods of assessing whether a subject is afflicted with cancer or at risk for developing cancer comprising comparing the amount, structure, and/or activity of a marker in a subject sample, wherein the marker is a marker which resides in an MCR listed in Table 1, and the normal amount, structure, and/or activity of the marker, wherein a significant difference between the amount, structure, and/or activity of the marker in the sample and the normal amount, structure, and/or activity is an indication that the subject is afflicted with cancer or at risk for developing cancer. In one embodiment, the marker is selected from the group consisting of the markers listed in Table 4 or Table 5. In another embodiment, the amount of the marker is determined by determining the level of expression of the marker. In yet another embodiment, the level of expression of the marker in the sample is assessed by detecting the presence in the sample of a protein corresponding to the marker. The presence of the protein may be detected using a reagent which specifically binds with the protein. In one embodiment, the reagent is selected from the group consisting of an antibody, an antibody derivative, and an antibody fragment. In another embodiment, the level of expression of the marker in the sample is assessed by detecting the presence in the sample of a transcribed polynucleotide or portion thereof, wherein the transcribed polynucleotide comprises the marker. In one embodiment, the transcribed polynucleotide is an mRNA or cDNA. The level of expression of the marker in the sample may also be assessed by detecting the presence in the sample of a transcribed polynucleotide which anneals with the marker or anneals with a portion of a polynucleotide wherein the polynucleotide comprises the marker, under stringent hybridization conditions.


In another embodiment, the amount of the marker is determined by determining copy number of the marker. The copy number of the MCRs or markers may be assessed by comparative genomic hybridization (CGH), e.g., array CGH. In still another embodiment, the normal amount, structure, and/or activity is obtained from a control sample. In yet another embodiment, the sample is selected from the group consisting of tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue.


In another aspect, the invention provides methods for monitoring the progression of cancer in a subject comprising a) detecting in a subject sample at a first point in time, the amount and/or activity of a marker, wherein the marker is a marker which resides in an MCR listed in Table 1; b) repeating step a) at a subsequent point in time; and c) comparing the amount and/or activity detected in steps a) and b), and therefrom monitoring the progression of cancer in the subject. In one embodiment, the marker is selected from the group consisting of the markers listed in Table 4 or Table 5. In another embodiment, the sample is selected from the group consisting of tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue. In still another embodiment, the sample comprises cells obtained from the subject. In yet another embodiment, between the first point in time and the subsequent point in time, the subject has undergone treatment for cancer, has completed treatment for cancer, and/or is in remission.


In still another aspect, the invention provides methods of assessing the efficacy of a test compound for inhibiting cancer in a subject comprising comparing the amount and/or activity of a marker in a first sample obtained from the subject and maintained in the presence of the test compound, wherein the marker is a marker which resides in an MCR listed in Table 1, and the amount and/or activity of the marker in a second sample obtained from the subject and maintained in the absence of the test compound, wherein a significantly higher amount and/or activity of a marker in the first sample which is deleted in cancer, relative to the second sample, is an indication that the test compound is efficacious for inhibiting cancer, and wherein a significantly lower amount and/or activity of the marker in the first sample which is amplified in cancer, relative to the second sample, is an indication that the test compound is efficacious for inhibiting cancer in the subject. In one embodiment, the first and second samples are portions of a single sample obtained from the subject. In another embodiment, the first and second samples are portions of pooled samples obtained from the subject. In one embodiment, the marker is selected from the group consisting of the markers listed in Table 4 or Table 5.


In yet another aspect, the invention provides methods of assessing the efficacy of a therapy for inhibiting cancer in a subject comprising comparing the amount and/or activity of a marker in the first sample obtained from the subject prior to providing at least a portion of the therapy to the subject, wherein the marker is a marker which resides in an MCR listed in Table 1, and the amount and/or activity of the marker in a second sample obtained from the subject following provision of the portion of the therapy, wherein a significantly higher amount and/or activity of a marker in the first sample which is deleted in cancer, relative to the second sample, is an indication that the test compound is efficacious for inhibiting cancer and wherein a significantly lower amount and/or activity of a marker in the first sample which is amplified in cancer, relative to the second sample, is an indication that the therapy is efficacious for inhibiting cancer in the subject. In one embodiment, the marker is selected from the group consisting of the markers listed in Table 4 or Table 5.


Another aspect of the invention provides methods of selecting a composition capable of modulating cancer comprising obtaining a sample comprising cancer cells; contacting said cells with a test compound; and determining the ability of the test compound to modulate the amount and/or activity of a marker, wherein the marker is a marker which resides in an MCR listed in Table 1, thereby identifying a modulator of cancer. In one embodiment, the marker is selected from the group consisting of the markers listed in Table 4 or Table 5. The cells may be isolated from, e.g., an animal model of cancer, a cancer cell line, e.g., a pancreatic cancer cell line originating from a pancreatic tumor, or from a subject suffering from cancer.


Yet another aspect of the invention provides methods of selecting a composition capable of modulating cancer comprising contacting a marker with a test compound; and determining the ability of the test compound to modulate the amount and/or activity of a marker, wherein the marker is a marker which resides in an MCR listed in Table 1, thereby identifying a composition capable of modulating cancer. In one embodiment, the marker is selected from the group consisting of the markers listed in Table 4 or Table 5. In another embodiment, the method further comprises administering the test compound to an animal model of cancer. In still another embodiment, the modulator inhibits the amount and/or activity of a gene or protein corresponding to a marker set forth in Table 1 which is amplified, e.g., a marker selected from the markers listed in Table 5. In yet another embodiment, the modulator increases the amount and/or activity of a gene or protein corresponding to a marker set forth in Table 1 which is deleted, e.g., a marker selected from the markers listed in Table 4.


In another aspect, the invention provides kits for assessing the ability of a compound to inhibit cancer comprising a reagent for assessing the amount, structure, and/or activity of a marker, wherein the marker is a marker which resides in an MCR listed in Table 1. In one embodiment, the marker selected from the group consisting of the markers listed in Table 4 or Table 5.


The invention also provides kits for assessing whether a subject is afflicted with cancer comprising a reagent for assessing the copy number of an MCR selected from the group consisting of the MCRs listed in Table 1, as well as kits for assessing whether a subject is afflicted with cancer, the kit comprising a reagent for assessing the amount, structure, and/or activity of a marker. In one embodiment, the marker selected from the group consisting of the markers listed in Table 4 or Table 5.


In another aspect, the invention provides kits for assessing the presence of human cancer cells comprising an antibody or fragment thereof, wherein the antibody or fragment thereof specifically binds with a protein corresponding to a marker, wherein the marker is a marker which resides in an MCR listed in Table 1. In one embodiment, the marker selected from the group consisting of the markers listed in Table 4 or Table 5.


In still another aspect, the invention provides kits for assessing the presence of cancer cells comprising a nucleic acid probe wherein the probe specifically binds with a transcribed polynucleotide corresponding to a marker, wherein the marker is a marker which resides in an MCR listed in Table 1. In one embodiment, the marker selected from the group consisting of the markers listed in Table 4 or Table 5.


In yet another aspect, the invention provides methods of treating a subject afflicted with cancer comprising administering to the subject a modulator of the amount and/or activity of a gene or protein corresponding to a marker, wherein the marker is a marker which resides in an MCR listed in Table 1. In one embodiment, the marker selected from the group consisting of the markers listed in Table 4 or Table 5.


The invention also provides methods of treating a subject afflicted with cancer comprising administering to the subject a compound which inhibits the amount and/or activity of a gene or protein corresponding to a marker which resides in an MCR listed in Table 1 which is amplified in cancer, e.g., a marker selected from the markers listed in Table 5, thereby treating a subject afflicted with cancer. In one embodiment, the compound is administered in a pharmaceutically acceptable formulation. In another embodiment, the compound is an antibody or an antigen binding fragment thereof, which specifically binds to a protein corresponding to the marker. For example, the antibody may be conjugated to a toxin or a chemotherapeutic agent. In still another embodiment, the compound is an RNA interfering agent, e.g., an siRNA molecule or an shRNA molecule, which inhibits expression of a gene corresponding to the marker. In yet another embodiment, the compound is an antisense oligonucleotide complementary to a gene corresponding to the marker. In still another embodiment, the compound is a peptide or peptidomimetic, a small molecule which inhibits activity of the marker, e.g., a small molecule which inhibits a protein-protein interaction between a marker and a target protein, or an aptamer which inhibits expression or activity of the marker.


In another aspect, the invention provides methods of treating a subject afflicted with cancer comprising administering to the subject a compound which increases expression or activity of a gene or protein corresponding to a marker which resides in an MCR listed in Table 1 which is deleted in cancer, e.g., a marker selected from the markers listed in Table 4, thereby treating a subject afflicted with cancer. In one embodiment, the compound is a small molecule.


The invention also includes methods of treating a subject afflicted with cancer comprising administering to the subject a protein corresponding to a marker, e.g., a marker selected from the markers listed in Table 4, thereby treating a subject afflicted with cancer. In one embodiment, the protein is provided to the cells of the subject, by a vector comprising a polynucleotide encoding the protein. In still another embodiment, the compound is administered in a pharmaceutically acceptable formulation.


The present invention also provides isolated proteins, or fragments thereof, corresponding to a marker selected from the markers listed in Table 4 or Table 5.


In another aspect, the invention provides isolated nucleic acid molecules, or fragments thereof, corresponding to a marker selected from the markers listed in Table 4 or Table 5.


In still another aspect, the invention provides isolated antibodies, or fragments thereof, which specifically bind to a protein corresponding to a marker selected from the markers listed in Table 4 or Table 5.


In yet another aspect, the invention provides an isolated nucleic acid molecule, or fragment thereof, contained within an MCR selected from the MCRs listed in Table 1, wherein said nucleic acid molecule has an altered amount, structure, and/or activity in cancer. The invention also provides an isolated polypeptide encoded by the nucleic acid molecules.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-1C depict the genomic profiles from pancreatic adenocarcinoma samples. Array-CGH profiles with x-axis coordinates representing cDNA probes ordered by genomic map positions. Segmented data is displayed in red, median filtered (3 nearest neighbors) in blue and raw data in black. (1A) Whole-genome profiles of primary tumor specimen PA.T.7692 (top) and cell line Panc 10.05 (bottom). Note the presence of focal high-level amplifications and deletions as well as large regional gains and losses in both samples. (1B) Recurrence of chromosomal alterations. Top: Integer-value recurrence of CNAs in segmented data (Y-axis) plotted for each cDNA probe evenly aligned along the x-axis in genome order. Dark red or green bars denote gain or loss of chromosome material. Bright red or green bars represent probes within regions of amplification or deletion. Bottom: TreeView showing discrete CNAs within all samples. Red represents chromosomal gain and green denotes chromosomal loss. (1C) CGH profiles of 12p12.3-q13.3 locus (Locus # 15 of Table 1) in three samples illustrating the definition of the physical extent, peak profile and MCRs for that locus. Note that the left MCR is defined by the overlap between samples on top and bottom, while the right MCR is defined by the overlap between the two samples on top. Since data points are plotted on the x-axis by genomic map positions, gaps in the profiles encompass regions of copy number transition for which there is no data point.



FIGS. 2A-2B depict that QPCR verifies complexity within CNAs. (2A) Chromosome 7 CGH profiles (Left panel) showing amplification of a discrete region of 7q22 in both the AsPC-1 cell line and PA.T.14172 (Locus # 9, Table 1), with MCR defined by both samples (outlined by dashed lines). Letters A-D indicate the relative positions of QPCR assays (Right panel), which confirm the gene copy alterations in AsPC-1 (dark gray bars) and PA.T.14172 (light gray bars). (2B) Chromosome 9 array-CGH profile (Left panel) for a complex CNA in the HUP-T3 cell line. Homozygous deletion of the known target p161nk4a is confirmed by QPCR (Right panel), which also verifies the existence of two discrete, focal amplicons and a narrow region of one-copy loss revealed by array-CGH. Note that CNAs covered by only one or two probes are not identified by the segmentation algorithm.



FIGS. 3A-3C depict that combined array-CGH and expression analysis facilitates identification of candidate genes. (3A) Analysis of 17q23.2-25.3 locus (Locus #21, Table 1) in cell line Hup T3. Top panel: array-CGH profile of HUP-T3. Bottom panel: Expression profile of genes on Affymetrix U133A array within the specified locus for the HUP-T3 cell line. The subset of genes exhibiting prominent gene-dosage correlated expression fall within the peak of the locus (arrows). (3B) Analysis of 9p24.3-21.2 locus (Locus #41, Table 1) in the cell line BxPC-3. Top panel: array-CGH profile of 9p region. Bottom panel: Affymetrix™ expression profile of genes mapping to the same region. Note the dramatically reduced expression of the p16INK4A gene (arrows) within the MCR. (3C) Correlation of p16INK4A expression and copy number in 24 cell lines was analyzed. Note the bimodal distribution of both expression values and copy number values for this gene across all samples (green lines). The box outlined in dots defines those samples (BxPC-3, MiaPaCa, Capan 1, Hup-T3 and Dan-G) in which p16INK4A is homozygously deleted and not expressed. The box outlined in solid lines encloses samples (Panc-1, Panc 03.27, SW1990, Panc 08.13, Hup-T4 and Panc 02.13) in which p16INK4A is present but with absent or reduced expression.



FIG. 4 depicts a histogram of segmented profiles showing the peak at a Log2 ratio of 0 as a result of mode centering. Outer lines mark 3% (del) and 97% (amp) quantiles; inner lines mark+/−4 standard deviation of middle 50% of data.



FIG. 5 depicts a comparison of array-CGH profiles of pancreatic adenocarcinoma primary tumors and cell lines. A pseudo-karyotype presentation of an integer-value recurrence plot by chromosome is shown here for both primary tumors (PT) and cell lines (CL). Gain is shown on the right of the chromosome and loss on the left of the chromosome. Important regions of similarity between PT and CL are highlighted with boxes and chromosomes with prominent discrepancies between PT and CL are circled.



FIG. 6 depicts a whole-genome correlation of copy number and expression in the cell line Capan-1. Median filtered (width=31 probes) array-CGH and expression data are indicated in light blue and gold, respectively. Note that fluctuations in average gene expression correlate with changes in chromosome copy number (R=0.66).





DETAILED DESCRIPTION OF THE INVENTION

The present invention is based, at least in part, on the identification of specific regions of the genome (referred to herein as minimal common regions (MCRs)), of recurrent copy number change which are contained within certain chromosomal regions (loci) and are associated with cancer. These MCRs were identified using a novel cDNA or oligomer-based platform and bioinformatics tools which allowed for the high-resolution characterization of copy-number alterations in the pancreatic adenocarcinoma genome (see Example 1).


To arrive at the identified loci and MCRs, array comparative genomic hybridization (array-CGH) was utilized to define copy number aberrations (CNAs) (gains and losses of chromosomal regions) in pancreatic adenocarcinoma cell lines and tumor specimens.


Segmentation analysis of the raw profiles to filter noise from the dataset (as described by Olshen and Venkatraman, Olshen, A. B., and Venkatraman, E. S. (2002) ASA Proceedings of the Joint Statistical Meetings 2530-2535; Ginzinger, D. G. (2002) Exp Hematol 30, 503-12; Golub, T. R., et al. (1999) Science 286, 531-7; Hyman, E., et al. (2002) Cancer Res 62, 6240-5; Lucito, R., et al. (2003) Genome Res 13, 2291-305) was performed and used to identify statistically significant changepoints in the data.


Identification of loci was based on an automated computer algorithm that utilized several basic criteria as follows: 1) segments above or below certain percentiles were identified as altered; 2) if two or more altered segments were adjacent in a single profile separated by less than 500KB, the entire region spanned by the segments was considered to be an altered span; 3) highly altered segments or spans that were shorter than 20 MB were retained as “informative spans” for defining discrete locus boundaries. Longer regions were not discarded, but were not included in defining locus boundaries; 4) informative to spans were compared across samples to identify overlapping groups of positive-value or negative-value segments; each group defines a locus; and 5) MCRs were defined as contiguous spans having at least 75% of the peak recurrence as calculated by counting the occurrence of highly altered segments. If two MCRs were separated by a gap of only one probe position, they were joined. If there were more than 3 MCRs in a locus, the whole region was reported as a single complex MCR.


A locus-identification algorithm was used that defines informative CNAs on the basis of size and achievement of a high significance threshold for the amplitude of change. Overlapping CNAs from multiple profiles were then merged in an automated fashion to define a discrete “locus” of regional copy number change, the bounds of which represent the combined physical extend to these overlapping CNAs (FIG. 1C). Each locus was characterized by a peak profile, the width and amplitude of which reflect the contour of the most prominent amplification or deletion for that locus. Furthermore, within each locus, one or more minimal common regions (MCRs) were identified across multiple tumor samples (FIG. 1C), with each MCR potentially harboring a distinct cancer-relevant gene targeted for copy number alteration across the sample set.


The locus-identification algorithm defined discrete MCRs within the dataset which were annotated in terms of recurrence, amplitude of change and representation in both cell lines and primary tumors. These discrete MCRs were prioritized based on four criteria that emphasize recurrent high-threshold changes in both primary tumors and cell lines (see Example 1). Implementation of this prioritization scheme yielded 64 MCRs of the present invention within 54 independent loci, that satisfied at least three of the four criteria (see Table 1).


The confidence-level ascribed to these prioritized loci was further validated by real-time quantitative PCR (QPCR), which demonstrated 100% concordance with 16 selected MCRs defined by array-CGH. When the MCRs in Table 1 were combined with an additional 81 MCRs (within 66 distinct loci) satisfying 2 out of 4 criteria, this genomic characterization has produced a set of 145 MCRs within 121 independent loci (Table 3).


The MCRs identified herein possess a median size of 2.7 Mb, with 21 (33%) MCRs spanning 1 Mb or less (median of 0.33 Mb) and possess an average of 15 annotated genes. Table 1 lists the cytogenetic bands for each of the 54 independent loci as well as the locus boundary (Mb) and locus peak profile. The positions of each of the identified MCRs are also listed in Table 1, as well as the size and recurrence for each. For example, locus #3 represents a chromosomal region of 5q31.1-q31.1 and has a locus boundary of 133.51-134.33. This locus contains an MCR at position 133.53-133.56.


Also in Table 1, the loci and MCRs are indicated as having either “gain and amplification” or “loss and deletion,” indicating that each locus and MCR has either (1) increased copy number and/or expression or (2) decreased copy number and/or expression, or deletion, in cancer. Furthermore, genes known to play important roles in the pathogenesis of pancreatic adenocarcinoma (the p16INK4A and TP53 tumor suppressors and the MYC, KRAS2 and AKT2 oncogenes) are present within the loci and are also set forth in Table 1.


Complementary expression profile analysis of a significant fraction of the genes residing within the MCRs of the present invention provided a subset of markers with statistically significant association between gene dosage and mRNA expression. Table 4 lists the markers of the invention which reside in MCRs of deletion and which consequently display decreased expression by comparison across pancreatic cancer cell lines. Table 5 lists the markers of the invention which reside in MCRs of amplification that are overexpressed by comparison, across pancreatic cancer cell lines. Additional markers within the MCRs that have not yet been annotated may also be used as markers for cancer as described herein, and are included in the invention.


The novel methods for identifying chromosomal regions of altered copy number, as described herein, may be applied to various data sets for various diseases, including, but not limited to, cancer. Other methods may be used to determine copy number aberrations as are known in the art, including, but not limited to oligonucleotide-based microarrays (Brennan, et al. (2004) In Press; Lucito, et al. (2003) Genome Res. 13:2291-2305; Bignell et al. (2004) Genome Res. 14:287-295; Zhao, et al (2004) Cancer Research, In Press), and other methods as described herein including, for example, hybridization methods (FISH).


The amplification or deletion of the MCRs identified herein correlate with the presence of cancer, e.g., pancreatic cancer and other epithelial cancers. Furthermore, analysis of copy number and/or expression levels of the genes residing within each MCR has led to the identification of individual markers and combinations of markers described herein, the increased and decreased expression and/or increased and decreased copy number of which correlate with the presence of cancer, e.g., pancreatic cancer, e.g., in a subject.


Accordingly, methods are provided herein for detecting the presence of cancer in a sample, the absence of cancer in a sample, and other characteristics of cancer that are relevant to prevention, diagnosis, characterization, and therapy of cancer in a subject by evaluating alterations in the amount, structure, and/or activity of a marker. For example, evaluation of the presence, absence or copy number of the MCRs identified herein, or by evaluating the copy number, expression level, protein level, protein activity, presence of mutations (e.g., substitution, deletion, or addition mutations) which affect activity of the marker, or methylation status of any one or more of the markers within the MCRs (e.g., the markers set forth in Tables 4 and 5), is within the scope of the invention.


Methods are also provided herein for the identification of compounds which are capable of inhibiting cancer, in a subject, and for the treatment, prevention, and/or inhibition of cancer using a modulator, e.g., an agonist or antagonist, of a gene or protein marker of the invention.


Although the MCRs and markers described herein were identified in pancreatic cancer samples, the methods of the invention are in no way limited to use for the prevention, diagnosis, characterization, therapy and prevention of pancreatic cancer, e.g., the methods of the invention may be applied to any cancer, as described herein.


Various aspects of the invention are described in further detail in the following subsections:


1. DEFINITIONS

As used herein, each of the following terms has the meaning associated with it in this section.


The articles “a” and “an” are used herein to refer to one or to more than one (i.e. to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.


The terms “tumor” or “cancer” refer to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Cancer cells are often in the form of a tumor, but such cells may exist alone within an animal, or may be a non-tumorigenic cancer cell, such as a leukemia cell. As used herein, the term “cancer” includes premalignant as well as malignant cancers. Cancers include, but are not limited to, pancreatic cancer, e.g., pancreatic adenocarcinoma, melanomas, breast cancer, lung cancer, bronchus cancer, colorectal cancer, prostate cancer, pancreas cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, cancer of hematological tissues, and the like.


The term “pancreatic cancer” or “neoplasia” as used herein, includes PanIns, adenomas, adenocarcinomas, gastrinomas, somatostatinomas, insulinomas and glucagonomas of the pancreas.


As used herein, the term “adenocarcinoma” is carcinoma that develops in the lining or inner surface of an organ and is derived from glandular tissue or in which the tumor cells form recognizable glandular structures.


As used interchangeably herein, the terms, “pancreatic adenocarcinoma,” or “pancreatic ductal adenocarcinoma” is an adenocarcinoma of the pancreas. In one embodiment, pancreatic adenocarcinomas arise from the progression of premalignant lesions that occur in the pancreatic ducts (pancreatic intraepithelial neoplasia, referred to herein as “PanIN”). The methods described herein may be used to detect premalignant cancers, e.g., PanIns, as well as malignant cancers.


A “minimal common region (MCR),” as used herein, refers to a contiguous chromosomal region which displays either gain and amplification (increased copy number) or loss and deletion (decreased copy number) in the genome of a cancer. An MCR includes at least one nucleic acid sequence which has increased or decreased copy number and which is associated with a cancer. The MCRs of the instant invention include, but are not limited to, those set forth in Table 1.


A “marker” is a gene or protein which may be altered, wherein said alteration is associated with cancer. The alteration may be in amount, structure, and/or activity in a cancer tissue or cancer cell, as compared to its amount, structure, and/or activity, in a normal or healthy tissue or cell (e.g., a control), and is associated with a disease state, such as cancer. For example, a marker of the invention which is associated with cancer may have altered copy number, expression level, protein level, protein activity, or methylation status, in a cancer tissue or cancer cell as compared to a normal, healthy tissue or cell. Furthermore, a “marker” includes a molecule whose structure is altered, e.g., mutated (contains an allelic variant), e.g., differs from the wild type sequence at the nucleotide or amino acid level, e.g., by substitution, deletion, or addition, when present in a tissue or cell associated with a disease state, such as cancer.


The term “altered amount” of a marker or “altered level” of a marker refers to increased or decreased copy number of a marker or chromosomal region, e.g., MCR, and/or increased or decreased expression level of a particular marker gene or genes in a cancer sample, as compared to the expression level or copy number of the marker in a control sample. The term “altered amount” of a marker also includes an increased or decreased protein level of a marker in a sample, e.g., a cancer sample, as compared to the protein level of the marker in a normal, control sample. Furthermore, an altered amount of a marker may be determined by detecting the methylation status of a marker, as described herein, which may affect the expression or activity of a marker.


The amount of a marker, e.g., expression or copy number of a marker or MCR, or protein level of a marker, in a subject is “significantly” higher or lower than the normal amount of a marker or MCR, if the amount of the marker is greater or less, respectively, than the normal level by an amount greater than the standard error of the assay employed to assess amount, and preferably at least twice, and more preferably three, four, five, ten or more times that amount. Alternately, the amount of the marker or MCR in the subject can be considered “significantly” higher or lower than the normal amount if the amount is at least about two, and preferably at least about three, four, or five times, higher or lower, respectively, than the normal amount of the marker or MCR.


The “copy number of a gene” or the “copy number of a marker” refers to the number of DNA sequences in a cell encoding a particular gene product. Generally, for a given gene, a mammal has two copies of each gene. The copy number can be increased, however, by gene amplification or duplication, or reduced by deletion.


The “normal” copy number of a marker or MCR or “normal” level of expression of a marker is the level of expression, copy number of the marker, or copy number of the MCR, in a biological sample, e.g., a sample containing tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue, from a subject, e.g. a human, not afflicted with cancer.


The term “altered level of expression” of a marker or MCR refers to an expression level or copy number of a marker in a test sample e.g., a sample derived from a patient suffering from cancer, that is greater or less than the standard error of the assay employed to assess expression or copy number, and is preferably at least twice, and more preferably three, four, five or ten or more times the expression level or copy number of the marker or MCR in a control sample (e.g., sample from a healthy subjects not having the associated disease) and preferably, the average expression level or copy number of the marker or MCR in several control samples. The altered level of expression is greater or less than the standard error of the assay employed to assess expression or copy number, and is preferably at least twice, and more preferably three, four, five or ten or more times the expression level or copy number of the marker or MCR in a control sample (e.g., sample from a healthy subjects not having the associated disease) and preferably, the average expression level or copy number of the marker or MCR in several control samples.


An “overexpression” or “significantly higher level of expression or copy number” of a marker or MCR refers to an expression level or copy number in a test sample that is greater than the standard error of the assay employed to assess expression or copy number, and is preferably at least twice, and more preferably three, four, five or ten or more times the expression level or copy number of the marker or MCR in a control sample (e.g., sample from a healthy subject not afflicted with cancer) and preferably, the average expression level or copy number of the marker or MCR in several control samples.


“Methylation status” of a marker refers to the methylation pattern, e.g., methylation of the promoter of the marker, and/or methylation levels of the marker. DNA methylation is a heritable, reversible and epigenetic change. Yet, DNA methylation has the potential to alter gene expression, which has developmental and genetic consequences. DNA methylation has been linked to cancer, as described in, for example, Laird, et al. (1994) Human Molecular Genetics 3:1487-1495 and Laird, P. (2003) Nature 3:253-266, the contents of which are incorporated herein by reference. For example, methylation of CpG oligonucleotides in the promoters of tumor suppressor genes can lead to their inactivation. In addition, alterations in the normal methylation process are associated with genomic instability (Lengauer et al. Proc. Natl. Acad. Sci. USA 94:2545-2550, 1997). Such abnormal epigenetic changes may be found in many types of cancer and can, therefore, serve as potential markers for oncogenic transformation.


Methods for determining methylation include restriction landmark genomic scanning (Kawai et al., Mol. Cell. Biol. 14:7421-7427, 1994), methylation-sensitive arbitrarily primed PCR (Gonzalgo et al., Cancer Res. 57:594-599, 1997); digestion of genomic DNA with methylation-sensitive restriction enzymes followed by Southern analysis of the regions of interest (digestion-Southern method); PCR-based process that involves digestion of genomic DNA with methylation-sensitive restriction enzymes prior to PCR amplification (Singer-Sam et al., Nucl. Acids Res. 18:687, 1990); genomic sequencing using bisulfite treatment (Frommer et al., Proc. Natl. Acad. Sci. USA 89:1827-1831, 1992); methylation-specific PCR (MSP) (Herman et al. Proc. Natl. Acad. Sci. USA 93:9821-9826, 1992); and restriction enzyme digestion of PCR products amplified from bisulfite-converted DNA (Sadri and Hornsby, Nucl. Acids Res. 24:5058-5059, 1996; and Xiong and Laird, Nucl. Acids. Res. 25:2532-2534, 1997); PCR techniques for detection of gene mutations (Kuppuswamyet et al., Proc. Natl. Acad. Sci. USA 88:1143-1147, 1991) and quantitation of allelic-specific expression (Szabo and Mann, Genes Dev. 9:3097-3108, 1995; and Singer-Sam et al., PCR Methods Appl. 1:160-163, 1992); and methods described in U.S. Pat. No. 6,251,594, the contents of which are incorporated herein by reference. An integrated genomic and epigenomic analysis as described in Zardo, et al. (2000) Nature Genetics 32:453-458, may also be used.


The term “altered activity” of a marker refers to an activity of a marker which is increased or decreased in a disease state, e.g., in a cancer sample, as compared to the activity of the marker in a normal, control sample. Altered activity of a marker may be the result of, for example, altered expression of the marker, altered protein level of the marker, altered structure of the marker, or, e.g., an altered interaction with other proteins involved in the same or different pathway as the marker or altered interaction with transcriptional activators or inhibitors, or altered methylation status.


The term “altered structure” of a marker refers to the presence of mutations or allelic variants within the marker gene or maker protein, e.g., mutations which affect expression or activity of the marker, as compared to the normal or wild-type gene or protein. For example, mutations include, but are not limited to substitutions, deletions, or addition mutations. Mutations may be present in the coding or non-coding region of the marker.


A “marker nucleic acid” is a nucleic acid (e.g., DNA, mRNA, cDNA) encoded by or corresponding to a marker of the invention. For example, such marker nucleic acid molecules include DNA (e.g., cDNA) comprising the entire or a partial sequence of any of the nucleic acid sequences set forth in Tables 4 or 5 or the complement or hybridizing fragment of such a sequence. The marker nucleic acid molecules also include RNA comprising the entire or a partial sequence of any of the nucleic acid sequences set forth in Tables 4 or 5 or the complement of such a sequence, wherein all thymidine residues are replaced with uridine residues. A “marker protein” is a protein encoded by or corresponding to a marker of the invention. A marker protein comprises the entire or a partial sequence of a protein encoded by any of the sequences set forth in Tables 4 or 5 or a fragment thereof. The terms “protein” and “polypeptide” are used interchangeably herein.


A “marker,” as used herein, includes any nucleic acid sequence present in an MCR as set forth in Table 1, or a protein encoded by such a sequence.


Markers identified herein include diagnostic and therapeutic markers. A single marker may be a diagnostic marker, a therapeutic marker, or both a diagnostic and therapeutic marker.


As used herein, the term “therapeutic marker” includes markers, e.g., markers set forth in Tables 4 and 5, which are believed to be involved in the development (including maintenance, progression, angiogenesis, and/or metastasis) of cancer. The cancer-related functions of a therapeutic marker may be confirmed by, e.g., (1) increased or decreased copy number (by, e.g., fluorescence in situ hybridization (FISH) or quantitative PCR (qPCR)) or mutation (e.g., by sequencing), overexpression or underexpression (e.g., by in situ hybridization (ISH), Northern Blot, or qPCR), increased or decreased protein levels (e.g., by immunohistochemistry (IHC)), or increased or decreased protein activity (determined by, for example, modulation of a pathway in which the marker is involved), e.g., in more than about 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 20%, 25%, or more of human cancers; (2) the inhibition of cancer cell proliferation and growth, e.g., in soft agar, by, e.g., RNA interference (“RNAi”) of the marker; (3) the ability of the marker to enhance transformation of mouse embryo fibroblasts (MEFs) by oncogenes, e.g., Myc and RAS, or by RAS alone; (4) the ability of the marker to enhance or decrease the growth of tumor cell lines, e.g., in soft agar; (5) the ability of the marker to transform primary mouse cells in SCID explant; and/or; (6) the prevention of maintenance or formation of tumors, e.g., tumors arising de novo in an animal or tumors derived from human cancer cell lines, by inhibiting or activating the marker. In one embodiment, a therapeutic marker may be used as a diagnostic marker.


As used herein, the term “diagnostic marker” includes markers, e.g., markers set forth in Tables 4 and 5, which are useful in the diagnosis of cancer, e.g., over- or under-activity emergence, expression, growth, remission, recurrence or resistance of tumors before, during or after therapy. The predictive functions of the marker may be confirmed by, e.g., (1) increased or decreased copy number (e.g., by FISH or qPCR), overexpression or underexpression (e.g., by ISH, Northern Blot, or qPCR), increased or decreased protein level (e.g., by IHC), or increased or decreased activity (determined by, for example, modulation of a pathway in which the marker is involved), e.g., in more than about 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 20%, 25%, or more of human cancers; (2) its presence or absence in a biological sample, e.g., a sample containing tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue from a subject, e.g. a human, afflicted with cancer; (3) its presence or absence in clinical subset of patients with cancer (e.g., those responding to a particular therapy or those developing resistance). Diagnostic markers also include “surrogate markers,” e.g., markers which are indirect markers of disease progression.


The term “probe” refers to any molecule which is capable of selectively binding to a specifically intended target molecule, for example a marker of the invention. Probes can be either synthesized by one skilled in the art, or derived from appropriate biological preparations. For purposes of detection of the target molecule, probes may be specifically designed to be labeled, as described herein. Examples of molecules that can be utilized as probes include, but are not limited to, RNA, DNA, proteins, antibodies, and organic monomers.


As used herein, the term “promoter/regulatory sequence” means a nucleic acid sequence which is required for expression of a gene product operably linked to the promoter/regulatory sequence. In some instances, this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product. The promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue-specific manner.


An “RNA interfering agent” as used herein, is defined as any agent which interferes with or inhibits expression of a target gene, e.g., a marker of the invention, by RNA interference (RNAi). Such RNA interfering agents include, but are not limited to, nucleic acid molecules including RNA molecules which are homologous to the target gene, e.g., a marker of the invention, or a fragment thereof, short interfering RNA (siRNA), and small molecules which interfere with or inhibit expression of a target gene by RNA interference (RNAi).


“RNA interference (RNAi)” is an evolutionally conserved process whereby the expression or introduction of RNA of a sequence that is identical or highly similar to a target gene results in the sequence specific degradation or specific post-transcriptional gene silencing (PTGS) of messenger RNA (mRNA) transcribed from that targeted gene (see Coburn, G. and Cullen, B. (2002) J of Virology 76(18):9225), thereby inhibiting expression of the target gene. In one embodiment, the RNA is double stranded RNA (dsRNA). This process has been described in plants, invertebrates, and mammalian cells. In nature, RNAi is initiated by the dsRNA-specific endonuclease Dicer, which promotes processive cleavage of long dsRNA into double-stranded fragments termed siRNAs. siRNAs are incorporated into a protein complex that recognizes and cleaves target mRNAs. RNAi can also be initiated by introducing nucleic acid molecules, e.g., synthetic siRNAs or RNA interfering agents, to inhibit or silence the expression of target genes. As used herein, “inhibition of target gene expression” or “inhibition of marker gene expression” includes any decrease in expression or protein activity or level of the target gene (e.g., a marker gene of the invention) or protein encoded by the target gene, e.g., a marker protein of the invention. The decrease may be of at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 99% or more as compared to the expression of a target gene or the activity or level of the protein encoded by a target gene which has not been targeted by an RNA interfering agent.


“Short interfering RNA” (siRNA), also referred to herein as “small interfering RNA” is defined as an agent which functions to inhibit expression of a target gene, e.g., by RNAi. An siRNA may be chemically synthesized, may be produced by in vitro transcription, or may be produced within a host cell. In one embodiment, siRNA is a double stranded RNA (dsRNA) molecule of about 15 to about 40 nucleotides in length, preferably about 15 to about 28 nucleotides, more preferably about 19 to about 25 nucleotides in length, and more preferably about 19, 20, 21, or 22 nucleotides in length, and may contain a 3′ and/or 5′ overhang on each strand having a length of about 0, 1, 2, 3, 4, or 5 nucleotides. The length of the overhang is independent between the two strands, i.e., the length of the over hang on one strand is not dependent on the length of the overhang on the second strand. Preferably the siRNA is capable of promoting RNA interference through degradation or specific post-transcriptional gene silencing (PTGS) of the target messenger RNA (mRNA).


In another embodiment, an siRNA is a small hairpin (also called stem loop) RNA (shRNA). In one embodiment, these shRNAs are composed of a short (e.g., 19-25 nucleotide) antisense strand, followed by a 5-9 nucleotide loop, and the analogous sense strand. Alternatively, the sense strand may precede the nucleotide loop structure and the antisense strand may follow. These shRNAs may be contained in plasmids, retroviruses, and lentiviruses and expressed from, for example, the pol III U6 promoter, or another promoter (see, e.g., Stewart, et al. (2003) RNA April; 9(4):493-501 incorporated be reference herein).


RNA interfering agents, e.g., siRNA molecules, may be administered to a patient having or at risk for having cancer, to inhibit expression of a marker gene of the invention, e.g., a marker gene which is overexpressed in cancer (such as the markers listed in Table 5) and thereby treat, prevent, or inhibit cancer in the subject.


A “constitutive” promoter is a nucleotide sequence which, when operably linked with a polynucleotide which encodes or specifies a gene product, causes the gene product to be produced in a living human cell under most or all physiological conditions of the cell.


An “inducible” promoter is a nucleotide sequence which, when operably linked with a polynucleotide which encodes or specifies a gene product, causes the gene product to be produced in a living human cell substantially only when an inducer which corresponds to the promoter is present in the cell.


A “tissue-specific” promoter is a nucleotide sequence which, when operably linked with a polynucleotide which encodes or specifies a gene product, causes the gene product to be produced in a living human cell substantially only if the cell is a cell of the tissue type corresponding to the promoter.


A “transcribed polynucleotide” is a polynucleotide (e.g. an RNA, a cDNA, or an analog of one of an RNA or cDNA) which is complementary to or homologous with all or a portion of a mature RNA made by transcription of a marker of the invention and normal post-transcriptional processing (e.g. splicing), if any, of the transcript, and reverse transcription of the transcript.


“Complementary” refers to the broad concept of sequence complementarity between regions of two nucleic acid strands or between two regions of the same nucleic acid strand. It is known that an adenine residue of a first nucleic acid region is capable of forming specific hydrogen bonds (“base pairing”) with a residue of a second nucleic acid region which is antiparallel to the first region if the residue is thymine or uracil. Similarly, it is known that a cytosine residue of a first nucleic acid strand is capable of base pairing with a residue of a second nucleic acid strand which is antiparallel to the first strand if the residue is guanine. A first region of a nucleic acid is complementary to a second region of the same or a different nucleic acid if, when the two regions are arranged in an antiparallel fashion, at least one nucleotide residue of the first region is capable of base pairing with a residue of the second region. Preferably, the first region comprises a first portion and the second region comprises a second portion, whereby, when the first and second portions are arranged in an antiparallel fashion, at least about 50%, and preferably at least about 75%, at least about 90%, or at least about 95% of the nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion. More preferably, all nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion.


The terms “homology” or “identity,” as used interchangeably herein, refer to sequence similarity between two polynucleotide sequences or between two polypeptide sequences, with identity being a more strict comparison. The phrases “percent identity or homology” and “% identity or homology” refer to the percentage of sequence similarity found in a comparison of two or more polynucleotide sequences or two or more polypeptide sequences. “Sequence similarity” refers to the percent similarity in base pair sequence (as determined by any suitable method) between two or more polynucleotide sequences. Two or more sequences can be anywhere from 0-100% similar, or any integer value there between. Identity or similarity can be determined by comparing a position in each sequence that may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same nucleotide base or amino acid, then the molecules are identical at that position. A degree of similarity or identity between polynucleotide sequences is a function of the number of identical or matching nucleotides at positions shared by the polynucleotide sequences. A degree of identity of polypeptide sequences is a function of the number of identical amino acids at positions shared by the polypeptide sequences. A degree of homology or similarity of polypeptide sequences is a function of the number of amino acids at positions shared by the polypeptide sequences. The term “substantial homology,” as used herein, refers to homology of at least 50%, more preferably, 60%, 70%, 80%, 90%, 95% or more.


A marker is “fixed” to a substrate if it is covalently or non-covalently associated with the substrate such the substrate can be rinsed with a fluid (e.g. standard saline citrate, pH 7.4) without a substantial fraction of the marker dissociating from the substrate.


As used herein, a “naturally-occurring” nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g. encodes a natural protein).


Cancer is “inhibited” if at least one symptom of the cancer is alleviated, terminated, slowed, or prevented. As used herein, cancer is also “inhibited” if recurrence or metastasis of the cancer is reduced, slowed, delayed, or prevented.


A kit is any manufacture (e.g. a package or container) comprising at least one reagent, e.g. a probe, for specifically detecting a marker of the invention, the manufacture being promoted, distributed, or sold as a unit for performing the methods of the present invention.


II. Uses of the Invention

The present invention is based, in part, on the identification of chromosomal regions (MCRs) which are structurally altered leading to a different copy number in cancer cells as compared to normal (i.e. non-cancerous) cells. Furthermore, the present invention is based, in part, on the identification of markers, e.g., markers which reside in the MCRs of the invention, which have an altered amount, structure, and/or activity in cancer cells as compared to normal (i.e., non-cancerous) cells. The markers of the invention correspond to DNA, cDNA, RNA, and polypeptide molecules which can be detected in one or both of normal and cancerous cells.


The amount, structure, and/or activity, e.g., the presence, absence, copy number, expression level, protein level, protein activity, presence of mutations, e.g., mutations which affect activity of the marker (e.g., substitution, deletion, or addition mutations), and/or methylation status, of one or more of these markers in a sample, e.g., a sample containing tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue, is herein correlated with the cancerous state of the tissue. In addition, the presence, absence, and/or copy number of one or more of the MCRs of the invention in a sample is also correlated with the cancerous state of the tissue. The invention thus provides compositions, kits, and methods for assessing the cancerous state of cells (e.g. cells obtained from a non-human, cultured non-human cells, and in vivo cells) as well as methods for treatment, prevention, and/or inhibition of cancer using a modulator, e.g., an agonist or antagonist, of a marker of the invention.


The compositions, kits, and methods of the invention have the following uses, among others:

    • 1) assessing whether a subject is afflicted with cancer;
    • 2) assessing the stage of cancer in a human subject;
    • 3) assessing the grade of cancer in a subject;
    • 4) assessing the benign or malignant nature of cancer in a subject;
    • 5) assessing the metastatic potential of cancer in a subject;
    • 6) assessing the histological type of neoplasm associated with cancer in a subject;
    • 7) making antibodies, antibody fragments or antibody derivatives that are useful for treating cancer and/or assessing whether a subject is afflicted with cancer;
    • 8) assessing the presence of cancer cells;
    • 9) assessing the efficacy of one or more test compounds for inhibiting cancer in a subject;
    • 10) assessing the efficacy of a therapy for inhibiting cancer in a subject;
    • 11) monitoring the progression of cancer in a subject;
    • 12) selecting a composition or therapy for inhibiting cancer, e.g., in a subject;
    • 13) treating a subject afflicted with cancer;
    • 14) inhibiting cancer in a subject;
    • 15) assessing the carcinogenic potential of a test compound; and
    • 16) preventing the onset of cancer in a subject at risk for developing cancer.


The invention thus includes a method of assessing whether a subject is afflicted with cancer or is at risk for developing cancer. This method comprises comparing the amount, structure, and/or activity, e.g., the presence, absence, copy number, expression level, protein level, protein activity, presence of mutations, e.g., mutations which affect activity of the marker (e.g., substitution, deletion, or addition mutations), and/or methylation status, of a marker in a subject sample with the normal level. A significant difference between the amount, structure, or activity of the marker in the subject sample and the normal level is an indication that the subject is afflicted with cancer. The invention also provides a method for assessing whether a subject is afflicted with cancer or is at risk for developing cancer by comparing the level of expression of marker(s) within an MCR or copy number of an MCR in a cancer sample with the level of expression of marker(s) within an MCR or copy number of an MCR in a normal, control sample. A significant difference between the level of expression of marker(s) within an MCR or copy number of the MCR in the subject sample and the normal level is an indication that the subject is afflicted with cancer. The MCR is selected from the group consisting of those listed in Table 1.


The marker is selected from the group consisting of the markers listed in Tables 4 and 5. Table 4 lists markers which have a highly significant correlation between gene expression and gene dosage (p, 0.05). The level of expression or copy number of these markers is decreased in samples histologically identified as pancreatic cancer, e.g., pancreatic adenocarcinoma. Table 4 also lists the chromosome, physical position in Mb, Gene Weight, p-value, Affymetrix™ probe(s) number corresponding to each UniGene ID, Genebank Accession No. (i.e., “GI” number), and SEQ ID NO. for each of the markers. Although one or more molecules corresponding to the markers listed in Table 4 may have been described by others, the significance of these markers with regard to the cancerous state of cells, has not previously been identified.


Table 5 also lists markers which have a highly significant correlation between gene expression and gene dosage (p, 0.05). The level of expression or copy number of these markers is increased in samples histologically identified as pancreatic cancer, e.g., pancreatic adenocarcinoma. Table 5 also lists the chromosome, physical position in Mb, Gene Weight, p-value, Affymetrix™ probe(s) number corresponding to each UniGene ID, Genebank Accession No. (i.e., “GI” number), and SEQ ID NO. for each of these markers. Although one or more molecules corresponding to the markers listed in Table 5 may have been described by others, the significance of these markers with regard to the cancerous state of cells, has not previously been identified.


Any marker or combination of markers listed in Tables 4 or 5 or any MCR or combination of MCRs listed in Table 1, may be used in the compositions, kits, and methods of the present invention. In general, it is preferable to use markers for which the difference between the amount, e.g., level of expression or copy number, and/or activity of the marker or MCR in cancer cells and the amount, e.g., level of expression or copy number, and/or activity of the same marker in normal cells, is as great as possible. Although this difference can be as small as the limit of detection of the method for assessing amount and/or activity of the marker, it is preferred that the difference be at least greater than the standard error of the assessment method, and preferably a difference of at least 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, 10-15-, 20-, 25-, 100-, 500-, 1000-fold or greater than the amount, e.g., level of expression or copy number, and/or activity of the same biomarker in normal tissue.


It is understood that by routine screening of additional subject samples using one or more of the markers of the invention, it will be realized that certain of the markers have altered amount, structure, and/or activity in cancers of various types, including specific pancreatic cancers, as well as other cancers, e.g., carcinoma, sarcoma, lymphoma or leukemia, examples of which include, but are not limited to, melanomas, breast cancer, lung cancer, bronchus cancer, colorectal cancer, prostate cancer, pancreas cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, cancer of hematological tissues, and the like.


For example, it will be confirmed that some of the markers of the invention have altered amount, structure, and/or activity in some, i.e., 10%, 20%, 30%, or 40%, or most (i.e. 50% or more) or substantially all (i.e. 80% or more) of cancer, e.g., pancreatic cancer. Furthermore, it will be confirmed that certain of the markers of the invention are associated with cancer of various histologic subtypes.


In addition, as a greater number of subject samples are assessed for altered amount, structure, and/or activity of the markers or altered expression or copy number MCRs of the invention and the outcomes of the individual subjects from whom the samples were obtained are correlated, it will also be confirmed that markers have altered amount, structure, and/or activity of certain of the markers or altered expression or copy number of MCRs of the invention are strongly correlated with malignant cancers and that altered expression of other markers of the invention are strongly correlated with benign tumors or premalignant states. The compositions, kits, and methods of the invention are thus useful for characterizing one or more of the stage, grade, histological type, and benign/premalignant/malignant nature of cancer in subjects.


When the compositions, kits, and methods of the invention are used for characterizing one or more of the stage, grade, histological type, and benign/premalignant/malignant nature of cancer, in a subject, it is preferred that the marker or MCR or panel of markers or MCRs of the invention be selected such that a positive result is obtained in at least about 20%, and preferably at least about 40%, 60%, or 80%, and more preferably, in substantially all, subjects afflicted with cancer, of the corresponding stage, grade, histological type, or benign/premalignant/malignant nature. Preferably, the marker or panel of markers of the invention is selected such that a PPV (positive predictive value) of greater than about 10% is obtained for the general population (more preferably coupled with an assay specificity greater than 99.5%).


When a plurality of markers or MCRs of the invention are used in the compositions, kits, and methods of the invention, the amount, structure, and/or activity of each marker or level of expression or copy number can be compared with the normal amount, structure, and/or activity of each of the plurality of markers or level of expression or copy number, in non-cancerous samples of the same type, either in a single reaction mixture (i.e. using reagents, such as different fluorescent probes, for each marker) or in individual reaction mixtures corresponding to one or more of the markers or MCRs.


In one embodiment, a significantly altered amount, structure, and/or activity of more than one of the plurality of markers, or significantly altered copy number of one or more of the MCRs in the sample, relative to the corresponding normal levels, is an indication that the subject is afflicted with cancer. For example, a significantly lower copy number in the sample of each of the plurality of markers or MCRs, relative to the corresponding normal levels or copy number, is an indication that the subject is afflicted with cancer. In yet another embodiment, a significantly enhanced copy number of one or more markers or MCRs and a significantly lower level of expression or copy number of one or more markers or MCRs in a sample relative to the corresponding normal levels, is an indication that the subject is afflicted with cancer. Also, for example, a significantly enhanced copy number in the sample of each of the plurality of markers or MCRs, relative to the corresponding normal copy number, is an indication that the subject is afflicted with cancer. In yet another embodiment, a significantly enhanced copy number of one or more markers or MCRs and a significantly lower copy number of one or more markers or MCRs in a sample relative to the corresponding normal levels, is an indication that the subject is afflicted with cancer.


When a plurality of markers or MCRs are used, it is preferred that 2, 3, 4, 5, 8, 10, 12, 15, 20, 30, or 50 or more individual markers or MCRs be used or identified, wherein fewer markers or MCRs are preferred.


Only a small number of markers are known to be associated with, for example, pancreatic cancer (e.g., AKT2, p16INK4a, c-MYC, SMAD4, and TP53; Lynch, supra). These markers or other markers which are known to be associated with other types of cancer may be used together with one or more markers of the invention in, for example, a panel of markers. In addition, frequent gains have been mapped to 3q, 5p, 7p, 8q, 11q, 12p, 17q and 20q and losses to 3p, 4q, 6q, 8p, 9p, 10q, 12q, 13q, 17p, 18q and 21q and 22q in pancreatic cancer. In some instances, validated oncogenes and tumor suppressor genes residing within these loci have been identified, including MYC (8q24), p16INK4A (9p21), p53 (17p13), SMAD4 (18q21) and AKT2 (19q13). It is well known that certain types of genes, such as oncogenes, tumor suppressor genes, growth factor-like genes, protease-like genes, and protein kinase-like genes are often involved with development of cancers of various types. Thus, among the markers of the invention, use of those which correspond to proteins which resemble known proteins encoded by known oncogenes and tumor suppressor genes, and those which correspond to proteins which resemble growth factors, proteases, and protein kinases, are preferred.


It is recognized that the compositions, kits, and methods of the invention will be of particular utility to subjects having an enhanced risk of developing cancer, and their medical advisors. Subjects recognized as having an enhanced risk of developing cancer, include, for example, subjects having a familial history of cancer, subjects identified as having a mutant oncogene (i.e. at least one allele), and subjects of advancing age.


An alteration, e.g. copy number, amount, structure, and/or activity of a marker in normal (i.e. non-cancerous) human tissue can be assessed in a variety of ways. In one embodiment, the normal level of expression or copy number is assessed by assessing the level of expression and/or copy number of the marker or MCR in a portion of cells which appear to be non-cancerous and by comparing this normal level of expression or copy number with the level of expression or copy number in a portion of the cells which are suspected of being cancerous. For example, when laparoscopy or other medical procedure, reveals the presence of a tumor on one portion of an organ, the normal level of expression or copy number of a marker or MCR may be assessed using the non-affected portion of the organ, and this normal level of expression or copy number may be compared with the level of expression or copy number of the same marker in an affected portion (i.e., the tumor) of the organ. Alternately, and particularly as further information becomes available as a result of routine performance of the methods described herein, population-average values for “normal” copy number, amount, structure, and/or activity of the markers or MCRs of the invention may be used. In other embodiments, the “normal” copy number, amount, structure, and/or activity of a marker or MCR may be determined by assessing copy number, amount, structure, and/or activity of the marker or MCR in a subject sample obtained from a non-cancer-afflicted subject, from a subject sample obtained from a subject before the suspected onset of cancer in the subject, from archived subject samples, and the like.


The invention includes compositions, kits, and methods for assessing the presence of cancer cells in a sample (e.g. an archived tissue sample or a sample obtained from a subject). These compositions, kits, and methods are substantially the same as those described above, except that, where necessary, the compositions, kits, and methods are adapted for use with certain types of samples. For example, when the sample is a parafinized, archived human tissue sample, it may be necessary to adjust the ratio of compounds in the compositions of the invention, in the kits of the invention, or the methods used. Such methods are well known in the art and within the skill of the ordinary artisan.


The invention thus includes a kit for assessing the presence of cancer cells (e.g. in a sample such as a subject sample). The kit may comprise one or more reagents capable of identifying a marker or MCR of the invention, e.g., binding specifically with a nucleic acid or polypeptide corresponding to a marker or MCR of the invention. Suitable reagents for binding with a polypeptide corresponding to a marker of the invention include antibodies, antibody derivatives, antibody fragments, and the like. Suitable reagents for binding with a nucleic acid (e.g. a genomic DNA, an mRNA, a spliced mRNA, a cDNA, or the like) include complementary nucleic acids. For example, the nucleic acid reagents may include oligonucleotides (labeled or non-labeled) fixed to a substrate, labeled oligonucleotides not bound with a substrate, pairs of PCR primers, molecular beacon probes, and the like.


The kit of the invention may optionally comprise additional components useful for performing the methods of the invention. By way of example, the kit may comprise fluids (e.g., SSC buffer) suitable for annealing complementary nucleic acids or for binding an antibody with a protein with which it specifically binds, one or more sample compartments, an instructional material which describes performance of a method of the invention, a sample of normal cells, a sample of cancer cells, and the like.


A kit of the invention may comprise a reagent useful for determining protein level or protein activity of a marker. In another embodiment, a kit of the invention may comprise a reagent for determining methylation status of a marker, or may comprise a reagent for determining alteration of structure of a marker, e.g., the presence of a mutation.


The invention also includes a method of making an isolated hybridoma which produces an antibody useful in methods and kits of the present invention. A protein corresponding to a marker of the invention may be isolated (e.g. by purification from a cell in which it is expressed or by transcription and translation of a nucleic acid encoding the protein in vivo or in vitro using known methods) and a vertebrate, preferably a mammal such as a mouse, rat, rabbit, or sheep, is immunized using the isolated protein. The vertebrate may optionally (and preferably) be immunized at least one additional time with the isolated protein, so that the vertebrate exhibits a robust immune response to the protein. Splenocytes are isolated from the immunized vertebrate and fused with an immortalized cell line to form hybridomas, using any of a variety of methods well known in the art. Hybridomas formed in this manner are then screened using standard methods to identify one or more hybridomas which produce an antibody which specifically binds with the protein. The invention also includes hybridomas made by this method and antibodies made using such hybridomas.


The invention also includes a method of assessing the efficacy of a test compound for inhibiting cancer cells. As described above, differences in the amount, structure, and/or activity of the markers of the invention, or level of expression or copy number of the MCRs of the invention, correlate with the cancerous state of cells. Although it is recognized that changes in the levels of amount, e.g., expression or copy number, structure, and/or activity of certain of the markers or expression or copy number of the MCRs of the invention likely result from the cancerous state of cells, it is likewise recognized that changes in the amount may induce, maintain, and promote the cancerous state. Thus, compounds which inhibit cancer, in a subject may cause a change, e.g., a change in expression and/or activity of one or more of the markers of the invention to a level nearer the normal level for that marker (e.g., the amount, e.g., expression, and/or activity for the marker in non-cancerous cells).


This method thus comprises comparing amount, e.g., expression, and/or activity of a marker in a first cell sample and maintained in the presence of the test compound and amount, e.g., expression, and/or activity of the marker in a second cell sample and maintained in the absence of the test compound. A significant increase in the amount, e.g., expression, and/or activity of a marker listed in Table 4 (e.g., a marker that was shown to be decreased in cancer), a significant decrease in the amount, e.g., expression, and/or activity of a marker listed in Table 5 (e.g., a marker that was shown to be increased in cancer), is an indication that the test compound inhibits cancer. The cell samples may, for example, be aliquots of a single sample of normal cells obtained from a subject, pooled samples of normal cells obtained from a subject, cells of a normal cell lines, aliquots of a single sample of cancer, cells obtained from a subject, pooled samples of cancer, cells obtained from a subject, cells of a cancer cell line, cells from an animal model of cancer, or the like. In one embodiment, the samples are cancer cells obtained from a subject and a plurality of compounds known to be effective for inhibiting various cancers, are tested in order to identify the compound which is likely to best inhibit the cancer in the subject.


This method may likewise be used to assess the efficacy of a therapy, e.g., chermotherapy, radiation therapy, surgery, or any other therapeutic approach useful for inhibiting cancer in a subject. In this method, the amount, e.g., expression, and/or activity of one or more markers of the invention in a pair of samples (one subjected to the therapy, the other not subjected to the therapy) is assessed. As with the method of assessing the efficacy of test compounds, if the therapy induces a significant decrease in the amount, e.g., expression, and/or activity of a marker listed in Table 5 (e.g., a marker that was shown to be increased in cancer), blocks induction of a marker listed in Table 5 (e.g., a marker that was shown to be increased in cancer), or if the therapy induces a significant enhancement of the amount, e.g., expression, and/or activity of a marker listed in Table 4 (e.g., a marker that was shown to be decreased in cancer), then the therapy is efficacious for inhibiting cancer. As above, if samples from a selected subject are used in this method, then alternative therapies can be assessed in vitro in order to select a therapy most likely to be efficacious for inhibiting cancer in the subject.


This method may likewise be used to monitor the progression of cancer in a subject, wherein if a sample in a subject has a significant decrease in the amount, e.g., expression, and/or activity of a marker listed in Table 5 (e.g., a marker that was shown to be increased in cancer, or blocks induction of a marker listed in Table 5 (e.g., a marker that was shown to be increased in cancer), or a significant enhancement of the amount, e.g., expression, and/or activity of a marker listed in Table 4 (e.g., a marker that was shown to be decreased in cancer), during the progression of cancer, e.g., at a first point in time and a subsequent point in time, then the cancer has improved. In yet another embodiment, between the first point in time and a subsequent point in time, the subject has undergone treatment, e.g., chermotherapy, radiation therapy, surgery, or any other therapeutic approach useful for inhibiting cancer, has completed treatment, or is in remission.


As described herein, cancer in subjects is associated with an increase in amount, e.g., expression, and/or activity of one or more markers listed in Table 5 (e.g., a marker that was shown to be increased in cancer), and/or a decrease in amount, e.g., expression, and/or activity of one or more markers listed in Table 4 (e.g., a marker that was shown to be decreased in cancer). While, as discussed above, some of these changes in amount, e.g., expression, and/or activity number result from occurrence of the cancer, others of these changes induce, maintain, and promote the cancerous state of cancer cells. Thus, cancer characterized by an increase in the amount, e.g., expression, and/or activity of one or more markers listed in Table 5 (e.g., a marker that was shown to be increased in cancer), can be inhibited by inhibiting amount, e.g., expression, and/or activity of those markers. Likewise, cancer characterized by a decrease in the amount, e.g., expression, and/or activity of one or more markers listed in Table 4 (e.g., a marker that was shown to be decreased in cancer), can be inhibited by enhancing amount, e.g., expression, and/or activity of those markers.


Amount and/or activity of a marker listed in Table 5 (e.g., a marker that was shown to be increased in cancer), can be inhibited in a number of ways generally known in the art. For example, an antisense oligonucleotide can be provided to the cancer cells in order to inhibit transcription, translation, or both, of the marker(s). An RNA interfering agent, e.g., an siRNA molecule, which is targeted to a marker listed in Table 5, can be provided to the cancer cells in order to inhibit expression of the target marker, e.g., through degradation or specific post-transcriptional gene silencing (PTGS) of the messenger RNA (mRNA) of the target marker. Alternately, a polynucleotide encoding an antibody, an antibody derivative, or an antibody fragment, e.g., a fragment capable of binding an antigen, and operably linked with an appropriate promoter or regulator region, can be provided to the cell in order to generate intracellular antibodies which will inhibit the function, amount, and/or activity of the protein corresponding to the marker(s). Conjugated antibodies or fragments thereof, e.g., chemolabeled antibodies, radiolabeled antibodies, or immunotoxins targeting a marker of the invention may also be administered to treat, prevent or inhibit cancer.


A small molecule may also be used to modulate, e.g., inhibit, expression and/or activity of a marker listed in Table 5. In one embodiment, a small molecule functions to disrupt a protein-protein interaction between a marker of the invention and a target molecule or ligand, thereby modulating, e.g., increasing or decreasing the activity of the marker.


Using the methods described herein, a variety of molecules, particularly including molecules sufficiently small that they are able to cross the cell membrane, can be screened in order to identify molecules which inhibit amount and/or activity of the marker(s). The compound so identified can be provided to the subject in order to inhibit amount and/or activity of the marker(s) in the cancer cells of the subject.


Amount and/or activity of a marker listed in Table 4 (e.g., a marker that was shown to be decreased in cancer), can be enhanced in a number of ways generally known in the art. For example, a polynucleotide encoding the marker and operably linked with an appropriate promoter/regulator region can be provided to cells of the subject in order to induce enhanced expression and/or activity of the protein (and mRNA) corresponding to the marker therein. Alternatively, if the protein is capable of crossing the cell membrane, inserting itself in the cell membrane, or is normally a secreted protein, then amount and/or activity of the protein can be enhanced by providing the protein (e.g. directly or by way of the bloodstream) to cancer cells in the subject. A small molecule may also be used to modulate, e.g., increase, expression or activity of a marker listed in Table 4. Furthermore, in another embodiment, a modulator of a marker of the invention, e.g., a small molecule, may be used, for example, to re-express a silenced gene, e.g., a tumor suppressor, in order to treat or prevent cancer. For example, such a modulator may interfere with a DNA binding element or a methyltransferase.


As described above, the cancerous state of human cells is correlated with changes in the amount and/or activity of the markers of the invention. Thus, compounds which induce increased expression or activity of one or more of the markers listed in Table 5 (e.g., a marker that was shown to be increased in cancer), decreased amount and/or activity of one or more of the markers listed in Table 4 (e.g., a marker that was shown to be decreased in cancer), can induce cell carcinogenesis. The invention also includes a method for assessing the human cell carcinogenic potential of a test compound. This method comprises maintaining separate aliquots of human cells in the presence and absence of the test compound. Expression or activity of a marker of the invention in each of the aliquots is compared. A significant increase in the amount and/or activity of a marker listed in Table 5 (e.g., a marker that was shown to be increased in cancer), or a significant decrease in the amount and/or activity of a marker listed in Table 4 (e.g., a marker that was shown to be decreased in cancer), in the aliquot maintained in the presence of the test compound (relative to the aliquot maintained in the absence of the test compound) is an indication that the test compound possesses human cell carcinogenic potential. The relative carcinogenic potentials of various test compounds can be assessed by comparing the degree of enhancement or inhibition of the amount and/or activity of the relevant markers, by comparing the number of markers for which the amount and/or activity is enhanced or inhibited, or by comparing both.


Various aspects of the invention are described in further detail in the following subsections.


III. Isolated Nucleic Acid Molecules

One aspect of the invention pertains to isolated nucleic acid molecules that correspond to a marker of the invention, including nucleic acids which encode a polypeptide corresponding to a marker of the invention or a portion of such a polypeptide. The nucleic acid molecules of the invention include those nucleic acid molecules which reside in the MCRs identified herein. Isolated nucleic acid molecules of the invention also include nucleic acid molecules sufficient for use as hybridization probes to identify nucleic acid molecules that correspond to a marker of the invention, including nucleic acid molecules which encode a polypeptide corresponding to a marker of the invention, and fragments of such nucleic acid molecules, e.g., those suitable for use as PCR primers for the amplification or mutation of nucleic acid molecules. As used herein, the term “nucleic acid molecule” is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs. The nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA.


An “isolated” nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid molecule. Preferably, an “isolated” nucleic acid molecule is free of sequences (preferably protein-encoding sequences) which naturally flank the nucleic acid (i.e., sequences located at the 5′ and 3′ ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated nucleic acid molecule can contain less than about 5 kB, 4 kB, 3 kB, 2 kB, 1 kB, 0.5 kB or 0.1 kB of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived. Moreover, an “isolated” nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.


A nucleic acid molecule of the present invention, e.g., a nucleic acid molecules encoding a protein corresponding to a marker listed in Tables 4 or 5, can be isolated using standard molecular biology techniques and the sequence information in the database records described herein. Using all or a portion of such nucleic acid sequences, nucleic acid molecules of the invention can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook et al., ed., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989).


A nucleic acid molecule of the invention can be amplified using cDNA, mRNA, or genomic DNA as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid molecules so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to all or a portion of a nucleic acid molecule of the invention can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.


In another preferred embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule which has a nucleotide sequence complementary to the nucleotide sequence of a nucleic acid corresponding to a marker of the invention or to the nucleotide sequence of a nucleic acid encoding a protein which corresponds to a marker of the invention. A nucleic acid molecule which is complementary to a given nucleotide sequence is one which is sufficiently complementary to the given nucleotide sequence that it can hybridize to the given nucleotide sequence thereby forming a stable duplex.


Moreover, a nucleic acid molecule of the invention can comprise only a portion of a nucleic acid sequence, wherein the full length nucleic acid sequence comprises a marker of the invention or which encodes a polypeptide corresponding to a marker of the invention. Such nucleic acid molecules can be used, for example, as a probe or primer. The probe/primer typically is used as one or more substantially purified oligonucleotides. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 7, preferably about 15, more preferably about 25, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, or 400 or more consecutive nucleotides of a nucleic acid of the invention.


Probes based on the sequence of a nucleic acid molecule of the invention can be used to detect transcripts or genomic sequences corresponding to one or more markers of the invention. The probe comprises a label group attached thereto, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as part of a diagnostic test kit for identifying cells or tissues which mis-express the protein, such as by measuring levels of a nucleic acid molecule encoding the protein in a sample of cells from a subject, e.g., detecting mRNA levels or determining whether a gene encoding the protein has been mutated or deleted.


The invention further encompasses nucleic acid molecules that differ, due to degeneracy of the genetic code, from the nucleotide sequence of nucleic acid molecules encoding a protein which corresponds to a marker of the invention, and thus encode the same protein.


In addition to the nucleotide sequences described in Tables 4 or 5, it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequence can exist within a population (e.g., the human population). Such genetic polymorphisms can exist among individuals within a population due to natural allelic variation. An allele is one of a group of genes which occur alternatively at a given genetic locus. In addition, it will be appreciated that DNA polymorphisms that affect RNA expression levels can also exist that may affect the overall expression level of that gene (e.g., by affecting regulation or degradation).


As used herein, the phrase “allelic variant” refers to a nucleotide sequence which occurs at a given locus or to a polypeptide encoded by the nucleotide sequence.


As used herein, the terms “gene” and “recombinant gene” refer to nucleic acid molecules comprising an open reading frame encoding a polypeptide corresponding to a marker of the invention. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of a given gene. Alternative alleles can be identified by sequencing the gene of interest in a number of different individuals. This can be readily carried out by using hybridization probes to identify the same genetic locus in a variety of individuals. Any and all such nucleotide variations and resulting amino acid polymorphisms or variations that are the result of natural allelic variation and that do not alter the functional activity are intended to be within the scope of the invention.


In another embodiment, an isolated nucleic acid molecule of the invention is at least 7, 15, 20, 25, 30, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 550, 650, 700, 800, 900, 1000, 1200, 1400, 1600, 1800, 2000, 2200, 2400, 2600, 2800, 3000, 3500, 4000, 4500, or more nucleotides in length and hybridizes under stringent conditions to a nucleic acid molecule corresponding to a marker of the invention or to a nucleic acid molecule encoding a protein corresponding to a marker of the invention. As used herein, the term “hybridizes under stringent conditions” is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% (65%, 70%, preferably 75%) identical to each other typically remain hybridized to each other. Such stringent conditions are known to those skilled in the art and can be found in sections 6.3.1-6.3.6 of Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989). A preferred, non-limiting example of stringent hybridization conditions are hybridization in 6× sodium chloride/sodium citrate (SSC) at about 45° C., followed by one or more washes in 0.2×SSC, 0.1% SDS at 50-65° C.


In addition to naturally-occurring allelic variants of a nucleic acid molecule of the invention that can exist in the population, the skilled artisan will further appreciate that sequence changes can be introduced by mutation thereby leading to changes in the amino acid sequence of the encoded protein, without altering the biological activity of the protein encoded thereby. For example, one can make nucleotide substitutions leading to amino acid substitutions at “non-essential” amino acid residues. A “non-essential” amino acid residue is a residue that can be altered from the wild-type sequence without altering the biological activity, whereas an “essential” amino acid residue is required for biological activity. For example, amino acid residues that are not conserved or only semi-conserved among homologs of various species may be non-essential for activity and thus would be likely targets for alteration. Alternatively, amino acid residues that are conserved among the homologs of various species (e.g., murine and human) may be essential for activity and thus would not be likely targets for alteration.


Accordingly, another aspect of the invention pertains to nucleic acid molecules encoding a polypeptide of the invention that contain changes in amino acid residues that are not essential for activity. Such polypeptides differ in amino acid sequence from the naturally-occurring proteins which correspond to the markers of the invention, yet retain biological activity. In one embodiment, such a protein has an amino acid sequence that is at least about 40% identical, 50%, 60%, 70%, 80%, 90%, 95%, or 98% identical to the amino acid sequence of one of the proteins which correspond to the markers of the invention.


An isolated nucleic acid molecule encoding a variant protein can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of nucleic acids of the invention, such that one or more amino acid residue substitutions, additions, or deletions are introduced into the encoded protein. Mutations can be introduced by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues. A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), non-polar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Alternatively, mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity. Following mutagenesis, the encoded protein can be expressed recombinantly and the activity of the protein can be determined.


The present invention encompasses antisense nucleic acid molecules, i.e., molecules which are complementary to a sense nucleic acid of the invention, e.g., complementary to the coding strand of a double-stranded cDNA molecule corresponding to a marker of the invention or complementary to an mRNA sequence corresponding to a marker of the invention. Accordingly, an antisense nucleic acid molecule of the invention can hydrogen bond to (i.e. anneal with) a sense nucleic acid of the invention. The antisense nucleic acid can be complementary to an entire coding strand, or to only a portion thereof, e.g., all or part of the protein coding region (or open reading frame). An antisense nucleic acid molecule can also be antisense to all or part of a non-coding region of the coding strand of a nucleotide sequence encoding a polypeptide of the invention. The non-coding regions (“5′ and 3“untranslated regions”) are the 5′ and 3′ sequences which flank the coding region and are not translated into amino acids.


An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 or more nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used. Examples of modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been sub-cloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).


The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a polypeptide corresponding to a selected marker of the invention to thereby inhibit expression of the marker, e.g., by inhibiting transcription and/or translation. The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix. Examples of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site or infusion of the antisense nucleic acid into an ovary-associated body fluid. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens. The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.


An antisense nucleic acid molecule of the invention can be an α-anomeric nucleic acid molecule. An α-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual α-units, the strands run parallel to each other (Gaultier et al., 1987, Nucleic Acids Res. 15:6625-6641). The antisense nucleic acid molecule can also comprise a 2′-o-methylribonucleotide (Inoue et al., 1987, Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et al., 1987, FEBS Lett. 215:327-330).


The invention also encompasses ribozymes. Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes as described in Haselhoff and Gerlach, 1988, Nature 334:585-591) can be used to catalytically cleave mRNA transcripts to thereby inhibit translation of the protein encoded by the mRNA. A ribozyme having specificity for a nucleic acid molecule encoding a polypeptide corresponding to a marker of the invention can be designed based upon the nucleotide sequence of a cDNA corresponding to the marker. For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved (see Cech et al. U.S. Pat. No. 4,987,071; and Cech et al. U.S. Pat. No. 5,116,742). Alternatively, an mRNA encoding a polypeptide of the invention can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules (see, e.g., Bartel and Szostak, 1993, Science 261:1411-1418).


The invention also encompasses nucleic acid molecules which form triple helical structures. For example, expression of a polypeptide of the invention can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the gene encoding the polypeptide (e.g., the promoter and/or enhancer) to form triple helical structures that prevent transcription of the gene in target cells. See generally Helene (1991) Anticancer Drug Des. 6(6):569-84; Helene (1992) Ann. N.Y. Acad. Sci. 660:27-36; and Maher (1992) Bioassays 14(12):807-15.


In various embodiments, the nucleic acid molecules of the invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acid molecules can be modified to generate peptide nucleic acid molecules (see Hyrup et al., 1996, Bioorganic & Medicinal Chemistry 4(1): 5-23). As used herein, the terms “peptide nucleic acids” or “PNAs” refer to nucleic acid mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup et al. (1996), supra; Perry-O'Keefe et al. (1996) Proc. Natl. Acad. Sci. USA 93:14670-675.


PNAs can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs can also be used, e.g., in the analysis of single base pair mutations in a gene by, e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., S1 nucleases (Hyrup (1996), supra; or as probes or primers for DNA sequence and hybridization (Hyrup, 1996, supra; Perry-O'Keefe et al., 1996, Proc. Natl. Acad. Sci. USA 93:14670-675).


In another embodiment, PNAs can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras can be generated which can combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes, e.g., RNASE H and DNA polymerases, to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (Hyrup, 1996, supra). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup (1996), supra, and Finn et al. (1996) Nucleic Acids Res. 24(17):3357-63. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs. Compounds such as 5′-(4-methoxytrityl)amino-5′-deoxy-thymidine phosphoramidite can be used as a link between the PNA and the 5′ end of DNA (Mag et al., 1989, Nucleic Acids Res. 17:5973-88). PNA monomers are then coupled in a step-wise manner to produce a chimeric molecule with a 5′ PNA segment and a 3′ DNA segment (Finn et al., 1996, Nucleic Acids Res. 24(17):3357-63). Alternatively, chimeric molecules can be synthesized with a 5′ DNA segment and a 3′ PNA segment (Peterser et al., 1975, Bioorganic Med. Chem. Lett. 5:1119-11124).


In other embodiments, the oligonucleotide can include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al., 1989, Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al., 1987, Proc. Natl. Acad. Sci. USA 84:648-652; PCT Publication No. WO 88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. WO 89/10134). In addition, oligonucleotides can be modified with hybridization-triggered cleavage agents (see, e.g., Krol et al., 1988, Bio/Techniques 6:958-976) or intercalating agents (see, e.g., Zon, 1988, Pharm. Res. 5:539-549). To this end, the oligonucleotide can be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc.


The invention also includes molecular beacon nucleic acid molecules having at least one region which is complementary to a nucleic acid molecule of the invention, such that the molecular beacon is useful for quantitating the presence of the nucleic acid molecule of the invention in a sample. A “molecular beacon” nucleic acid is a nucleic acid molecule comprising a pair of complementary regions and having a fluorophore and a fluorescent quencher associated therewith. The fluorophore and quencher are associated with different portions of the nucleic acid in such an orientation that when the complementary regions are annealed with one another, fluorescence of the fluorophore is quenched by the quencher. When the complementary regions of the nucleic acid molecules are not annealed with one another, fluorescence of the fluorophore is quenched to a lesser degree. Molecular beacon nucleic acid molecules are described, for example, in U.S. Pat. No. 5,876,930.


IV. Isolated Proteins and Antibodies

One aspect of the invention pertains to isolated proteins which correspond to individual markers of the invention, and biologically active portions thereof, as well as polypeptide fragments suitable for use as immunogens to raise antibodies directed against a polypeptide corresponding to a marker of the invention. In one embodiment, the native polypeptide corresponding to a marker can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques. In another embodiment, polypeptides corresponding to a marker of the invention are produced by recombinant DNA techniques. Alternative to recombinant expression, a polypeptide corresponding to a marker of the invention can be synthesized chemically using standard peptide synthesis techniques.


An “isolated” or “purified” protein or biologically active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the protein is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized. The language “substantially free of cellular material” includes preparations of protein in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced. Thus, protein that is substantially free of cellular material includes preparations of protein having less than about 30%, 20%, 10%, or 5% (by dry weight) of heterologous protein (also referred to herein as a “contaminating protein”). When the protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, 10%, or 5% of the volume of the protein preparation. When the protein is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly such preparations of the protein have less than about 30%, 20%, 10%, 5% (by dry weight) of chemical precursors or compounds other than the polypeptide of interest.


Biologically active portions of a polypeptide corresponding to a marker of the invention include polypeptides comprising amino acid sequences sufficiently identical to or derived from the amino acid sequence of the protein corresponding to the marker (e.g., the protein encoded by the nucleic acid molecules listed in Tables 4 or 5), which include fewer amino acids than the full length protein, and exhibit at least one activity of the corresponding full-length protein. Typically, biologically active portions comprise a domain or motif with at least one activity of the corresponding protein. A biologically active portion of a protein of the invention can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acids in length. Moreover, other biologically active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of the native form of a polypeptide of the invention.


Preferred polypeptides have an amino acid sequence of a protein encoded by a nucleic acid molecule listed in Tables 4 or 5. Other useful proteins are substantially identical (e.g., at least about 40%, preferably 50%, 60%, 70%, 80%, 90%, 95%, or 99%) to one of these sequences and retain the functional activity of the protein of the corresponding naturally-occurring protein yet differ in amino acid sequence due to natural allelic variation or mutagenesis.


To determine the percent identity of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity of identical positions/total # of positions (e.g., overlapping positions)×100). In one embodiment the two sequences are the same length.


The determination of percent identity between two sequences can be accomplished using a mathematical algorithm. A preferred, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul, et al. (1990) J. Mol. Biol. 215:403-410. BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to a nucleic acid molecules of the invention. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to a protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402. Alternatively, PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules. When utilizing BLAST, Gapped BLAST, and PSI-Blast programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See http://www.ncbi.nlm.nih.gov. Another preferred, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, (1988) Comput Appl Biosci, 4:11-7. Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Yet another useful algorithm for identifying regions of local sequence similarity and alignment is the FASTA algorithm as described in Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85:2444-2448. When using the FASTA algorithm for comparing nucleotide or amino acid sequences, a PAM120 weight residue table can, for example, be used with a k-tuple value of 2.


The percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, only exact matches are counted.


The invention also provides chimeric or fusion proteins corresponding to a marker of the invention. As used herein, a “chimeric protein” or “fusion protein” comprises all or part (preferably a biologically active part) of a polypeptide corresponding to a marker of the invention operably linked to a heterologous polypeptide (i.e., a polypeptide other than the polypeptide corresponding to the marker). Within the fusion protein, the term “operably linked” is intended to indicate that the polypeptide of the invention and the heterologous polypeptide are fused in-frame to each other. The heterologous polypeptide can be fused to the amino-terminus or the carboxyl-terminus of the polypeptide of the invention.


One useful fusion protein is a GST fusion protein in which a polypeptide corresponding to a marker of the invention is fused to the carboxyl terminus of GST sequences. Such fusion proteins can facilitate the purification of a recombinant polypeptide of the invention.


In another embodiment, the fusion protein contains a heterologous signal sequence at its amino terminus. For example, the native signal sequence of a polypeptide corresponding to a marker of the invention can be removed and replaced with a signal sequence from another protein. For example, the gp67 secretory sequence of the baculovirus envelope protein can be used as a heterologous signal sequence (Ausubel et al., ed., Current Protocols in Molecular Biology, John Wiley & Sons, NY, 1992). Other examples of eukaryotic heterologous signal sequences include the secretory sequences of melittin and human placental alkaline phosphatase (Stratagene; La Jolla, Calif.). In yet another example, useful prokaryotic heterologous signal sequences include the phoA secretory signal (Sambrook et al., supra) and the protein A secretory signal (Pharmacia Biotech; Piscataway, N.J.).


In yet another embodiment, the fusion protein is an immunoglobulin fusion protein in which all or part of a polypeptide corresponding to a marker of the invention is fused to sequences derived from a member of the immunoglobulin protein family. The immunoglobulin fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject to inhibit an interaction between a ligand (soluble or membrane-bound) and a protein on the surface of a cell (receptor), to thereby suppress signal transduction in vivo. The immunoglobulin fusion protein can be used to affect the bioavailability of a cognate ligand of a polypeptide of the invention. Inhibition of ligand/receptor interaction can be useful therapeutically, both for treating proliferative and differentiative disorders and for modulating (e.g. promoting or inhibiting) cell survival. Moreover, the immunoglobulin fusion proteins of the invention can be used as immunogens to produce antibodies directed against a polypeptide of the invention in a subject, to purify ligands and in screening assays to identify molecules which inhibit the interaction of receptors with ligands.


Chimeric and fusion proteins of the invention can be produced by standard recombinant DNA techniques. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and re-amplified to generate a chimeric gene sequence (see, e.g., Ausubel et al., supra). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). A nucleic acid encoding a polypeptide of the invention can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the polypeptide of the invention.


A signal sequence can be used to facilitate secretion and isolation of the secreted protein or other proteins of interest. Signal sequences are typically characterized by a core of hydrophobic amino acids which are generally cleaved from the mature protein during secretion in one or more cleavage events. Such signal peptides contain processing sites that allow cleavage of the signal sequence from the mature proteins as they pass through the secretory pathway. Thus, the invention pertains to the described polypeptides having a signal sequence, as well as to polypeptides from which the signal sequence has been proteolytically cleaved (i.e., the cleavage products). In one embodiment, a nucleic acid sequence encoding a signal sequence can be operably linked in an expression vector to a protein of interest, such as a protein which is ordinarily not secreted or is otherwise difficult to isolate. The signal sequence directs secretion of the protein, such as from a eukaryotic host into which the expression vector is transformed, and the signal sequence is subsequently or concurrently cleaved. The protein can then be readily purified from the extracellular medium by art recognized methods. Alternatively, the signal sequence can be linked to the protein of interest using a sequence which facilitates purification, such as with a GST domain.


The present invention also pertains to variants of the polypeptides corresponding to individual markers of the invention. Such variants have an altered amino acid sequence which can function as either agonists (mimetics) or as antagonists. Variants can be generated by mutagenesis, e.g., discrete point mutation or truncation. An agonist can retain substantially the same, or a subset, of the biological activities of the naturally occurring form of the protein. An antagonist of a protein can inhibit one or more of the activities of the naturally occurring form of the protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the protein of interest. Thus, specific biological effects can be elicited by treatment with a variant of limited function. Treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein can have fewer side effects in a subject relative to treatment with the naturally occurring form of the protein.


Variants of a protein of the invention which function as either agonists (mimetics) or as antagonists can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the protein of the invention for agonist or antagonist activity. In one embodiment, a variegated library of variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential protein sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display). There are a variety of methods which can be used to produce libraries of potential variants of the polypeptides of the invention from a degenerate oligonucleotide sequence. Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang, 1983, Tetrahedron 39:3; Itakura et al., 1984, Annu. Rev. Biochem. 53:323; Itakura et al., 1984, Science 198:1056; Ike et al., 1983 Nucleic Acid Res. 11:477).


In addition, libraries of fragments of the coding sequence of a polypeptide corresponding to a marker of the invention can be used to generate a variegated population of polypeptides for screening and subsequent selection of variants. For example, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of the coding sequence of interest with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with S1 nuclease, and ligating the resulting fragment library into an expression vector. By this method, an expression library can be derived which encodes amino terminal and internal fragments of various sizes of the protein of interest.


Several techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. The most widely used techniques, which are amenable to high throughput analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a technique which enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify variants of a protein of the invention (Arkin and Yourvan, 1992, Proc. Natl. Acad. Sci. USA 89:7811-7815; Delgrave et al., 1993, Protein Engineering 6(3):327-331).


An isolated polypeptide corresponding to a marker of the invention, or a fragment thereof, can be used as an immunogen to generate antibodies using standard techniques for polyclonal and monoclonal antibody preparation. The full-length polypeptide or protein can be used or, alternatively, the invention provides antigenic peptide fragments for use as immunogens. The antigenic peptide of a protein of the invention comprises at least 8 (preferably 10, 15, 20, or 30 or more) amino acid residues of the amino acid sequence of one of the polypeptides of the invention, and encompasses an epitope of the protein such that an antibody raised against the peptide forms a specific immune complex with a marker of the invention to which the protein corresponds. Preferred epitopes encompassed by the antigenic peptide are regions that are located on the surface of the protein, e.g., hydrophilic regions. Hydrophobicity sequence analysis, hydrophilicity sequence analysis, or similar analyses can be used to identify hydrophilic regions.


An immunogen typically is used to prepare antibodies by immunizing a suitable (i.e. immunocompetent) subject such as a rabbit, goat, mouse, or other mammal or vertebrate. An appropriate immunogenic preparation can contain, for example, recombinantly-expressed or chemically-synthesized polypeptide. The preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or a similar immunostimulatory agent.


Accordingly, another aspect of the invention pertains to antibodies directed against a polypeptide of the invention. The terms “antibody” and “antibody substance” as used interchangeably herein refer to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site which specifically binds an antigen, such as a polypeptide of the invention. A molecule which specifically binds to a given polypeptide of the invention is a molecule which binds the polypeptide, but does not substantially bind other molecules in a sample, e.g., a biological sample, which naturally contains the polypeptide. Examples of immunologically active portions of immunoglobulin molecules include F(ab) and F(ab′)2 fragments which can be generated by treating the antibody with an enzyme such as pepsin. The invention provides polyclonal and monoclonal antibodies. The term “monoclonal antibody” or “monoclonal antibody composition”, as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope.


Polyclonal antibodies can be prepared as described above by immunizing a suitable subject with a polypeptide of the invention as an immunogen. The antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized polypeptide. If desired, the antibody molecules can be harvested or isolated from the subject (e.g., from the blood or serum of the subject) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction. At an appropriate time after immunization, e.g., when the specific antibody titers are highest, antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (see Kozbor et al., 1983, Immunol. Today 4:72), the EBV-hybridoma technique (see Cole et al., pp. 77-96 In Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., 1985) or trioma techniques. The technology for producing hybridomas is well known (see generally Current Protocols in Immunology, Coligan et al. ed., John Wiley & Sons, New York, 1994). Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind the polypeptide of interest, e.g., using a standard ELISA assay.


Alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal antibody directed against a polypeptide of the invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide of interest. Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAP Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Pat. No. 5,223,409; PCT Publication No. WO 92/18619; PCT Publication No. WO 91/17271; PCT Publication No. WO 92/20791; PCT Publication No. WO 92/15679; PCT Publication No. WO 93/01288; PCT Publication No. WO 92/01047; PCT Publication No. WO 92/09690; PCT Publication No. WO 90/02809; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum. Antibod. Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; Griffiths et al. (1993) EMBO J. 12:725-734.


Additionally, recombinant antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in PCT Publication No. WO 87/02671; European Patent Application 184,187; European Patent Application 171,496; European Patent Application 173,494; PCT Publication No. WO 86/01533; U.S. Pat. No. 4,816,567; European Patent Application 125,023; Better et al. (1988) Science 240:1041-1043; Liu et al. (1987) Proc. Natl. Acad. Sci. USA 84:3439-3443; Liu et al. (1987) J. Immunol. 139:3521-3526; Sun et al. (1987) Proc. Natl. Acad. Sci. USA 84:214-218; Nishimura et al. (1987) Cancer Res. 47:999-1005; Wood et al. (1985) Nature 314:446-449; and Shaw et al. (1988) J. Natl. Cancer Inst. 80:1553-1559); Morrison (1985) Science 229:1202-1207; Oi et al. (1986) Bio/Techniques 4:214; U.S. Pat. No. 5,225,539; Jones et al. (1986) Nature 321:552-525; Verhoeyan et al. (1988) Science 239:1534; and Beidler et al. (1988) J. Immunol. 141:4053-4060.


Completely human antibodies are particularly desirable for therapeutic treatment of human subjects. Such antibodies can be produced using transgenic mice which are incapable of expressing endogenous immunoglobulin heavy and light chains genes, but which can express human heavy and light chain genes. The transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide corresponding to a marker of the invention. Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology. The human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA and IgE antibodies. For an overview of this technology for producing human antibodies, see Lonberg and Huszar (1995) Int. Rev. Immunol. 13:65-93). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., U.S. Pat. No. 5,625,126; U.S. Pat. No. 5,633,425; U.S. Pat. No. 5,569,825; U.S. Pat. No. 5,661,016; and U.S. Pat. No. 5,545,806. In addition, companies such as Abgenix, Inc. (Freemont, Calif.), can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.


Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as “guided selection.” In this approach a selected non-human monoclonal antibody, e.g., a murine antibody, is used to guide the selection of a completely human antibody recognizing the same epitope (Jespers et al., 1994, Bio/technology 12:899-903).


An antibody, antibody derivative, or fragment thereof, which specifically binds a marker of the invention which is overexpressed in cancer (e.g., a marker set forth in Table 5), may be used to inhibit activity of a marker, e.g., a marker set forth in Table 5, and therefore may be administered to a subject to treat, inhibit, or prevent cancer in the subject. Furthermore, conjugated antibodies may also be used to treat, inhibit, or prevent cancer in a subject. Conjugated antibodies, preferably monoclonal antibodies, or fragments thereof, are antibodies which are joined to drugs, toxins, or radioactive atoms, and used as delivery vehicles to deliver those substances directly to cancer cells. The antibody, e.g., an antibody which specifically binds a marker of the invention (e.g., a marker listed in Table 5), is administered to a subject and binds the marker, thereby delivering the toxic substance to the cancer cell, minimizing damage to normal cells in other parts of the body.


Conjugated antibodies are also referred to as “tagged,” “labeled,” or “loaded.” Antibodies with chemotherapeutic agents attached are generally referred to as chemolabeled. Antibodies with radioactive particles attached are referred to as radiolabeled, and this type of therapy is known as radioimmunotherapy (RIT). Aside from being used to treat cancer, radiolabeled antibodies can also be used to detect areas of cancer spread in the body. Antibodies attached to toxins are called immunotoxins.


Immunotoxins are made by attaching toxins (e.g., poisonous substances from plants or bacteria) to monoclonal antibodies. Immunotoxins may be produced by attaching monoclonal antibodies to bacterial toxins such as diphtherial toxin (DT) or pseudomonal exotoxin (PE40), or to plant toxins such as ricin A or saporin.


An antibody directed against a polypeptide corresponding to a marker of the invention (e.g., a monoclonal antibody) can be used to isolate the polypeptide by standard techniques, such as affinity chromatography or immunoprecipitation. Moreover, such an antibody can be used to detect the marker (e.g., in a cellular lysate or cell supernatant) in order to evaluate the level and pattern of expression of the marker. The antibodies can also be used diagnostically to monitor protein levels in tissues or body fluids (e.g. in an ovary-associated body fluid) as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125I, 131I, 35S or 3H.


V. Recombinant Expression Vectors and Host Cells

Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding a polypeptide corresponding to a marker of the invention (or a portion of such a polypeptide). As used herein, the term “vector” refers to a to nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors, namely expression vectors, are capable of directing the expression of genes to which they are operably linked. In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids (vectors). However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.


The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell. This means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operably linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, “operably linked” is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, Methods in Enzymology: Gene Expression Technology vol. 185, Academic Press, San Diego, Calif. (1991). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, and the like. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein.


The recombinant expression vectors of the invention can be designed for expression of a polypeptide corresponding to a marker of the invention in prokaryotic (e.g., E. coli) or eukaryotic cells (e.g., insect cells {using baculovirus expression vectors}, yeast cells or mammalian cells). Suitable host cells are discussed further in Goeddel, supra. Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.


Expression of proteins in prokaryotes is most often carried out in E. coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three purposes: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson, 1988, Gene 67:31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.


Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al., 1988, Gene 69:301-315) and pET 11d (Studier et al., p. 60-89, In Gene Expression Technology Methods in Enzymology vol. 185, Academic Press, San Diego, Calif., 1991). Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid trp-lac fusion promoter. Target gene expression from the pET 11d vector relies on transcription from a T7 gn10-lac fusion promoter mediated by a co-expressed viral RNA polymerase (T7 gn1). This viral polymerase is supplied by host strains BL21(DE3) or HMS174(DE3) from a resident prophage harboring a T7 gn1 gene under the transcriptional control of the lacUV 5 promoter.


One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein (Gottesman, p. 119-128, In Gene Expression Technology: Methods in Enzymology vol. 185, Academic Press, San Diego, Calif., 1990. Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (Wada et al., 1992, Nucleic Acids Res. 20:2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.


In another embodiment, the expression vector is a yeast expression vector. Examples of vectors for expression in yeast S. cerevisiae include pYepSec1 (Baldari et al., 1987, EMBO J. 6:229-234), pMFa (Kurjan and Herskowitz, 1982, Cell 30:933-943), pJRY88 (Schultz et al., 1987, Gene 54:113-123), pYES2 (Invitrogen Corporation, San Diego, Calif.), and pPicZ (Invitrogen Corp, San Diego, Calif.).


Alternatively, the expression vector is a baculovirus expression vector. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., Sf 9 cells) include the pAc series (Smith et al., 1983, Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers, 1989, Virology 170:31-39).


In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, 1987, Nature 329:840) and pMT2PC (Kaufman et al., 1987, EMBO J. 6:187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook et al., supra.


In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al., 1987, Genes Dev. 1:268-277), lymphoid-specific promoters (Calame and Eaton, 1988, Adv. Immunol. 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore, 1989, EMBO J. 8:729-733) and immunoglobulins (Banerji et al., 1983, Cell 33:729-740; Queen and Baltimore, 1983, Cell 33:741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle, 1989, Proc. Natl. Acad. Sci. USA 86:5473-5477), pancreas-specific promoters (Edlund et al., 1985, Science 230:912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No. 4,873,316 and European Application Publication No. 264,166). Developmentally-regulated promoters are also encompassed, for example the murine hox promoters (Kessel and Gruss, 1990, Science 249:374-379) and the α-fetoprotein promoter (Camper and Tilghman, 1989, Genes Dev. 3:537-546).


The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation: That is, the DNA molecule is operably linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an RNA molecule which is antisense to the mRNA encoding a polypeptide of the invention. Regulatory sequences operably linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue-specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid, or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes see Weintraub et al., 1986, Trends in Genetics, Vol. 1 (1).


Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms “host cell” and “recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.


A host cell can be any prokaryotic (e.g., E. coli) or eukaryotic cell (e.g., insect cells, yeast or mammalian cells).


Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms “transformation” and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (supra), and other laboratory manuals.


For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., for resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Preferred selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).


A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce a polypeptide corresponding to a marker of the invention. Accordingly, the invention further provides methods for producing a polypeptide corresponding to a marker of the invention using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding a polypeptide of the invention has been introduced) in a suitable medium such that the marker is produced. In another embodiment, the method further comprises isolating the marker polypeptide from the medium or the host cell.


The host cells of the invention can also be used to produce nonhuman transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which sequences encoding a polypeptide corresponding to a marker of the invention have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous sequences encoding a marker protein of the invention have been introduced into their genome or homologous recombinant animals in which endogenous gene(s) encoding a polypeptide corresponding to a marker of the invention sequences have been altered. Such animals are useful for studying the function and/or activity of the polypeptide corresponding to the marker, for identifying and/or evaluating modulators of polypeptide activity, as well as in pre-clinical testing of therapeutics or diagnostic molecules, for marker discovery or evaluation, e.g., therapeutic and diagnostic marker discovery or evaluation, or as surrogates of drug efficacy and specificity.


As used herein, a “transgenic animal” is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, an “homologous recombinant animal” is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal. Transgenic animals also include inducible transgenic animals, such as those described in, for example, Chan I. T., et al. (2004) J Clin Invest. 113(4):528-38 and Chin L. et al (1999) Nature 400(6743):468-72.


A transgenic animal of the invention can be created by introducing a nucleic acid encoding a polypeptide corresponding to a marker of the invention into the male pronuclei of a fertilized oocyte, e.g., by microinjection, retroviral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal. Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequence(s) can be operably linked to the transgene to direct expression of the polypeptide of the invention to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866 and 4,870,009, U.S. Pat. No. 4,873,191 and in Hogan, Manipulating the Mouse Embryo, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the transgene in its genome and/or expression of mRNA encoding the transgene in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying the transgene can further be bred to other transgenic animals carrying other transgenes.


To create an homologous recombinant animal, a vector is prepared which contains at least a portion of a gene encoding a polypeptide corresponding to a marker of the invention into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the gene. In a preferred embodiment, the vector is designed such that, upon homologous recombination, the endogenous gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a “knock out” vector). Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous protein). In the homologous recombination vector, the altered portion of the gene is flanked at its 5′ and 3′ ends by additional nucleic acid of the gene to allow for homologous recombination to occur between the exogenous gene carried by the vector and an endogenous gene in an embryonic stem cell. The additional flanking nucleic acid sequences are of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5′ and 3′ ends) are included in the vector (see, e.g., Thomas and Capecchi, 1987, Cell 51:503 for a description of homologous recombination vectors). The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced gene has homologously recombined with the endogenous gene are selected (see, e.g., Li et al., 1992, Cell 69:915). The selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras (see, e.g., Bradley, Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, Robertson, Ed., IRL, Oxford, 1987, pp. 113-152). A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley (1991) Current Opinion in Bio/Technology 2:823-829 and in PCT Publication NOS. WO 90/11354, WO 91/01140, WO 92/0968, and WO 93/04169.


In another embodiment, transgenic non-human animals can be produced which contain selected systems which allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage P1. For a description of the cre/loxP recombinase system, see, e.g., Lakso et al. (1992) Proc. Natl. Acad. Sci. USA 89:6232-6236. Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al., 1991, Science 251:1351-1355). If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of “double” transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.


Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut et al. (1997) Nature 385:810-813 and PCT Publication NOS. WO 97/07668 and WO 97/07669.


VI. Methods of Treatment

The present invention provides for both prophylactic and therapeutic methods of treating a subject, e.g., a human, who has or is at risk of (or susceptible to) cancer, e.g., pancreatic cancer. As used herein, “treatment” of a subject includes the application or administration of a therapeutic agent to a subject, or application or administration of a therapeutic agent to a cell or tissue from a subject, who has a diseases or disorder, has a symptom of a disease or disorder, or is at risk of (or susceptible to) a disease or disorder, with the purpose of curing, inhibiting, healing, alleviating, relieving, altering, remedying, ameliorating, improving, or affecting the disease or disorder, the symptom of the disease or disorder, or the risk of (or susceptibility to) the disease or disorder. As used herein, a “therapeutic agent” or “compound” includes, but is not limited to, small molecules, peptides, peptidomimetics, polypeptides, RNA interfering agents, e.g., siRNA molecules, antibodies, ribozymes, and antisense oligonucleotides.


As described herein, cancer in subjects is associated with a change, e.g., an increase in the amount and/or activity, or a change in the structure, of one or more markers listed in Table 5 (e.g., a marker that was shown to be increased in cancer), and/or a decrease in the amount and/or activity, or a change in the structure of one or more markers listed in Table 4 (e.g. a marker that was shown to be decreased in cancer). While, as discussed above, some of these changes in amount, structure, and/or activity, result from occurrence of the cancer, others of these changes induce, maintain, and promote the cancerous state of cancer, cells. Thus, cancer, characterized by an increase in the amount and/or activity, or a change in the structure, of one or more markers listed in Table 5 (e.g., a marker that is shown to be increased in cancer), can be inhibited by inhibiting amount, e.g., expression or protein level, and/or activity of those markers. Likewise, cancer characterized by a decrease in the amount and/or activity, or a change in the structure, of one or more markers listed in Table 4 (e.g., a marker that is shown to be decreased in cancer), can be inhibited by enhancing amount, e.g., expression or protein level, and/or activity of those markers.


Accordingly, another aspect of the invention pertains to methods for treating a subject suffering from cancer. These methods involve administering to a subject a compound which modulates amount and/or activity of one or more markers of the invention. For example, methods of treatment or prevention of cancer include administering to a subject a compound which decreases the amount and/or activity of one or more markers listed in Table 5 (e.g., a marker that was shown to be increased in cancer). Compounds, e.g., antagonists, which may be used to inhibit amount and/or activity of a marker listed in Table 5, to thereby treat or prevent cancer include antibodies (e.g., conjugated antibodies), small molecules, RNA interfering agents, e.g., siRNA molecules, ribozymes, and antisense oligonucleotides. In one embodiment, an antibody used for treatment is conjugated to a toxin, a chemotherapeutic agent, or radioactive particles.


Methods of treatment or prevention of cancer also include administering to a subject a compound which increases the amount and/or activity of one or more markers listed in Table 4 (e.g., a marker that was shown to be decreased in cancer). Compounds, e.g., agonists, which may be used to increase expression or activity of a marker listed in Table 4, to thereby treat or prevent cancer include small molecules, peptides, peptoids, peptidomimetics, and polypeptides.


Small molecules used in the methods of the invention include those which inhibit a protein-protein interaction and thereby either increase or decrease marker amount and/or activity. Furthermore, modulators, e.g., small molecules, which cause re-expression of silenced genes, e.g., tumor suppressors, are also included herein. For example, such molecules include compounds which interfere with DNA binding or methyltransferas activity.


An aptamer may also be used to modulate, e.g., increase or inhibit expression or activity of a marker of the invention to thereby treat, prevent or inhibit cancer. Aptamers are DNA or RNA molecules that have been selected from random pools based on their ability to bind other molecules. Aptamers may be selected which bind nucleic acids or proteins.


VII. Screening Assays

The invention also provides methods (also referred to herein as “screening assays”) for identifying modulators, i.e., candidate or test compounds or agents (e.g., proteins, peptides, peptidomimetics, peptoids, small molecules or other drugs) which (a) bind to the marker, or (b) have a modulatory (e.g., stimulatory or inhibitory) effect on the activity of the marker or, more specifically, (c) have a modulatory effect on the interactions of the marker with one or more of its natural substrates (e.g., peptide, protein, hormone, co-factor, or nucleic acid), or (d) have a modulatory effect on the expression of the marker. Such assays typically comprise a reaction between the marker and one or more assay components. The other components may be either the test compound itself, or a combination of test compound and a natural binding partner of the marker. Compounds identified via assays such as those described herein may be useful, for example, for modulating, e.g., inhibiting, ameliorating, treating, or preventing cancer.


The test compounds of the present invention may be obtained from any available source, including systematic libraries of natural and/or synthetic compounds. Test compounds may also be obtained by any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; peptoid libraries (libraries of molecules having the functionalities of peptides, but with a novel, non-peptide backbone which are resistant to enzymatic degradation but which nevertheless remain bioactive; see, e.g., Zuckermann et al., 1994, J. Med. Chem. 37:2678-85); spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the ‘one-bead one-compound’ library method; and synthetic library methods using affinity chromatography selection. The biological library and peptoid library approaches are limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, 1997, Anticancer Drug Des. 12:145).


Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt et al. (1993) Proc. Natl. Acad. Sci. U.S.A. 90:6909; Erb et al. (1994) Proc. Natl. Acad. Sci. USA 91:11422; Zuckermann et al. (1994). J. Med. Chem. 37:2678; Cho et al. (1993) Science 261:1303; Carrell et al. (1994) Angew. Chem. Int. Ed. Engl. 33:2059; Carell et al. (1994) Angew. Chem. Int. Ed. Engl. 33:2061; and in Gallop et al. (1994)J. Med. Chem. 37:1233.


Libraries of compounds may be presented in solution (e.g., Houghten, 1992, Biotechniques 13:412-421), or on beads (Lam, 1991, Nature 354:82-84), chips (Fodor, 1993, Nature 364:555-556), bacteria and/or spores, (Ladner, U.S. Pat. No. 5,223,409), plasmids (Cull et al, 1992, Proc Natl Acad Sci USA 89:1865-1869) or on phage (Scott and Smith, 1990, Science 249:386-390; Devlin, 1990, Science 249:404-406; Cwirla-et al, 1990, Proc. Natl. Acad. Sci. 87:6378-6382; Felici, 1991, J. Mol. Biol. 222:301-310; Ladner, supra.).


In one embodiment, the invention provides assays for screening candidate or test compounds which are substrates of a marker or biologically active portion thereof. In another embodiment, the invention provides assays for screening candidate or test compounds which bind to a marker or biologically active portion thereof. Determining the ability of the test compound to directly bind to a marker can be accomplished, for example, by coupling the compound with a radioisotope or enzymatic label such that binding of the compound to the marker can be determined by detecting the labeled marker compound in a complex. For example, compounds (e.g., marker substrates) can be labeled with 125I, 35S, 14C, or 3H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting. Alternatively, assay components can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product.


In another embodiment, the invention provides assays for screening candidate or test compounds which modulate the activity of a marker or a biologically active portion thereof. In all likelihood, the marker can, in vivo, interact with one or more molecules, such as, but not limited to, peptides, proteins, hormones, cofactors and nucleic acids. For the purposes of this discussion, such cellular and extracellular molecules are referred to herein as “binding partners” or marker “substrate”.


One necessary embodiment of the invention in order to facilitate such screening is the use of the marker to identify its natural in vivo binding partners. There are many ways to accomplish this which are known to one skilled in the art. One example is the use of the marker protein as “bait protein” in a two-hybrid assay or three-hybrid assay (see, e.g., U.S. Pat. No. 5,283,317; Zervos et al, 1993, Cell 72:223-232; Madura et al, 1993, J. Biol. Chem. 268:12046-12054; Bartel et al, 1993, Biotechniques 14:920-924; Iwabuchi et al, 1993 Oncogene 8:1693-1696; Brent WO94/10300) in order to identify other proteins which bind to or interact with the marker (binding partners) and, therefore, are possibly involved in the natural function of the marker. Such marker binding partners are also likely to be involved in the propagation of signals by the marker or downstream elements of a marker-mediated signaling pathway. Alternatively, such marker binding partners may also be found to be inhibitors of the marker.


The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. In one construct, the gene that encodes a marker protein fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other construct, a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein (“prey” or “sample”) is fused to a gene that codes for the activation domain of the known transcription factor. If the “bait” and the “prey” proteins are able to interact, in vivo, forming a marker-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be readily detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene which encodes the protein which interacts with the marker protein.


In a further embodiment, assays may be devised through the use of the invention for the purpose of identifying compounds which modulate (e.g., affect either positively or negatively) interactions between a marker and its substrates and/or binding partners. Such compounds can include, but are not limited to, molecules such as antibodies, peptides, hormones, oligonucleotides, nucleic acids, and analogs thereof. Such compounds may also be obtained from any available source, including systematic libraries of natural and/or synthetic compounds. The preferred assay components for use in this embodiment is a cancer, marker identified herein, the known binding partner and/or substrate of same, and the test compound. Test compounds can be supplied from any source.


The basic principle of the assay systems used to identify compounds that interfere with the interaction between the marker and its binding partner involves preparing a reaction mixture containing the marker and its binding partner under conditions and for a time sufficient to allow the two products to interact and bind, thus forming a complex. In order to test an agent for inhibitory activity, the reaction mixture is prepared in the presence and absence of the test compound. The test compound can be initially included in the reaction mixture, or can be added at a time subsequent to the addition of the marker and its binding partner. Control reaction mixtures are incubated without the test compound or with a placebo. The formation of any complexes between the marker and its binding partner is then detected. The formation of a complex in the control reaction, but less or no such formation in the reaction mixture containing the test compound, indicates that the compound interferes with the interaction of the marker and its binding partner. Conversely, the formation of more complex in the presence of compound than in the control reaction indicates that the compound may enhance interaction of the marker and its binding partner. The assay for compounds that interfere with the interaction of the marker with its binding partner may be conducted in a heterogeneous or homogeneous format. Heterogeneous assays involve anchoring either the marker or its binding partner onto a solid phase and detecting complexes anchored to the solid phase at the end of the reaction. In homogeneous assays, the entire reaction is carried out in a liquid phase. In either approach, the order of addition of reactants can be varied to obtain different information about the compounds being tested. For example, test compounds that interfere with the interaction between the markers and the binding partners (e.g., by competition) can be identified by conducting the reaction in the presence of the test substance, i.e., by adding the test substance to the reaction mixture prior to or simultaneously with the marker and its interactive binding partner. Alternatively, test compounds that disrupt preformed complexes, e.g. compounds with higher binding constants that displace one of the components from the complex, can be tested by adding the test compound to the reaction mixture after complexes have been formed. The various formats are briefly described below.


In a heterogeneous assay system, either the marker or its binding partner is anchored onto a solid surface or matrix, while the other corresponding non-anchored component may be labeled, either directly or indirectly. In practice, microtitre plates are often utilized for this approach. The anchored species can be immobilized by a number of methods, either non-covalent or covalent, that are typically well known to one who practices the art. Non-covalent attachment can often be accomplished simply by coating the solid surface with a solution of the marker or its binding partner and drying. Alternatively, an immobilized antibody specific for the assay component to be anchored can be used for this purpose. Such surfaces can often be prepared in advance and stored.


In related embodiments, a fusion protein can be provided which adds a domain that allows one or both of the assay components to be anchored to a matrix. For example, glutathione-S-transferase/marker fusion proteins or glutathione-S-transferase/binding partner can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, Mo.) or glutathione derivatized microtiter plates, which are then combined with the test compound or the test compound and either the non-adsorbed marker or its binding partner, and the mixture incubated under conditions conducive to complex formation (e.g., physiological conditions). Following incubation, the beads or microtiter plate wells are washed to remove any unbound assay components, the immobilized complex assessed either directly or indirectly, for example, as described above. Alternatively, the complexes can be dissociated from the matrix, and the level of marker binding or activity determined using standard techniques.


Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either a marker or a marker binding partner can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated marker protein or target molecules can be prepared from biotin-NHS(N-hydroxy-succinimide) using techniques known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). In certain embodiments, the protein-immobilized surfaces can be prepared in advance and stored.


In order to conduct the assay, the corresponding partner of the immobilized assay component is exposed to the coated surface with or without the test compound. After the reaction is complete, unreacted assay components are removed (e.g., by washing) and any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the non-immobilized component is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the non-immobilized component is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the initially non-immobilized species (the antibody, in turn, can be directly labeled or indirectly labeled with, e.g., a labeled anti-Ig antibody). Depending upon the order of addition of reaction components, test compounds which modulate (inhibit or enhance) complex formation or which disrupt preformed complexes can be detected.


In an alternate embodiment of the invention, a homogeneous assay may be used. This is typically a reaction, analogous to those mentioned above, which is conducted in a liquid phase in the presence or absence of the test compound. The formed complexes are then separated from unreacted components, and the amount of complex formed is determined. As mentioned for heterogeneous assay systems, the order of addition of reactants to the liquid phase can yield information about which test compounds modulate (inhibit or enhance) complex formation and which disrupt preformed complexes.


In such a homogeneous assay, the reaction products may be separated from unreacted assay components by any of a number of standard techniques, including but not limited to: differential centrifugation, chromatography, electrophoresis and immunoprecipitation. In differential centrifugation, complexes of molecules may be separated from uncomplexed molecules through a series of centrifugal steps, due to the different sedimentation equilibria of complexes based on their different sizes and densities (see, for example, Rivas, G., and Minton, A. P., Trends Biochem Sci 1993 August; 18(8):284-7). Standard chromatographic techniques may also be utilized to separate complexed molecules from uncomplexed ones. For example, gel filtration chromatography separates molecules based on size, and through the utilization of an appropriate gel filtration resin in a column format, for example, the relatively larger complex may be separated from the relatively smaller uncomplexed components. Similarly, the relatively different charge properties of the complex as compared to the uncomplexed molecules may be exploited to differentially separate the complex from the remaining individual reactants, for example through the use of ion-exchange chromatography resins. Such resins and chromatographic techniques are well known to one skilled in the art (see, e.g., Heegaard, 1998, J. Mol. Recognit. 11:141-148; Hage and Tweed, 1997, J. Chromatogr. B. Biomed. Sci. Appl., 699:499-525). Gel electrophoresis may also be employed to separate complexed molecules from unbound species (see, e.g., Ausubel et al (eds.), In: Current Protocols in Molecular Biology, J. Wiley & Sons, New York. 1999). In this technique, protein or nucleic acid complexes are separated based on size or charge, for example. In order to maintain the binding interaction during the electrophoretic process, nondenaturing gels in the absence of reducing agent are typically preferred, but conditions appropriate to the particular interactants will be well known to one skilled in the art. Immunoprecipitation is another common technique utilized for the isolation of a protein-protein complex from solution (see, e.g., Ausubel et al (eds.), In: Current Protocols in Molecular Biology, J. Wiley & Sons, New York. 1999). In this technique, all proteins binding to an antibody specific to one of the binding molecules are precipitated from solution by conjugating the antibody to a polymer bead that may be readily collected by centrifugation. The bound assay components are released from the beads (through a specific proteolysis event or other technique well known in the art which will not disturb the protein-protein interaction in the complex), and a second immunoprecipitation step is performed, this time utilizing antibodies specific for the correspondingly different interacting assay component. In this manner, only formed complexes should remain attached to the beads. Variations in complex formation in both the presence and the absence of a test compound can be compared, thus offering information about the ability of the compound to modulate interactions between the marker and its binding partner.


Also within the scope of the present invention are methods for direct detection of interactions between the marker and its natural binding partner and/or a test compound in a homogeneous or heterogeneous assay system without further sample manipulation. For example, the technique of fluorescence energy transfer may be utilized (see, e.g., Lakowicz et al, U.S. Pat. No. 5,631,169; Stavrianopoulos et al, U.S. Pat. No. 4,868,103). Generally, this technique involves the addition of a fluorophore label on a first ‘donor’ molecule (e.g., marker or test compound) such that its emitted fluorescent energy will be absorbed by a fluorescent label on a second, ‘acceptor’ molecule (e.g., marker or test compound), which in turn is able to fluoresce due to the absorbed energy. Alternately, the ‘donor’ protein molecule may simply utilize the natural fluorescent energy of tryptophan residues. Labels are chosen that emit different wavelengths of light, such that the ‘acceptor’ molecule label may be differentiated from that of the ‘donor’. Since the efficiency of energy transfer between the labels is related to the distance separating the molecules, spatial relationships between the molecules can be assessed. In a situation in which binding occurs between the molecules, the fluorescent emission of the ‘acceptor’ molecule label in the assay should be maximal. An FET binding event can be conveniently measured through standard fluorometric detection means well known in the art (e.g., using a fluorimeter). A test substance which either enhances or hinders participation of one of the species in the preformed complex will result in the generation of a signal variant to that of background. In this way, test substances that modulate interactions between a marker and its binding partner can be identified in controlled assays.


In another embodiment, modulators of marker expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of mRNA or protein, corresponding to a marker in the cell, is determined. The level of expression of mRNA or protein in the presence of the candidate compound is compared to the level of expression of mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of marker expression based on this comparison. For example, when expression of marker mRNA or protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of marker mRNA or protein expression. Conversely, when expression of marker mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of marker mRNA or protein expression. The level of marker mRNA or protein expression in the cells can be determined by methods described herein for detecting marker mRNA or protein.


In another aspect, the invention pertains to a combination of two or more of the assays described herein. For example, a modulating agent can be identified using a cell-based or a cell free assay, and the ability of the agent to modulate the activity of a marker protein can be further confirmed in vivo, e.g., in a whole animal model for cancer, cellular transformation and/or tumorigenesis. An animal model for pancreatic cancer is described in, for example, Aguirre A., et al. (2003) Genes Dev. December 15; 17(24):3112-26, the contents of which are expressly incorporated herein by reference. Additional animal based models of cancer are well known in the art (reviewed in Animal Models of Cancer Predisposition Syndromes, Hiai, H and Hino, O (eds.) 1999, Progress in Experimental Tumor Research, Vol. 35; Clarke A R Carcinogenesis (2000) 21:435-41) and include, for example, carcinogen-induced tumors (Rithidech, K et al. Mutat Res (1999) 428:33-39; Miller, M L et al. Environ Mol Mutagen (2000) 35:319-327), injection and/or transplantation of tumor cells into an animal, as well as animals bearing mutations in growth regulatory genes, for example, oncogenes (e.g., ras) (Arbeit, J M et al. Am J Pathol (1993) 142:1187-1197; Sinn, E et al. Cell (1987) 49:465-475; Thorgeirsson, S S et al. Toxicol Lett (2000) 112-113:553-555) and tumor suppressor genes (e.g., p53) (Vooijs, M et al. Oncogene (1999) 18:5293-5303; Clark A R Cancer Metast Rev (1995) 14:125-148; Kumar, T R et al. J Intern Med (1995) 238:233-238; Donehower, L A et al. (1992) Nature 356215-221). Furthermore, experimental model systems are available for the study of, for example, ovarian cancer (Hamilton, T C et al. Semin Oncol (1984) 11:285-298; Rahman, N A et al. Mol Cell Endocrinol (1998) 145:167-174; Beamer, W G et al. Toxicol Pathol (1998) 26:704-710), gastric cancer (Thompson, J et al. Int J Cancer (2000) 86:863-869; Fodde, R et al. Cytogenet Cell Genet (1999) 86:105-111), breast cancer (Li, M et al. Oncogene (2000) 19:1010-1019; Green, J E et al. Oncogene (2000) 19:1020-1027), melanoma (Satyamoorthy, K et al. Cancer Metast Rev (1999) 18:401-405), and prostate cancer (Shirai, T et al. Mutat Res (2000) 462:219-226; Bostwick, D G et al. Prostate (2000) 43:286-294). Animal models described in, for example, Chin L. et al (1999) Nature 400(6743):468-72, may also be used in the methods of the invention.


This invention further pertains to novel agents identified by the above-described screening assays. Accordingly, it is within the scope of this invention to further use an agent identified as described herein in an appropriate animal model. For example, an agent identified as described herein (e.g., a marker modulating agent, a small molecule, an antisense marker nucleic acid molecule, a ribozyme, a marker-specific antibody, or fragment thereof, a marker protein, a marker nucleic acid molecule, an RNA interfering agent, e.g., an siRNA molecule targeting a marker of the invention, or a marker-binding partner) can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent. Alternatively, an agent identified as described herein can be used in an animal model to determine the mechanism of action of such an agent. Furthermore, this invention pertains to uses of novel agents identified by the above-described screening assays for treatments as described herein.


VIII. Pharmaceutical Compositions

The small molecules, peptides, peptoids, peptidomimetics, polypeptides, RNA interfering agents, e.g., siRNA molecules, antibodies, ribozymes, and antisense oligonucleotides (also referred to herein as “active compounds” or “compounds”) corresponding to a marker of the invention can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the small molecules, peptides, peptoids, peptidomimetics, polypeptides, RNA interfering agents, e.g., siRNA molecules, antibodies, ribozymes, or antisense oligonucleotides and a pharmaceutically acceptable carrier. As used herein the language “pharmaceutically acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.


The invention includes methods for preparing pharmaceutical compositions for modulating the expression or activity of a polypeptide or nucleic acid corresponding to a marker of the invention. Such methods comprise formulating a pharmaceutically acceptable carrier with an agent which modulates expression or activity of a polypeptide or nucleic acid corresponding to a marker of the invention. Such compositions can further include additional active agents. Thus, the invention further includes methods for preparing a pharmaceutical composition by formulating a pharmaceutically acceptable carrier with an agent which modulates expression or activity of a polypeptide or nucleic acid corresponding to a marker of the invention and one or more additional active compounds.


It is understood that appropriate doses of small molecule agents and protein or polypeptide agents depends upon a number of factors within the knowledge of the ordinarily skilled physician, veterinarian, or researcher. The dose(s) of these agents will vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable, and the effect which the practitioner desires the agent to have upon the nucleic acid molecule or polypeptide of the invention. Small molecules include, but are not limited to, peptides, peptidomimetics, amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, nucleotides, nucleotide analogs, organic or inorganic compounds (i.e., including heteroorganic and organometallic compounds) having a molecular weight less than about 10,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 1,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 500 grams per mole, and salts, esters, and other pharmaceutically acceptable forms of such compounds.


Exemplary doses of a small molecule include milligram or microgram amounts per kilogram of subject or sample weight (e.g. about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram).


As defined herein, a therapeutically effective amount of an RNA interfering agent, e.g., siRNA, (i.e., an effective dosage) ranges from about 0.001 to 3,000 mg/kg body weight, preferably about 0.01 to 2500 mg/kg body weight, more preferably about 0.1 to 2000, about 0.1 to 1000 mg/kg body weight, 0.1 to 500 mg/kg body weight, 0.1 to 100 mg/kg body weight, 0.1 to 50 mg/kg body weight, 0.1 to 25 mg/kg body weight, and even more preferably about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight. Treatment of a subject with a therapeutically effective amount of an RNA interfering agent can include a single treatment or, preferably, can include a series of treatments. In a preferred example, a subject is treated with an RNA interfering agent in the range of between about 0.1 to 20 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks.


Exemplary doses of a protein or polypeptide include gram, milligram or microgram amounts per kilogram of subject or sample weight (e.g. about 1 microgram per kilogram to about 5 grams per kilogram, about 100 micrograms per kilogram to about 500 milligrams per kilogram, or about 1 milligram per kilogram to about 50 milligrams per kilogram). It is furthermore understood that appropriate doses of one of these agents depend upon the potency of the agent with respect to the expression or activity to be modulated. Such appropriate doses can be determined using the assays described herein. When one or more of these agents is to be administered to an animal (e.g. a human) in order to modulate expression or activity of a polypeptide or nucleic acid of the invention, a physician, veterinarian, or researcher can, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained. In addition, it is understood that the specific dose level for any particular animal subject will depend upon a variety of factors including the activity of the specific agent employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.


A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediamine-tetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampules, disposable syringes or multiple dose vials made of glass or plastic.


Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF; Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions can be prepared by incorporating the active compound (e.g., a polypeptide or antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium, and then incorporating the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.


Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.


Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches, and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.


For administration by inhalation, the compounds are delivered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant, e.g. a gas such as carbon dioxide, or a nebulizer.


Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.


The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.


In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes having monoclonal antibodies incorporated therein or thereon) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.


It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.


For antibodies, the preferred dosage is 0.1 mg/kg to 100 mg/kg of body weight (generally 10 mg/kg to 20 mg/kg). If the antibody is to act in the brain, a dosage of 50 mg/kg to 100 mg/kg is usually appropriate. Generally, partially human antibodies and fully human antibodies have a longer half-life within the human body than other antibodies. Accordingly, lower dosages and less frequent administration is often possible. Modifications such as lipidation can be used to stabilize antibodies and to enhance uptake and tissue penetration (e.g., into the epithelium). A method for lipidation of antibodies is described by Cruikshank et al. (1997) J Acquired Immune Deficiency Syndromes and Human Retrovirology 14:193.


The nucleic acid molecules corresponding to a marker of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (U.S. Pat. No. 5,328,470), or by stereotactic injection (see, e.g., Chen et al., 1994, Proc. Natl. Acad. Sci. USA 91:3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g. retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system.


The RNA interfering agents, e.g., siRNAs used in the methods of the invention can be inserted into vectors. These constructs can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Pat. No. 5,328,470) or by stereotactic injection (see e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA 91:3054-3057). The pharmaceutical preparation of the vector can include the RNA interfering agent, e.g., the siRNA vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system.


The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.


IX. Predictive Medicine

The present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trails are used for prognostic (predictive) purposes to thereby treat an individual prophylactically. Accordingly, one aspect of the present invention relates to diagnostic assays for determining the amount, structure, and/or activity of polypeptides or nucleic acids corresponding to one or more markers of the invention, in order to determine whether an individual is at risk of developing cancer. Such assays can be used for prognostic or predictive purposes to thereby prophylactically treat an individual prior to the onset of the cancer.


Yet another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs or other compounds administered either to inhibit cancer, or to treat or prevent any other disorder {i.e. in order to understand any carcinogenic effects that such treatment may have}) on the amount, structure, and/or activity of a marker of the invention in clinical trials. These and other agents are described in further detail in the following sections.


A. Diagnostic Assays


1. Methods for Detection of Copy Number


Methods of evaluating the copy number of a particular marker or chromosomal region (e.g., an MCR) are well known to those of skill in the art. The presence or absence of chromosomal gain or loss can be evaluated simply by a determination of copy number of the regions or markers identified herein.


Methods for evaluating copy number of encoding nucleic acid in a sample include, but are not limited to, hybridization-based assays. For example, one method for evaluating the copy number of encoding nucleic acid in a sample involves a Southern Blot. In a Southern Blot, the genomic DNA (typically fragmented and separated on an electrophoretic gel) is hybridized to a probe specific for the target region. Comparison of the intensity of the hybridization signal from the probe for the target region with control probe signal from analysis of normal genomic DNA (e.g., a non-amplified portion of the same or related cell, tissue, organ, etc.) provides an estimate of the relative copy number of the target nucleic acid.


An alternative means for determining the copy number is in situ hybridization (e.g., Angerer (1987) Meth. Enzymol 152: 649). Generally, in situ hybridization comprises the following steps: (1) fixation of tissue or biological structure to be analyzed; (2) prehybridization treatment of the biological structure to increase accessibility of target DNA, and to reduce nonspecific binding; (3) hybridization of the mixture of nucleic acids to the nucleic acid in the biological structure or tissue; (4) post-hybridization washes to remove nucleic acid fragments not bound in the hybridization and (5) detection of the hybridized nucleic acid fragments. The reagent used in each of these steps and the conditions for use vary depending on the particular application.


Preferred hybridization-based assays include, but are not limited to, traditional “direct probe” methods such as Southern blots or in situ hybridization (e.g., FISH), and “comparative probe” methods such as comparative genomic hybridization (CGH), e.g., cDNA-based or oligonucleotide-based CGH. The methods can be used in a wide variety of formats including, but not limited to, substrate (e.g. membrane or glass) bound methods or array-based approaches.


In a typical in situ hybridization assay, cells are fixed to a solid support, typically a glass slide. If a nucleic acid is to be probed, the cells are typically denatured with heat or alkali. The cells are then contacted with a hybridization solution at a moderate temperature to permit annealing of labeled probes specific to the nucleic acid sequence encoding the protein. The targets (e.g., cells) are then typically washed at a predetermined stringency or at an increasing stringency until an appropriate signal to noise ratio is obtained.


The probes are typically labeled, e.g., with radioisotopes or fluorescent reporters. Preferred probes are sufficiently long so as to specifically hybridize with the target nucleic acid(s) under stringent conditions. The preferred size range is from about 200 bases to about 1000 bases.


In some applications it is necessary to block the hybridization capacity of repetitive sequences. Thus, in some embodiments, tRNA, human genomic DNA, or Cot-I DNA is used to block non-specific hybridization.


In CGH methods, a first collection of nucleic acids (e.g. from a sample, e.g., a possible tumor) is labeled with a first label, while a second collection of nucleic acids (e.g. a control, e.g., from a healthy cell/tissue) is labeled with a second label. The ratio of hybridization of the nucleic acids is determined by the ratio of the two (first and second) labels binding to each fiber in the array. Where there are chromosomal deletions or multiplications, differences in the ratio of the signals from the two labels will be detected and the ratio will provide a measure of the copy number. Array-based CGH may also be performed with single-color labeling (as opposed to labeling the control and the possible tumor sample with two different dyes and mixing them prior to hybridization, which will yield ratio due to competitive hybridization to probes on the arrays). In single color CGH, the control is labeled and hybridized to one array and absolute signals are read, and the possible tumor sample is labeled and hybridized to a second array (with identical content) and absolute signals are read. Copy number difference is calculated based on absolute signals from the two arrays. Hybridization protocols suitable for use with the methods of the invention are described, e.g., in Albertson (1984) EMBO J. 3: 1227-1234; Pinkel (1988) Proc. Natl. Acad. Sci. USA 85: 9138-9142; EPO Pub. No. 430, 402; Methods in Molecular Biology, Vol. 33: In Situ Hybridization Protocols, Choo, ed., Humana Press, Totowa, N.J. (1994), etc. In one embodiment, the hybridization protocol of Pinkel et al. (1998) Nature Genetics 20: 207-211, or of Kallioniemi (1992) Proc. Natl Acad Sci USA 89:5321-5325 (1992) is used.


The methods of the invention are particularly well suited to array-based hybridization formats. Array-based CGH is described in U.S. Pat. No. 6,455,258, the contents of which are incorporated herein by reference.


In still another embodiment, amplification-based assays can be used to measure copy number. In such amplification-based assays, the nucleic acid sequences act as a template in an amplification reaction (e.g., Polymerase Chain Reaction (PCR). In a quantitative amplification, the amount of amplification product will be proportional to the amount of template in the original sample. Comparison to appropriate controls, e.g. healthy tissue, provides a measure of the copy number.


Methods of “quantitative” amplification are well known to those of skill in the art. For example, quantitative PCR involves simultaneously co-amplifying a known quantity of a control sequence using the same primers. This provides an internal standard that may be used to calibrate the PCR reaction. Detailed protocols for quantitative PCR are provided in Innis et al. (1990) PCR Protocols, A Guide to Methods and Applications, Academic Press, Inc. N.Y.). Measurement of DNA copy number at microsatellite loci using quantitative PCR analysis is described in Ginzonger, et al (2000) Cancer Research 60:5405-5409. The known nucleic acid sequence for the genes is sufficient to enable one of skill in the art to routinely select primers to amplify any portion of the gene. Fluorogenic quantitative PCR may also be used in the methods of the invention. In fluorogenic quantitative PCR, quantitation is based on amount of fluorescence signals, e.g., TaqMan and sybr green.


Other suitable amplification methods include, but are not limited to, ligase chain reaction (LCR) (see Wu and Wallace (1989) Genomics 4: 560, Landegren et al. (1988) Science 241: 1077, and Barringer et al. (1990) Gene 89: 117, transcription amplification (Kwoh et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173), self-sustained sequence replication (Guatelli et al. (1990) Proc. Nat. Acad. Sci. USA 87: 1874), dot PCR, and linker adapter PCR, etc.


Loss of heterozygosity (LOH) mapping (Wang Z. C. et al. (2004) Cancer Res 64(1):64-71; Seymour, A. B., et al. (1994) Cancer Res 54, 2761-4; Hahn, S. A., et al. (1995) Cancer Res 55, 4670-5; Kimura, M., et al. (1996) Genes Chromosomes Cancer 17, 88-93) may also be used to identify regions of amplification or deletion.


2. Methods for Detection of Gene Expression


Marker expression level can also be assayed as a method for diagnosis of cancer or risk for developing cancer. Expression of a marker of the invention may be assessed by any of a wide variety of well known methods for detecting expression of a transcribed molecule or protein. Non-limiting examples of such methods include immunological methods for detection of secreted, cell-surface, cytoplasmic, or nuclear proteins, protein purification methods, protein function or activity assays, nucleic acid hybridization methods, nucleic acid reverse transcription methods, and nucleic acid amplification methods.


In preferred embodiments, activity of a particular gene is characterized by a measure of gene transcript (e.g. mRNA), by a measure of the quantity of translated protein, or by a measure of gene product activity. Marker expression can be monitored in a variety of ways, including by detecting mRNA levels, protein levels, or protein activity, any of which can be measured using standard techniques. Detection can involve quantification of the level of gene expression (e.g., genomic DNA, cDNA, mRNA, protein, or enzyme activity), or, alternatively, can be a qualitative assessment of the level of gene expression, in particular in comparison with a control level. The type of level being detected will be clear from the context.


Methods of detecting and/or quantifying the gene transcript (mRNA or cDNA made therefrom) using nucleic acid hybridization techniques are known to those of skill in the art (see Sambrook et al. supra). For example, one method for evaluating the presence, absence, or quantity of cDNA involves a Southern transfer as described above. Briefly, the mRNA is isolated (e.g. using an acid guanidinium-phenol-chloroform extraction method, Sambrook et al. supra.) and reverse transcribed to produce cDNA. The cDNA is then optionally digested and run on a gel in buffer and transferred to membranes. Hybridization is then carried out using the nucleic acid probes specific for the target cDNA.


A general principle of such diagnostic and prognostic assays involves preparing a sample or reaction mixture that may contain a marker, and a probe, under appropriate conditions and for a time sufficient to allow the marker and probe to interact and bind, thus forming a complex that can be removed and/or detected in the reaction mixture. These assays can be conducted in a variety of ways.


For example, one method to conduct such an assay would involve anchoring the marker or probe onto a solid phase support, also referred to as a substrate, and detecting target marker/probe complexes anchored on the solid phase at the end of the reaction. In one embodiment of such a method, a sample from a subject, which is to be assayed for presence and/or concentration of marker, can be anchored onto a carrier or solid phase support. In another embodiment, the reverse situation is possible, in which the probe can be anchored to a solid phase and a sample from a subject can be allowed to react as an unanchored component of the assay.


There are many established methods for anchoring assay components to a solid phase. These include, without limitation, marker or probe molecules which are immobilized through conjugation of biotin and streptavidin. Such biotinylated assay components can be prepared from biotin-NHS(N-hydroxy-succinimide) using techniques known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). In certain embodiments, the surfaces with immobilized assay components can be prepared in advance and stored.


Other suitable carriers or solid phase supports for such assays include any material capable of binding the class of molecule to which the marker or probe belongs. Well-known supports or carriers include, but are not limited to, glass, polystyrene, nylon, polypropylene, nylon, polyethylene, dextran, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.


In order to conduct assays with the above mentioned approaches, the non-immobilized component is added to the solid phase upon which the second component is anchored. After the reaction is complete, uncomplexed components may be removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized upon the solid phase. The detection of marker/probe complexes anchored to the solid phase can be accomplished in a number of methods outlined herein.


In a preferred embodiment, the probe, when it is the unanchored assay component, can be labeled for the purpose of detection and readout of the assay, either directly or indirectly, with detectable labels discussed herein and which are well-known to one skilled in the art.


It is also possible to directly detect marker/probe complex formation without further manipulation or labeling of either component (marker or probe), for example by utilizing the technique of fluorescence energy transfer (see, for example, Lakowicz et al., U.S. Pat. No. 5,631,169; Stavrianopoulos, et al., U.S. Pat. No. 4,868,103). A fluorophore label on the first, ‘donor’ molecule is selected such that, upon excitation with incident light of appropriate wavelength, its emitted fluorescent energy will be absorbed by a fluorescent label on a second ‘acceptor’ molecule, which in turn is able to fluoresce due to the absorbed energy. Alternately, the ‘donor’ protein molecule may simply utilize the natural fluorescent energy of tryptophan residues. Labels are chosen that emit different wavelengths of light, such that the ‘acceptor’ molecule label may be differentiated from that of the ‘donor’. Since the efficiency of energy transfer between the labels is related to the distance separating the molecules, spatial relationships between the molecules can be assessed. In a situation in which binding occurs between the molecules, the fluorescent emission of the ‘acceptor’ molecule label in the assay should be maximal. An FET binding event can be conveniently measured through standard fluorometric detection means well known in the art (e.g., using a fluorimeter).


In another embodiment, determination of the ability of a probe to recognize a marker can be accomplished without labeling either assay component (probe or marker) by utilizing a technology such as real-time Biomolecular Interaction Analysis (BIA) (see, e.g., Sjolander, S, and Urbaniczky, C., 1991, Anal. Chem. 63:2338-2345 and Szabo et al., 1995, Curr. Opin. Struct. Biol. 5:699-705). As used herein, “BIA” or “surface plasmon resonance” is a technology for studying biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcore). Changes in the mass at the binding surface (indicative of a binding event) result in alterations of the refractive index of light near the surface (the optical phenomenon of surface plasmon resonance (SPR)), resulting in a detectable signal which can be used as an indication of real-time reactions between biological molecules.


Alternatively, in another embodiment, analogous diagnostic and prognostic assays can be conducted with marker and probe as solutes in a liquid phase. In such an assay, the complexed marker and probe are separated from uncomplexed components by any of a number of standard techniques, including but not limited to: differential centrifugation, chromatography, electrophoresis and immunoprecipitation. In differential centrifugation, marker/probe complexes may be separated from uncomplexed assay components through a series of centrifugal steps, due to the different sedimentation equilibria of complexes based on their different sizes and densities (see, for example, Rivas, G., and Minton, A. P., 1993, Trends Biochem Sci. 18(8):284-7). Standard chromatographic techniques may also be utilized to separate complexed molecules from uncomplexed ones. For example, gel filtration chromatography separates molecules based on size, and through the utilization of an appropriate gel filtration resin in a column format, for example, the relatively larger complex may be separated from the relatively smaller uncomplexed components. Similarly, the relatively different charge properties of the marker/probe complex as compared to the uncomplexed components may be exploited to differentiate the complex from uncomplexed components, for example through the utilization of ion-exchange chromatography resins. Such resins and chromatographic techniques are well known to one skilled in the art (see, e.g., Heegaard, N. H., 1998, J. Mol. Recognit. Winter 11 (1-6):141-8; Hage, D. S., and Tweed, S. A. J Chromatogr B Biomed Sci Appl 1997 Oct. 10; 699 (1-2):499-525). Gel electrophoresis may also be employed to separate complexed assay components from unbound components (see, e.g., Ausubel et al., ed., Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1987-1999). In this technique, protein or nucleic acid complexes are separated based on size or charge, for example. In order to maintain the binding interaction during the electrophoretic process, non-denaturing gel matrix materials and conditions in the absence of reducing agent are typically preferred. Appropriate conditions to the particular assay and components thereof will be well known to one skilled in the art.


In a particular embodiment, the level of mRNA corresponding to the marker can be determined both by in situ and by in vitro formats in a biological sample using methods known in the art. The term “biological sample” is intended to include tissues, cells, biological fluids and isolates thereof, isolated from a subject, as well as tissues, cells and fluids present within a subject. Many expression detection methods use isolated RNA. For in vitro methods, any RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of RNA from cells (see, e.g., Ausubel et al., ed., Current Protocols in Molecular Biology, John Wiley & Sons, New York 1987-1999). Additionally, large numbers of tissue samples can readily be processed using techniques well known to those of skill in the art, such as, for example, the single-step RNA isolation process of Chomczynski (1989, U.S. Pat. No. 4,843,155).


The isolated nucleic acid can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses and probe arrays. One preferred diagnostic method for the detection of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to the mRNA encoded by the gene being detected. The nucleic acid probe can be, for example, a full-length cDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to a mRNA or genomic DNA encoding a marker of the present invention. Other suitable probes for use in the diagnostic assays of the invention are described herein. Hybridization of an mRNA with the probe indicates that the marker in question is being expressed.


In one format, the mRNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose. In an alternative format, the probe(s) are immobilized on a solid surface and the mRNA is contacted with the probe(s), for example, in an Affymetrix gene chip array. A skilled artisan can readily adapt known mRNA detection methods for use in detecting the level of mRNA encoded by the markers of the present invention.


The probes can be full length or less than the full length of the nucleic acid sequence encoding the protein. Shorter probes are empirically tested for specificity. Preferably nucleic acid probes are 20 bases or longer in length. (See, e.g., Sambrook et al. for methods of selecting nucleic acid probe sequences for use in nucleic acid hybridization.) Visualization of the hybridized portions allows the qualitative determination of the presence or absence of cDNA.


An alternative method for determining the level of a transcript corresponding to a marker of the present invention in a sample involves the process of nucleic acid amplification, e.g., by rtPCR (the experimental embodiment set forth in Mullis, 1987, U.S. Pat. No. 4,683,202), ligase chain reaction (Barany, 1991, Proc. Natl. Acad. Sci. USA, 88:189-193), self sustained sequence replication (Guatelli et al., 1990, Proc. Natl. Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh et al., 1989, Proc. Natl. Acad. Sci. USA 86:1173-1177), Q-Beta Replicase (Lizardi et al., 1988, Bio/Technology 6:1197), rolling circle replication (Lizardi et al., U.S. Pat. No. 5,854,033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. Fluorogenic rtPCR may also be used in the methods of the invention. In fluorogenic rtPCR, quantitation is based on amount of fluorescence signals, e.g., TaqMan and sybr green. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. As used herein, amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5′ or 3′ regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between. In general, amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers. For in situ methods, mRNA does not need to be isolated from the cells prior to detection. In such methods, a cell or tissue sample is prepared/processed using known histological methods. The sample is then immobilized on a support, typically a glass slide, and then contacted with a probe that can hybridize to mRNA that encodes the marker.


As an alternative to making determinations based on the absolute expression level of the marker, determinations may be based on the normalized expression level of the marker. Expression levels are normalized by correcting the absolute expression level of a marker by comparing its expression to the expression of a gene that is not a marker, e.g., a housekeeping gene that is constitutively expressed. Suitable genes for normalization include housekeeping genes such as the actin gene, or epithelial cell-specific genes. This normalization allows the comparison of the expression level in one sample, e.g., a subject sample, to another sample, e.g., a non-cancerous sample, or between samples from different sources.


Alternatively, the expression level can be provided as a relative expression level. To determine a relative expression level of a marker, the level of expression of the marker is determined for 10 or more samples of normal versus cancer cell isolates, preferably 50 or more samples, prior to the determination of the expression level for the sample in question. The mean expression level of each of the genes assayed in the larger number of samples is determined and this is used as a baseline expression level for the marker. The expression level of the marker determined for the test sample (absolute level of expression) is then divided by the mean expression value obtained for that marker. This provides a relative expression level.


Preferably, the samples used in the baseline determination will be from cancer cells or normal cells of the same tissue type. The choice of the cell source is dependent on the use of the relative expression level. Using expression found in normal tissues as a mean expression score aids in validating whether the marker assayed is specific to the tissue from which the cell was derived (versus normal cells). In addition, as more data is accumulated, the mean expression value can be revised, providing improved relative expression values based on accumulated data. Expression data from normal cells provides a means for grading the severity of the cancer state.


In another preferred embodiment, expression of a marker is assessed by preparing genomic DNA or mRNA/cDNA (i.e. a transcribed polynucleotide) from cells in a subject sample, and by hybridizing the genomic DNA or mRNA/cDNA with a reference polynucleotide which is a complement of a polynucleotide comprising the marker, and fragments thereof. cDNA can, optionally, be amplified using any of a variety of polymerase chain reaction methods prior to hybridization with the reference polynucleotide. Expression of one or more markers can likewise be detected using quantitative PCR (QPCR) to assess the level of expression of the marker(s). Alternatively, any of the many known methods of detecting mutations or variants (e.g. single nucleotide polymorphisms, deletions, etc.) of a marker of the invention may be used to detect occurrence of a mutated marker in a subject.


In a related embodiment, a mixture of transcribed polynucleotides obtained from the sample is contacted with a substrate having fixed thereto a polynucleotide complementary to or homologous with at least a portion (e.g. at least 7, 10, 15, 20, 25, 30, 40, 50, 100, 500, or more nucleotide residues) of a marker of the invention. If polynucleotides complementary to or homologous with are differentially detectable on the substrate (e.g. detectable using different chromophores or fluorophores, or fixed to different selected positions), then the levels of expression of a plurality of markers can be assessed simultaneously using a single substrate (e.g. a “gene chip” microarray of polynucleotides fixed at selected positions). When a method of assessing marker expression is used which involves hybridization of one nucleic acid with another, it is preferred that the hybridization be performed under stringent hybridization conditions.


In another embodiment, a combination of methods to assess the expression of a marker is utilized.


Because the compositions, kits, and methods of the invention rely on detection of a difference in expression levels or copy number of one or more markers of the invention, it is preferable that the level of expression or copy number of the marker is significantly greater than the minimum detection limit of the method used to assess expression or copy number in at least one of normal cells and cancerous cells.


3. Methods for Detection of Expressed Protein


The activity or level of a marker protein can also be detected and/or quantified by detecting or quantifying the expressed polypeptide. The polypeptide can be detected and quantified by any of a number of means well known to those of skill in the art. These may include analytic biochemical methods such as electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, and the like, or various immunological methods such as fluid or gel precipitin reactions, immunodiffusion (single or double), immunoelectrophoresis, radioimmunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, western blotting, and the like. A skilled artisan can readily adapt known protein/antibody detection methods for use in determining whether cells express a marker of the present invention.


A preferred agent for detecting a polypeptide of the invention is an antibody capable of binding to a polypeptide corresponding to a marker of the invention, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab′)2) can be used. The term “labeled”, with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin.


In a preferred embodiment, the antibody is labeled, e.g. a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody). In another embodiment, an antibody derivative (e.g. an antibody conjugated with a substrate or with the protein or ligand of a protein-ligand pair {e.g. biotin-streptavidin}), or an antibody fragment (e.g. a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically with a protein corresponding to the marker, such as the protein encoded by the open reading frame corresponding to the marker or such a protein which has undergone all or a portion of its normal post-translational modification, is used.


Proteins from cells can be isolated using techniques that are well known to those of skill in the art. The protein isolation methods employed can, for example, be such as those described in Harlow and Lane (Harlow and Lane, 1988, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.).


In one format, antibodies, or antibody fragments, can be used in methods such as Western blots or immunofluorescence techniques to detect the expressed proteins. In such uses, it is generally preferable to immobilize either the antibody or proteins on a solid support. Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.


One skilled in the art will know many other suitable carriers for binding antibody or antigen, and will be able to adapt such support for use with the present invention. For example, protein isolated from cells can be run on a polyacrylamide gel electrophoresis and immobilized onto a solid phase support such as nitrocellulose. The support can then be washed with suitable buffers followed by treatment with the detectably labeled antibody. The solid phase support can then be washed with the buffer a second time to remove unbound antibody. The amount of bound label on the solid support can then be detected by conventional means. Means of detecting proteins using electrophoretic techniques are well known to those of skill in the art (see generally, R. Scopes (1982) Protein Purification, Springer-Verlag, N.Y.; Deutscher, (1990) Methods in Enzymology Vol. 182: Guide to Protein Purification, Academic Press, Inc., N.Y.).


In another preferred embodiment, Western blot (immunoblot) analysis is used to detect and quantify the presence of a polypeptide in the sample. This technique generally comprises separating sample proteins by gel electrophoresis on the basis of molecular weight, transferring the separated proteins to a suitable solid support, (such as a nitrocellulose filter, a nylon filter, or derivatized nylon filter), and incubating the sample with the antibodies that specifically bind a polypeptide. The anti-polypeptide antibodies specifically bind to the polypeptide on the solid support. These antibodies may be directly labeled or alternatively may be subsequently detected using labeled antibodies (e.g., labeled sheep anti-mouse antibodies) that specifically bind to the anti-polypeptide.


In a more preferred embodiment, the polypeptide is detected using an immunoassay. As used herein, an immunoassay is an assay that utilizes an antibody to specifically bind to the analyte. The immunoassay is thus characterized by detection of specific binding of a polypeptide to an anti-antibody as opposed to the use of other physical or chemical properties to isolate, target, and quantify the analyte.


The polypeptide is detected and/or quantified using any of a number of well recognized immunological binding assays (see, e.g., U.S. Pat. Nos. 4,366,241; 4,376,110; 4,517,288; and 4,837,168). For a review of the general immunoassays, see also Asai (1993) Methods in Cell Biology Volume 37: Antibodies in Cell Biology, Academic Press, Inc. New York; Stites & Terr (1991) Basic and Clinical Immunology 7th Edition.


Immunological binding assays (or immunoassays) typically utilize a “capture agent” to specifically bind to and often immobilize the analyte (polypeptide or subsequence). The capture agent is a moiety that specifically binds to the analyte. In a preferred embodiment, the capture agent is an antibody that specifically binds a polypeptide. The antibody (anti-peptide) may be produced by any of a number of means well known to those of skill in the art.


Immunoassays also often utilize a labeling agent to specifically bind to and label the binding complex formed by the capture agent and the analyte. The labeling agent may itself be one of the moieties comprising the antibody/analyte complex. Thus, the labeling agent may be a labeled polypeptide or a labeled anti-antibody. Alternatively, the labeling agent may be a third moiety, such as another antibody, that specifically binds to the antibody/polypeptide complex.


In one preferred embodiment, the labeling agent is a second human antibody bearing a label. Alternatively, the second antibody may lack a label, but it may, in turn, be bound by a labeled third antibody specific to antibodies of the species from which the second antibody is derived. The second can be modified with a detectable moiety, e.g. as biotin, to which a third labeled molecule can specifically bind, such as enzyme-labeled streptavidin.


Other proteins capable of specifically binding immunoglobulin constant regions, such as protein A or protein G may also be used as the label agent. These proteins are normal constituents of the cell walls of streptococcal bacteria. They exhibit a strong non-immunogenic reactivity with immunoglobulin constant regions from a variety of species (see, generally Kronval, et al. (1973) J. Immunol., 111: 1401-1406, and Akerstrom (1985) J. Immunol., 135: 2589-2542).


As indicated above, immunoassays for the detection and/or quantification of a polypeptide can take a wide variety of formats well known to those of skill in the art.


Preferred immunoassays for detecting a polypeptide are either competitive or noncompetitive. Noncompetitive immunoassays are assays in which the amount of captured analyte is directly measured. In one preferred “sandwich” assay, for example, the capture agent (anti-peptide antibodies) can be bound directly to a solid substrate where they are immobilized. These immobilized antibodies then capture polypeptide present in the test sample. The polypeptide thus immobilized is then bound by a labeling agent, such as a second human antibody bearing a label.


In competitive assays, the amount of analyte (polypeptide) present in the sample is measured indirectly by measuring the amount of an added (exogenous) analyte (polypeptide) displaced (or competed away) from a capture agent (anti peptide antibody) by the analyte present in the sample. In one competitive assay, a known amount of, in this case, a polypeptide is added to the sample and the sample is then contacted with a capture agent. The amount of polypeptide bound to the antibody is inversely proportional to the concentration of polypeptide present in the sample.


In one particularly preferred embodiment, the antibody is immobilized on a solid substrate. The amount of polypeptide bound to the antibody may be determined either by measuring the amount of polypeptide present in a polypeptide/antibody complex, or alternatively by measuring the amount of remaining uncomplexed polypeptide. The amount of polypeptide may be detected by providing a labeled polypeptide.


The assays of this invention are scored (as positive or negative or quantity of polypeptide) according to standard methods well known to those of skill in the art. The particular method of scoring will depend on the assay format and choice of label. For example, a Western Blot assay can be scored by visualizing the colored product produced by the enzymatic label. A clearly visible colored band or spot at the correct molecular weight is scored as a positive result, while the absence of a clearly visible spot or band is scored as a negative. The intensity of the band or spot can provide a quantitative measure of polypeptide.


Antibodies for use in the various immunoassays described herein, can be produced as described below.


In another embodiment, level (activity) is assayed by measuring the enzymatic activity of the gene product. Methods of assaying the activity of an enzyme are well known to those of skill in the art.


In vivo techniques for detection of a biomarker protein include introducing into a subject a labeled antibody directed against the protein. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.


Certain markers identified by the methods of the invention may be secreted proteins. It is a simple matter for the skilled artisan to determine whether any particular marker protein is a secreted protein. In order to make this determination, the marker protein is expressed in, for example, a mammalian cell, preferably a human cell line, extracellular fluid is collected, and the presence or absence of the protein in the extracellular fluid is assessed (e.g. using a labeled antibody which binds specifically with the protein).


The following is an example of a method which can be used to detect secretion of a protein. About 8×105 293T cells are incubated at 37° C. in wells containing growth medium (Dulbecco's modified Eagle's medium {DMEM} supplemented with 10% fetal bovine serum) under a 5% (v/v) CO2, 95% air atmosphere to about 60-70% confluence. The cells are then transfected using a standard transfection mixture comprising 2 micrograms of DNA comprising an expression vector encoding the protein and 10 microliters of LipofectAMINE™ (GIBCO/BRL Catalog no. 18342-012) per well. The transfection mixture is maintained for about 5 hours, and then replaced with fresh growth medium and maintained in an air atmosphere. Each well is gently rinsed twice with DMEM which does not contain methionine or cysteine (DMEM-MC; ICN Catalog no. 16-424-54). About 1 milliliter of DMEM-MC and about 50 microcuries of Trans-35STM reagent (ICN Catalog no. 51006) are added to each well. The wells are maintained under the 5% CO2 atmosphere described above and incubated at 37° C. for a selected period. Following incubation, 150 microliters of conditioned medium is removed and centrifuged to remove floating cells and debris. The presence of the protein in the supernatant is an indication that the protein is secreted.


It will be appreciated that subject samples, e.g., a sample containing tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue, may contain cells therein, particularly when the cells are cancerous, and, more particularly, when the cancer is metastasizing, and thus may be used in the methods of the present invention. The cell sample can, of course, be subjected to a variety of well-known post-collection preparative and storage techniques (e.g., nucleic acid and/or protein extraction, fixation, storage, freezing, ultrafiltration, concentration, evaporation, centrifugation, etc.) prior to assessing the level of expression of the marker in the sample. Thus, the compositions, kits, and methods of the invention can be used to detect expression of markers corresponding to proteins having at least one portion which is displayed on the surface of cells which express it. It is a simple matter for the skilled artisan to determine whether the protein corresponding to any particular marker comprises a cell-surface protein. For example, immunological methods may be used to detect such proteins on whole cells, or well known computer-based sequence analysis methods (e.g. the SIGNALP program; Nielsen et al., 1997, Protein Engineering 10:1-6) may be used to predict the presence of at least one extracellular domain (i.e. including both secreted proteins and proteins having at least one cell-surface domain). Expression of a marker corresponding to a protein having at least one portion which is displayed on the surface of a cell which expresses it may be detected without necessarily lysing the cell (e.g. using a labeled antibody which binds specifically with a cell-surface domain of the protein).


The invention also encompasses kits for detecting the presence of a polypeptide or nucleic acid corresponding to a marker of the invention in a biological sample, e.g., a sample containing tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue. Such kits can be used to determine if a subject is suffering from or is at increased risk of developing cancer. For example, the kit can comprise a labeled compound or agent capable of detecting a polypeptide or an mRNA encoding a polypeptide corresponding to a marker of the invention in a biological sample and means for determining the amount of the polypeptide or mRNA in the sample (e.g., an antibody which binds the polypeptide or an oligonucleotide probe which binds to DNA or mRNA encoding the polypeptide). Kits can also include instructions for interpreting the results obtained using the kit.


For antibody-based kits, the kit can comprise, for example: (1) a first antibody (e.g., attached to a solid support) which binds to a polypeptide corresponding to a marker of the invention; and, optionally, (2) a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable label.


For oligonucleotide-based kits, the kit can comprise, for example: (1) an oligonucleotide, e.g., a detectably labeled oligonucleotide, which hybridizes to a nucleic acid sequence encoding a polypeptide corresponding to a marker of the invention or (2) a pair of primers useful for amplifying a nucleic acid molecule corresponding to a marker of the invention. The kit can also comprise, e.g., a buffering agent, a preservative, or a protein stabilizing agent. The kit can further comprise components necessary for detecting the detectable label (e.g., an enzyme or a substrate). The kit can also contain a control sample or a series of control samples which can be assayed and compared to the test sample. Each component of the kit can be enclosed within an individual container and all of the various containers can be within a single package, along with instructions for interpreting the results of the assays performed using the kit.


4. Method for Detecting Structural Alterations


The invention also provides a method for assessing whether a subject is afflicted with cancer or is at risk for developing cancer by comparing the structural alterations, e.g., mutations or allelic variants, of a marker in a cancer sample with the structural alterations, e.g., mutations of a marker in a normal, e.g., control sample. The presence of a structural alteration, e.g., mutation or allelic variant in the marker in the cancer sample is an indication that the subject is afflicted with cancer.


A preferred detection method is allele specific hybridization using probes overlapping the polymorphic site and having about 5, 10, 20, 25, or 30 nucleotides around the polymorphic region. In a preferred embodiment of the invention, several probes capable of hybridizing specifically to allelic variants are attached to a solid phase support, e.g., a “chip”. Oligonucleotides can be bound to a solid support by a variety of processes, including lithography. For example a chip can hold up to 250,000 oligonucleotides (GeneChip, Affymetrix™). Mutation detection analysis using these chips comprising oligonucleotides, also termed “DNA probe arrays” is described e.g., in Cronin et al. (1996) Human Mutation 7:244. In one embodiment, a chip comprises all the allelic variants of at least one polymorphic region of a gene. The solid phase support is then contacted with a test nucleic acid and hybridization to the specific probes is detected. Accordingly, the identity of numerous allelic variants of one or more genes can be identified in a simple hybridization experiment. For example, the identity of the allelic variant of the nucleotide polymorphism in the 5′ upstream regulatory element can be determined in a single hybridization experiment.


In other detection methods, it is necessary to first amplify at least a portion of a marker prior to identifying the allelic variant. Amplification can be performed, e.g., by PCR and/or LCR (see Wu and Wallace (1989) Genomics 4:560), according to methods known in the art. In one embodiment, genomic DNA of a cell is exposed to two PCR primers and amplification for a number of cycles sufficient to produce the required amount of amplified DNA. In preferred embodiments, the primers are located between 150 and 350 base pairs apart.


Alternative amplification methods include: self sustained sequence replication (Guatelli, J. C. et al., (1990) Proc. Natl. Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh, D. Y. et al., (1989) Proc. Natl. Acad. Sci. USA 86:1173-1177), Q-Beta Replicase (Lizardi, P. M. et al., (1988) Bio/Technology 6:1197), and self-sustained sequence replication (Guatelli et al., (1989)Proc. Nat. Acad. Sci. 87:1874), and nucleic acid based sequence amplification (NABSA), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.


In one embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence at least a portion of a marker and detect allelic variants, e.g., mutations, by comparing the sequence of the sample sequence with the corresponding reference (control) sequence. Exemplary sequencing reactions include those based on techniques developed by Maxam and Gilbert (Proc. Natl. Acad Sci USA (1977) 74:560) or Sanger (Sanger et al. (1977) Proc. Nat. Acad. Sci. 74:5463). It is also contemplated that any of a variety of automated sequencing procedures may be utilized when performing the subject assays (Biotechniques (1995) 19:448), including sequencing by mass spectrometry (see, for example, U.S. Pat. No. 5,547,835 and international patent application Publication Number WO 94/16101, entitled DNA Sequencing by Mass Spectrometry by H. Köster; U.S. Pat. No. 5,547,835 and international patent application Publication Number WO 94/21822 entitled “DNA Sequencing by Mass Spectrometry Via Exonuclease Degradation” by H. Köster), and U.S. Pat. No. 5,605,798 and International Patent Application No. PCT/US96/03651 entitled DNA Diagnostics Based on Mass Spectrometry by H. Köster; Cohen et al. (1996) Adv Chromatogr 36:127-162; and Griffin et al. (1993) Appl Biochem Biotechnol 38:147-159). It will be evident to one skilled in the art that, for certain embodiments, the occurrence of only one, two or three of the nucleic acid bases need be determined in the sequencing reaction. For instance, A-track or the like, e.g., where only one nucleotide is detected, can be carried out.


Yet other sequencing methods are disclosed, e.g., in U.S. Pat. No. 5,580,732 entitled “Method of DNA sequencing employing a mixed DNA-polymer chain probe” and U.S. Pat. No. 5,571,676 entitled “Method for mismatch-directed in vitro DNA sequencing.”


In some cases, the presence of a specific allele of a marker in DNA from a subject can be shown by restriction enzyme analysis. For example, a specific nucleotide polymorphism can result in a nucleotide sequence comprising a restriction site which is absent from the nucleotide sequence of another allelic variant.


In a further embodiment, protection from cleavage agents (such as a nuclease, hydroxylamine or osmium tetroxide and with piperidine) can be used to detect mismatched bases in RNA/RNA DNA/DNA, or RNA/DNA heteroduplexes (Myers, et al. (1985) Science 230:1242). In general, the technique of “mismatch cleavage” starts by providing heteroduplexes formed by hybridizing a control nucleic acid, which is optionally labeled, e.g., RNA or DNA, comprising a nucleotide sequence of a marker allelic variant with a sample nucleic acid, e.g., RNA or DNA, obtained from a tissue sample. The double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as duplexes formed based on basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S1 nuclease to enzymatically digest the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine whether the control and sample nucleic acids have an identical nucleotide sequence or in which nucleotides they are different. See, for example, Cotton et al (1988) Proc. Natl Acad Sci USA 85:4397; Saleeba et al (1992) Methods Enzymol. 217:286-295. In a preferred embodiment, the control or sample nucleic acid is labeled for detection.


In another embodiment, an allelic variant can be identified by denaturing high-performance liquid chromatography (DHPLC) (Oefner and Underhill, (1995) Am. J. Human Gen. 57:Suppl. A266). DHPLC uses reverse-phase ion-pairing chromatography to detect the heteroduplexes that are generated during amplification of PCR fragments from individuals who are heterozygous at a particular nucleotide locus within that fragment (Oefner and Underhill (1995) Am. J. Human Gen. 57:Suppl. A266). In general, PCR products are produced using PCR primers flanking the DNA of interest. DHPLC analysis is carried out and the resulting chromatograms are analyzed to identify base pair alterations or deletions based on specific chromatographic profiles (see O'Donovan et al. (1998) Genomics 52:44-49).


In other embodiments, alterations in electrophoretic mobility is used to identify the type of marker allelic variant. For example, single strand conformation polymorphism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita et al. (1989) Proc Natl. Acad. Sci. USA 86:2766, see also Cotton (1993) Mutat Res 285:125-144; and Hayashi (1992) Genet Anal Tech Appl 9:73-79). Single-stranded DNA fragments of sample and control nucleic acids are denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In another preferred embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet. 7:5).


In yet another embodiment, the identity of an allelic variant of a polymorphic region is obtained by analyzing the movement of a nucleic acid comprising the polymorphic region in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. (1985) Nature 313:495). When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing agent gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys Chem 265:1275).


Examples of techniques for detecting differences of at least one nucleotide between two nucleic acids include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide probes may be prepared in which the known polymorphic nucleotide is placed centrally (allele-specific probes) and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324:163); Saiki et al (1989) Proc. Natl. Acad. Sci. USA 86:6230; and Wallace et al. (1979) Nucl. Acids Res. 6:3543). Such allele specific oligonucleotide hybridization techniques may be used for the simultaneous detection of several nucleotide changes in different polylmorphic regions of marker. For example, oligonucleotides having nucleotide sequences of specific allelic variants are attached to a hybridizing membrane and this membrane is then hybridized with labeled sample nucleic acid. Analysis of the hybridization signal will then reveal the identity of the nucleotides of the sample nucleic acid.


Alternatively, allele specific amplification technology which depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as primers for specific amplification may carry the allelic variant of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al (1989) Nucleic Acids Res. 17:2437-2448) or at the extreme 3′ end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner (1993) Tibtech 11:238; Newton et al. (1989) Nucl. Acids Res. 17:2503). This technique is also termed “PROBE” for Probe Oligo Base Extension. In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection (Gasparini et al (1992) Mol. Cell. Probes 6:1).


In another embodiment, identification of the allelic variant is carried out using an oligonucleotide ligation assay (OLA), as described, e.g., in U.S. Pat. No. 4,998,617 and in Landegren, U. et al., (1988) Science 241:1077-1080. The OLA protocol uses two oligonucleotides which are designed to be capable of hybridizing to abutting sequences of a single strand of a target. One of the oligonucleotides is linked to a separation marker, e.g., biotinylated, and the other is detectably labeled. If the precise complementary sequence is found in a target molecule, the oligonucleotides will hybridize such that their termini abut, and create a ligation substrate. Ligation then permits the labeled oligonucleotide to be recovered using avidin, or another biotin ligand. Nickerson, D. A. et al. have described a nucleic acid detection assay that combines attributes of PCR and OLA (Nickerson, D. A. et al., (1990) Proc. Natl. Acad. Sci. (U.S.A.) 87:8923-8927. In this method, PCR is used to achieve the exponential amplification of target DNA, which is then detected using OLA.


The invention further provides methods for detecting single nucleotide polymorphisms in a marker. Because single nucleotide polymorphisms constitute sites of variation flanked by regions of invariant sequence, their analysis requires no more than the determination of the identity of the single nucleotide present at the site of variation and it is unnecessary to determine a complete gene sequence for each subject. Several methods have been developed to facilitate the analysis of such single nucleotide polymorphisms.


In one embodiment, the single base polymorphism can be detected by using a specialized exonuclease-resistant nucleotide, as disclosed, e.g., in Mundy, C. R. (U.S. Pat. No. 4,656,127). According to the method, a primer complementary to the allelic sequence immediately 3′ to the polymorphic site is permitted to hybridize to a target molecule obtained from a particular animal or human. If the polymorphic site on the target molecule contains a nucleotide that is complementary to the particular exonuclease-resistant nucleotide derivative present, then that derivative will be incorporated onto the end of the hybridized primer. Such incorporation renders the primer resistant to exonuclease, and thereby permits its detection. Since the identity of the exonuclease-resistant derivative of the sample is known, a finding that the primer has become resistant to exonucleases reveals that the nucleotide present in the polymorphic site of the target molecule was complementary to that of the nucleotide derivative used in the reaction. This method has the advantage that it does not require the determination of large amounts of extraneous sequence data.


In another embodiment of the invention, a solution-based method is used for determining the identity of the nucleotide of a polymorphic site. Cohen, D. et al. (French Patent 2,650,840; PCT Appln. No. WO91/02087). As in the Mundy method of U.S. Pat. No. 4,656,127, a primer is employed that is complementary to allelic sequences immediately 3′ to a polymorphic site. The method determines the identity of the nucleotide of that site using labeled dideoxynucleotide derivatives, which, if complementary to the nucleotide of the polymorphic site will become incorporated onto the terminus of the primer.


An alternative method, known as Genetic Bit Analysis or GBA™ is described by Goelet, P. et al. (PCT Appln. No. 92/15712). The method of Goelet, P. et al. uses mixtures of labeled terminators and a primer that is complementary to the sequence 3′ to a polymorphic site. The labeled terminator that is incorporated is thus determined by, and complementary to, the nucleotide present in the polymorphic site of the target molecule being evaluated. In contrast to the method of Cohen et al. (French Patent 2,650,840; PCT Appln. No. WO91/02087) the method of Goelet, P. et al. is preferably a heterogeneous phase assay, in which the primer or the target molecule is immobilized to a solid phase.


Several primer-guided nucleotide incorporation procedures for assaying polymorphic sites in DNA have been described (Komher, J. S. et al., (1989) Nucl. Acids. Res. 17:7779-7784; Sokolov, B. P., (1990) Nucl. Acids Res. 18:3671; Syvanen, A.-C., et al., (990) Genomics 8:684-692; Kuppuswamy, M. N. et al., (1991) Proc. Natl. Acad. Sci. (U.S.A.) 88:1143-1147; Prezant, T. R. et al., (1992) Hum. Mutat. 1:159-164; Ugozzoli, L. et al., (1992) GATA 9:107-112; Nyren, P. (1993) et al., Anal Biochem. 208:171-175). These methods differ from GBA™ in that they all rely on the incorporation of labeled deoxynucleotides to discriminate between bases at a polymorphic site. In such a format, since the signal is proportional to the number of deoxynucleotides incorporated, polymorphisms that occur in runs of the same nucleotide can result in signals that are proportional to the length of the run (Syvanen, A. C., et al., (1993) Amer. J. Hum. Genet. 52:46-59).


For determining the identity of the allelic variant of a polymorphic region located in the coding region of a marker, yet other methods than those described above can be used. For example, identification of an allelic variant which encodes a mutated marker can be performed by using an antibody specifically recognizing the mutant protein in, e.g., immunohistochemistry or immunoprecipitation. Antibodies to wild-type marker or mutated forms of markers can be prepared according to methods known in the art.


Alternatively, one can also measure an activity of a marker, such as binding to a marker ligand. Binding assays are known in the art and involve, e.g., obtaining cells from a subject, and performing binding experiments with a labeled ligand, to determine whether binding to the mutated form of the protein differs from binding to the wild-type of the protein.


B. Pharmacogenomics


Agents or modulators which have a stimulatory or inhibitory effect on amount and/or activity of a marker of the invention can be administered to individuals to treat (prophylactically or therapeutically) cancer in the subject. In conjunction with such treatment, the pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug) of the individual may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug. Thus, the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the amount, structure, and/or activity of the invention in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.


Pharmacogenomics deals with clinically significant variations in the response to drugs due to altered drug disposition and abnormal action in affected persons. See, e.g., Linder (1997) Clin. Chem. 43(2):254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drugs act on the body are referred to as “altered drug action.” Genetic conditions transmitted as single factors altering the way the body acts on drugs are referred to as “altered drug metabolism”. These pharmacogenetic conditions can occur either as rare defects or as polymorphisms. For example, glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common inherited enzymopathy in which the main clinical complication is hemolysis after ingestion of oxidant drugs (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans.


As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymorphisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19) has provided an explanation as to why some subjects do not obtain the expected drug effects or show exaggerated drug response and serious toxicity after taking the standard and safe dose of a drug. These polymorphisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for CYP2D6 is highly polymorphic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, a PM will show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite morphine. The other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.


Thus, the amount, structure, and/or activity of a marker of the invention in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymorphic alleles encoding drug-metabolizing enzymes to the identification of an individual's drug responsiveness phenotype. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with a modulator of amount, structure, and/or activity of a marker of the invention.


C. Monitoring Clinical Trials


Monitoring the influence of agents (e.g., drug compounds) on amount, structure, and/or activity of a marker of the invention can be applied not only in basic drug screening, but also in clinical trials. For example, the effectiveness of an agent to affect marker amount, structure, and/or activity can be monitored in clinical trials of subjects receiving treatment for cancer. In a preferred embodiment, the present invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, antibody, nucleic acid, antisense nucleic acid, ribozyme, small molecule, RNA interfering agent, or other drug candidate) comprising the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the agent; (ii) detecting the amount, structure, and/or activity of one or more selected markers of the invention in the pre-administration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the amount, structure, and/or activity of the marker(s) in the post-administration samples; (v) comparing the level of expression of the marker(s) in the pre-administration sample with the amount, structure, and/or activity of the marker(s) in the post-administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly. For example, increased administration of the agent can be desirable to increase amount and/or activity of the marker(s) to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent can be desirable to decrease amount and/or activity of the marker(s) to lower levels than detected, i.e., to decrease the effectiveness of the agent.


EXAMPLES

This invention is further illustrated by the following examples which should not be construed as limiting. The contents of all references, figures, Sequence Listing, patents and published patent applications cited throughout this application are hereby incorporated by reference.


Example 1
A. Materials and Methods
Primary Tumors and Cell Lines

All cell lines were acquired from the American Type Culture Collection (ATCC) or the German Collection of Microorganisms and Cell Cultures (DSMZ). All fresh-frozen specimens of primary pancreatic ductal adenocarcinoma were obtained from the Memorial Sloan-Kettering Cancer Center tumor bank and histology was confirmed by hematoxylin and eosin (H&E) staining prior to inclusion in the study (Table 2). Table 2 lists the cell lines analyzed by array-CGH and expression profiling in the study.


Array-CGH Profiling on cDNA Microarrays


Genomic DNA was fragmented and random-prime labeled according to published protocol (Pollack, J. R., et al. (1999) Nat Genet. 23, 41-6) with modifications. Labeled DNAs were hybridized to human cDNA microarrays containing 14,160 cDNA clones (Agilent Technologies™, Human 1 clone set), for which approximately 9,420 unique map positions were defined (NCBI, Build 33). The median interval between mapped elements is 100.1 kilobase, 92.8% of intervals are less than 1 megabase, and 98.6% are less than 3 megabases.


Fluorescence ratios of scanned images of the arrays were calculated and the raw array-CGH profiles were processed to identify statistically significant transitions in copy number using a segmentation algorithm which employs permutation to determine the significance of change points in the raw data (Olshen, A. B., and Venkatraman, E. S. (2002) ASA Proceedings of the Joint Statistical Meetings, 2530-2535; Ginzinger, D. G. (2002) Exp Hematol 30, 503-12). Each segment was assigned a Log2 ratio that is the median of the contained probes. The data was centered by the tallest mode in the distribution of the segmented values. After mode-centering, gains and losses were defined as Log2 ratios of greater than or equal to +0.13 or −0.13 (+/−4 standard deviations of the middle 50% quantile of data), and amplification and deletion as ratio greater than 0.52 or less than −0.58, respectively (i.e., 97% or 3% quantiles) (FIG. 4).


Automated Locus Definition

Loci were defined by an automated algorithm applied to the segmented data based on the following rules:


1. Segments above 97th percentile or below 3rd percentile were identified as altered.


2. If two or more altered segments are adjacent in a single profile or separated by less than 500KB, the entire region spanned by the segments was considered to be an altered span.


3. Highly altered segments or spans that were shorter than 20 MB were retained as “informative spans” for defining discrete locus boundaries. Longer regions were not discarded, but were not included in defining locus boundaries.


4. Informative spans were compared across samples to identify overlapping groups of positive-value or negative-value segments; each group defined a locus.


5. Minimal common regions (MCRs) were defined as contiguous spans having at least 75% of the peak recurrence as calculated by counting the occurrence of highly altered segments. If two MCRs were separated by a gap of only one probe position, they were joined. If there were more than 3 MCRs in a locus, the whole region was reported as a single complex MCR.


MCR Characterization

For each MCR, the peak segment value was identified. Recurrence of gain or loss was calculated across all samples based on the lower thresholds previously defined (˜+/−0.13). As an additional measure of recurrence independent of thresholds for segment value or length, Median Aberration (MA) was calculated for each probe position by taking the median of all segment values above zero for amplified regions, below zero for deleted regions. This pair of values was compared to the distribution of values obtained after permuting the probe labels independently in each sample profile. Where the magnitude of the MA exceeds 95% of the permuted averages, the region was marked as significantly gained or lost, and this was used in the voting system for prioritization. The number of known genes and GENSCAN model predicted genes were counted based on the April 2003 human assembly at UCSC (genome.ucsc.edu).


QPCR Verification

PCR primers were designed to amplify products of 100-150 bp within target and control sequences. Primers for control sequences in each cell line were designed within a region of euploid copy number as shown by array-CGH analysis. Quantitative PCR was performed by monitoring in real-time the increase in fluorescence of SYBR Green dye (Qiagen™) with an ABI 7700 sequence detection system (Perkin Elmer Life Sciences™). Relative gene copy number was calculated by the comparative Ct method (Ginzinger, D. G. (2002) Exp Hematol 30, 503-12).


Expression Profiling on Affymetrix GeneChip™

Biotinylated target cRNA was generated from total sample RNA and hybridized to human oligonucleotide probe arrays (U133A, Affymetrix™, Santa Clara, Calif.) according to standard protocols (Golub, T. R., et al. (1999) Science 286, 531-7). Expression values for each gene were standardized by Log2 ratio to a middle value for the sample set, defined as the midpoint between 25% and 75% quantiles, and were mapped to genomic positions based on NCBI Build 33 of the human genome.


Integrated Copy Number and Expression Analysis

Array-CGH data was interpolated such that each expression value can be mapped to its corresponding copy number value. In order to maximize detection of focal CNAs, two separate interpolations were calculated: one selecting the higher bounding CGH probe and one choosing the lower. For each gene position, the samples were grouped based on whether array-CGH shows altered copy number or not based on interpolated CGH value. The effect of gene dosage on expression was measured by calculating a gene weight defined as the difference of the means of the expression value in the altered and unaltered sample groups divided by the sum of the standard deviations of the expression values in altered and unaltered sample groups (Hyman, E., et al. (2002) Cancer Res 62, 6240-5). The significance of the weight for each gene was estimated by permuting the sample labels 10,000 times and applying an alpha threshold of 0.05.


B. Results

Comprehensive Catalogue of CNAs in the Pancreatic Adenocarcinoma Genome


From a total of 75 primary pancreatic tumor specimens, 13 samples were identified that possessed greater than 60% neoplastic cellularity. Genomic DNAs from these primary tumor samples, along with DNAs derived from 24 established pancreatic cancer cell lines, e.g., pancreatic adenocarcinoma cell lines (Table 2), were subjected to genome-wide array-CGH profiling using a cDNA-based array platform that offers a median resolution of 100 kB. To facilitate identification of significant copy number events in these array-CGH profiles, a modified version of the circular binary segmentation methodology developed by Olshen and colleagues was employed (Olshen, A. B., and Venkatraman, E. S. (2002) ASA Proceedings of the Joint Statistical Meetings 2530-2535; Ginzinger, D. G. (2002) Exp Hematol 30, 503-12; Golub, T. R., et al. (1999) Science 286, 531-7; Hyman, E., et al. (2002) Cancer Res 62, 6240-5; Lucito, R., et al. (2003) Genome Res 13, 2291-305). This algorithm applies nonparametric statistical testing to identify and distinguish discrete copy number transition points from chance noise events in the primary dataset. As shown in FIG. 1A, the segmented array-CGH profiles readily identified large regional changes that are typically of low amplitude, hereafter referred to as ‘gain’ or ‘loss’. Similarly, focal high amplitude alterations representing ‘amplification’ or ‘deletion’ are evident in both primary tumor specimens and tumor cell lines (FIG. 1). Recurrence frequencies of the CNAs reported here match the frequencies described in the published literature (Solinas-Toldo, S., et al. (1996) Cancer Res 56, 3803-7; Mahlamaki, E. H., et al. (1997) Genes Chromosomes Cancer 20, 383-91; Mahlamaki, E. H., et al. (2002) Genes Chromosomes Cancer 35, 353-8; Fukushige, S., et al. (1997) Genes Chromosomes Cancer 19:161-9; Curtis, L. J., et al. (1998) Genomics 53, 42-55; Ghadimi, B. M., et al. (1999) Am J Pathol 154, 525-36; Armengol, G., et al. (2000) Cancer Genet Cytogenet 116, 133-41) (FIG. 1B). There is also strong concordance between primary tumors and cell lines with respect to gains on 3q, 8q and 20q and losses on I p, 3p, 6q, 9p, 17p and 18q (see FIG. 5 and FIG. 6). However, some differences were evident between primary tumor and cell line datasets and are likely attributable to the cellular heterogeneity within primary tumor samples and/or culture-induced genetic adaptation in the cell lines.


The identification of many novel CNAs, along with the high degree of structural complexity within each CNA, prompted the implementation of objective criteria to define and prioritize CNAs across the dataset. To that end, a locus-identification algorithm was developed that defines informative CNAs on the basis of size and achievement of a high significance threshold for the amplitude of change. Overlapping CNAs from multiple profiles are then merged in an automated fashion to define a discrete “locus” of regional copy number change, the bounds of which represent the combined physical extent of these overlapping CNAs (FIG. 1C). Each locus is characterized by a peak profile, the width and amplitude of which reflect the contour of the most prominent amplification or deletion for that locus. Furthermore, within each locus, one or more minimal common region (MCRs) can be identified across multiple tumor samples (FIG. 1C), with each MCR potentially harboring a distinct cancer-relevant gene targeted for copy number alteration across the sample set.


The locus identification algorithm is highly effective in delineating more discrete CNAs within previously described larger regions of gain or loss. For example, chromosome 6q has been reported as one of the most frequently deleted regions in pancreatic adenocarcinoma, but no validated tumor suppressor gene has yet been assigned to this locus. Analysis of 6q loss in the dataset presented herein has identified 5 distinct MCRs that range in size from 2.4 to 12.8 Mb, raising the possibility that there may be multiple targets for 6q loss. Notably, two of these MCRs (Table 3, Locus #75 and #76) coincide with previously identified regions of common allelic loss (Abe, T., et al. (1999) Genes Chromosomes Cancer 25, 60-4), an observation that provides further validation for the analytical approach described herein.


Selection of High-Priority Loci

The above locus-identification algorithm defined 287 discrete MCRs (from 256 independent autosomal loci) within this dataset and annotated each in terms of recurrence, amplitude of change and representation in both cell lines and primary tumors. Based upon extensive experience with this platform across many tumor types, recurrence across multiple independent samples and high amplitude signals are the two features most predictive of verification by independent assays. Hence, these discrete MCRs were prioritized based on four criteria that include (1) recurrence of high-threshold amplification or deletion (above 97th percentile or below 3rd percentile) in at least two specimens, (2) presence of a high-threshold event in at least one primary tumor specimen, (3) statistically significant Median Aberration (see M&M), and (4) a peak amplitude of equal to or greater than absolute Log2 value of 0.8 in either a cell line or primary tumor (beyond 0.5% quantiles).


Implementation of this prioritization scheme yielded 64 MCRs within 54 independent loci that satisfied at least three of the four criteria (Table 1). In Table 1, the high-confidence recurrent CNAs in pancreatic adenocarcinoma are depicted. For each MCR, the numbers of known (“K”) and predicted (“P”) (GenScan) transcripts (“Trspt”) are indicated. Of these, some are represented on Affymetrix™ U133A chip (“Total”), a subset of which show statistical significance (p<0.05) for copy number correlation (“Sig”). MCR recurrence is denoted as percentage of the total dataset. The numbers of primary tumors (T) or cell lines (C) with gain or loss, and amplification or deletion, are listed, respectively. Candidate genes within a locus for which there is literature support for involvement in pancreatic cancer are listed. Black diamonds denote the loci where the peak did not fall within a defined MCR. MCRs in bold have been validated by QPCR. Notably, genes known to play important roles in the pathogenesis of pancreatic adenocarcinoma—the p16INK4A and TP53 tumor suppressors and the MYC, KRAS2 and AKT2 oncogenes—were present within these high-confidence loci (Table 1). Within the prioritized MCRs, there was an average recurrence rate for gain/loss of 38% across the entire dataset and the maxima or minima absolute Log2 values for 34 of these 64 MCRs are greater than 1.0, placing them significantly above the threshold defined for amplification or deletion (FIG. 4). In the majority of cases, the peak profile of a locus coincided with one of the MCRs (47 of 54 Loci, Table 1) (Albertson, D. G., et al. (2000) Nat Genet. 25, 144-6). The median size of these 64 prioritized MCRs is 2.7 Mb, with 21 MCRs (33%) spanning 1 Mb or less (Table 1). Residing within these 21 highly focal MCRs with a median size of 0.33 Mb, there are on average 15 annotated and 8 GENSCAN predicted genes, rendering them highly attractive for target identification.


The confidence-level ascribed to these prioritized loci was further validated by real-time quantitative PCR (QPCR), which demonstrated 100% concordance with 16 selected MCRs defined by array-CGH (Table 1). For example, the MCR of an amplified locus at 7q21.11-7q32.2 was readily confirmed by QPCR (FIG. 2A). Furthermore, QPCR analyses also verified the structural details of complex CNAs reported by array-CGH. As shown in FIG. 2B, QPCR precisely mirrored each component of the complex 9p21 locus in HUP-T3, including homozygous deletion of p16INK4A, the known target for this CNA. Such detailed structural information will prove useful in dissecting the mechanisms responsible for the genesis of these cancer-associated chromosomal aberrations.


When high-priority MCRs in Table 1 were combined with an additional 81 moderate-priority MCRs (within 66 distinct loci) satisfying 2 out of 4 criteria, a genomic characterization produced a list of 145 MCRs within 121 independent loci (Table 3). Table 3 shows the combined list of 145 MCRs in 121 independent loci, including 64 high-confidence MCRs (≧3 votes) and 81 moderate-priority (2 votes) MCRs, that were prioritized by the automated algorithm as described herein. Each locus is assigned to a cytogenetic band, while the actual extent of the locus is defined more precisely by its physical location on the chromosome (in Mb) (NCBI, Built 33). The overall contour of the locus is reflected by the maxima/minima profile, which defines the most prominent point of amplification or deletion within the locus by its width (defined in physical Mb) and amplitude. Each locus is furthered defined by one or more Minimal Common Regions (MCRs), depending on the cytogenetic complexity of the locus. Each MCR is defined in a similar manner. In addition, the number of known and predicted (GENSCAN) transcripts as well as the total number of Affymetrix-represented genes and those with p-value<0.05 are shown for each MCR. MCR recurrence is denoted as percentage of the total dataset. The number of primary tumors (T) or cell lines (C) with gain or loss (90% and 10% quantiles, respectively) is listed. Furthermore, the subset of these tumors with gain/loss that meet the threshold for amplification or deletion (97% and 3% quantiles, respectively) are also indicated. The boundaries of the MCRs of each locus have been defined based on conservative parameters.


Integrated Analysis of Copy Number and Expression Information.

Copy number aberrations and their associated impact on gene expression patterns represent a common mechanism of oncogene activation and tumor suppressor inactivation. Indeed, integration of copy number and transcriptional profile datasets revealed a consistent influence of gene dosage on mRNA expression globally across the genome (FIG. 6) (Hyman, E., et al. (2002) Cancer Res 62, 6240-5; Pollack, J. R., et al. (2002) Proc Natl Acad Sci USA 99, 12963-8). Conversely, as previously demonstrated (Platzer, P., et al. (2002) Cancer Res 62, 1134-8), only a subset of genes within any given CNA show copy-number-driven expression changes—a feature that provides a first-pass means of distinguishing bystanders from potential cancer gene targets within the CNA. As a case in point, a novel locus of amplification on chromosome 17 in the cell line Hup-T3 (Locus # 21, Table 1) contains 455 genes of which 151 are present on the Affymetrix U133A array. Of these 151 genes, only 19 exhibited increased transcript levels above 2-fold. Moreover, these 19 genes reside within the peak of this locus (FIG. 3A). Similar correlations can be established in regions of deletion. For example, the 9p21 deletion locus in the BxPC-3 cell line demonstrated that only 5 out of 91 genes residing within the MCR show undetectable or decreased expression below 2-fold (FIG. 3B). Examination of p16INK4A, the known target for deletion, across the entire sample set shows that 11 of 24 cell lines show low or absent expression, of which 5 showed homozygous deletion while the remaining 6 were present at the DNA level (FIG. 3C). In the latter, epigenetic silencing is the presumed mechanism of p16INK4a inactivation.


In many cancer types, including pancreatic adenocarcinoma, the true cell-of-origin remains unknown and thus a premalignant physiological frame-of-reference is not available. In the examples above, a model for interfacing copy number and expression profiles by midpoint-centering the expression data and calculating a weighted statistic for assignment of significance values to genes with correlated copy number and expression was applied (Golub, T. R., et al. (1999) Science 286, 531-7; Hyman, E., et al. (2002) Cancer Res 62, 6240-5) (see M&M). Using this approach, the genes residing within the 64 high-confidence MCRs (Table 1) were prioritized based on the correlation of their expression with gene dosage. While only a subset of genes are represented, the Affymetrix™ U133A array permitted inclusion of 1,926 genes out of a total of 4,742 genes residing within these MCRs for this analysis. By weighing each of these 1,926 genes based on the magnitude of its expression alteration and its representation within CNAs across the dataset, the integrated copy number and expression analysis yielded a list of 603 genes that show a statistically significant association between gene copy number and mRNA expression (p<0.05, Tables 4 and 5). Tables 4 and 5 show the genes located within high-priority MCRs (Table 1) and having highly significant correlation between gene expression and gene dosage (p<0.05). The chromosome, physical position in Mb, Gene Weight (see M&M) and p-value are listed. Affymetrix probe(s) number(s) corresponding to each GenBank Accession Number (“GI” Number) and UniGene ID are listed, along with SEQ ID NOs for each nucleic acid and protein sequence. P-Values were calculated by permutation analysis of a gene weight statistic. Of these, 336 are located within regions of amplifications and 267 within regions of deletions. Importantly, among these 603 genes were known pancreatic cancer genes such as MYC (13), p16INK4A (Rozenblum, E., et al. (1997) Cancer Res 57, 1731-4; Caldas, C., et al. (1994) Nat Genet. 8, 27-32) and DUSP6 (Furukawa, T., et al. (2003) Am J Pathol 162, 1807-15) (Tables 4 and 5), thus reinforcing the value of integrating both copy number and expression information.


While incomplete representation of known and predicted genes on the Affymetrix U133A expression array precluded assessment of all possible target genes, the complementary analysis of array-CGH and expression profiles presented herein serves to prioritize the list of available cancer gene candidates and provides a basis for focus on a subset of high-probability candidates. In addition, integrating genomic datasets across species may also prove effective in facilitating cancer gene identification. A particularly productive path for oncogene identification may be the analysis of common integration sites (CISs) present in retrovirally-promoted leukemias and lymphomas (Neil, J. C. & Cameron, E. R. (2002) Cancer Cell 2, 253-5). Consistent with the paradigm that proviral integration primarily serves to activate endogenous proto-oncogene (Neil, J. C. & Cameron, E. R. (2002) Cancer Cell 2, 253-5), syntenic mapping of 232 CISs to the human genome (Akagi K., et al. (2004) Nucl. Acids Res 32) uncovered 19 CISs residing within MCRs of amplified loci in Table 1, whereas only 10 would be expected by chance alone (p<0.006). On the contrary, MCRs within regions of loss or deletion contained only 16 CIS's whereas 14.4 would have been expected by chance alone. Thus, the abundance of CIS's mapping to amplified loci may represent genes with pathogenetic relevance in mouse models of tumor progression as well as in human cancer, although there may be possible cell-type specific roles for these candidate genes.


EQUIVALENTS

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.









TABLE 1





List of High-Confidence MCRs.

















Minimal Common



Regions (MCRs)













Cytogenetic
Locus
Locus Peak Profile

Size













#
Band
Boundary (Mb)
Pos (MB)
Max/Min
Position (MB)
(MB)










Gain and Amplification













1
1p13.1-p12
116.83-119.49
119.07-119.2 
1.56
116.83-119.49
2.65


2
5p15.33-p15.31
0.28-6.69
0.28-0.51
0.79
0.28-6.69
6.4


3
5q31.1-q31.1
133.51-134.33
133.58-133.95
0.97
133.53-133.56
0.04


4
6p22.1-p21.32
28.12-32.72
31.98-32.44
1.36
31.98-32.11
0.13


5
6p21.1-p21.1
42.91-43.19
42.98-43.15
0.84
42.91-43.03
0.12


6
7p22.3-p22.1
0.72-4.53
0.72-2.48
0.93
0.72-2.28
1.56


7
7p15.1-p14.3
30.12-31.56
 30.5-30.81
0.82
30.12-31.66
1.44


8
7q11.21-q21.11
64.84-77.18
64.95-64.95
1.06
64.95-65.85
0.9


9
7q21.11-q32.2
 79.45-129.46
97.86-98.55
3.06
 92.33-112.27
19.94


10
8p12-p11.21
 37.7-41.76
37.72-38.02
1.71
 37.7-38.45
0.75







38.68-39.52
0.84


11
8q12.1-q12.3
59.09-63.68
59.23-62.26
0.58
59.23-60.02
1.6


12
8q21.3-q24.3
 90.7-145.83
133.72-134.16
1.78
118.97-145.83
26.86


13
9p21.3-p13.2
23.68-37.87
35.65-36.56
0.86
 35.6-36.56
0.96


14
11q14.1-q14.2
78.15-86.74
82.76-85.89
0.9
83.39-86.21
2.82


15
12p12.3-q13.13
 16.6-53.06
 20.7-22.54
2.52
21.82-22.39
0.57


16
12q15-q15
68.27-68.87
68.44-68.77
1.38
68.27-68.85
0.59


17
13q12.11-q14.13
18.68-43.95
19.21-24.22
0.89
20.65-24.85
4.19


18
13q34-q34
112.84-113.06
112.85-113.01
0.82
112.84-113.06
0.23


19
14q11.2-q24.3
18.82-74.47
 73.56-74.47*
1.08
28.12-33.17
5.05







52.41-63.53
11.12


20
17q12-q23.2
37.48-56.39
43.53-43.53
2.37
38.95-55.26
16.31


21
17q23.2-q25.3
59.83-79.64
 62.33-62.47*
3.61
74.02-74.22
0.2


22
18p11.21-q12.1
12.02-28.55
19.67-21.37
2.12
19.12-21.37
2.26


23
19p13.11-q13.32
23.32-50  
 45.03-45.39*
3.84
41.41-44.6 
3.2


24
19q13.32-q13.43
50.06-63.76
50.65-50.65
1.59
50.06-62.89
12.83


25
20p13-q13.33
 0.33-63.41
58.54-63.41
1.34
25.68-31.48
5.8







58.24-63.41
6.17


26
22q11.1-q12.1
14.65-26.58
22.7-22.7
1.04
22.64-22.83
0.2


27
22q12.2-q12.2
28.75-29.85
28.96-29.83
0.93
29.35-29.85
0.51







Loss and Deletion













28
1p36.21-p36.11
15.02-26.95
21.46-21.46
−0.94
21.08-21.56
0.48







22.82-26.95
4.13


29
1p36.3-p34.3
28.37-39.18
32.79-33.04
−0.87
28.37-31.18
2.81







32.67-34.68
2.01


30
1p21.2-p21.1
100.63-103.3 
103.25-103.25
−1.69
103.25-103.3 
0.05


31
2p25.3-p24.3
 0.21-16.09
10.92-11.3 
−0.85
 11.3-11.32
0.02


32
2p12-p11.2
75.23-89  
 79.31-85.51*
−1.03
 77.7-79.27
1.57


33
3p26.3-p24.3
1.33-18.8
13.85-14.52
−1
13.67-14.52
0.85


34
4q31.22-q32.1
148.03-158.75
149.46-153.1 
−0.93
151.71-154.37
2.65







155.09-158.56
3.47


35
4q34.1-q35.2
174.84-188.09
175.03-187.79
−1.28
174.84-188.09
13.26


36
5q23.2-q23.3
127.55-130.53
127.65-128.48
−0.62
127.55-130.53
2.98


37
6q21-q22.31
106.63-119.43
107.02-116.57
−1.11
106.63-119.43
12.8


38
6q23.3-q24.3
135.17-146.81
137.46-138.13
−0.91
135.17-146.81
11.64


39
6q27-q27
168.07-170.64
168.63-170.64
−0.72
168.07-170.64
2.47


40
8p23.3-p12
2.06-37.7
2.06-2.1*
−1.86
18.07-21.76
3.68







28.45-37.7 
9.25


41
9p24.3-p21.2
 0.47-27.18
20.77-21.31
−2.63
0.47-3.39
2.92







 6.3-23.68
18.39


42
11q14.2-q14.3
85.89-89.24
86.21-89.24
−0.9
85.89-89.24
3.35


43
12q12-q13.12
40.06-49.24
41.04-49.21
−0.71
40.06-49.24
9.18


44
12q13.12-q13.3
49.65-55.82
50.01-53.06
−0.7
49.65-63.06
3.41







63.17-55.82
2.65


45
12q14.1-q15
67.92-68.77
62.76-68.27
−0.8
62.76-68.77
6.01


46
12q21.2-q24.33
77.19-133.4
81.03-85.63
−1.19
77.19-91.44
14.25


47
16p13.3-p12.2
 0.03-23.87
0.76-23.7
−0.67
2.24-2.82
0.58


48
17p13.3-q11.1
 0.02-25.61
 8.16-14.05
−0.99
10.36-12.8 
2.44


49
18q11.2-q21.1
18.61-46.28
34.95-40.68
−1.19
34.16-43.14
8.99


50
18q22.1-q23
 60.4-77.63
74.45-76.84
−1.53
 60.4-77.63
17.23


51
19q13.2-q13.43
44.67-63.76
69.99-60.04
−1.31
59.85-60.18
0.33


52
21p11.2-q11.2
 9.96-13.43
 9.96-10.08
−1.24
 9.96-13.43
3.47


53
21q22.2-q22.3
39.76-46.94
 41.7-41.76*
−4.09
45.08-45.17
0.09







46.77-46.94
0.17


54
22q11.1-q13.2
14.49-39.46
22.7-22.7
−1.32
20.64-39.46
18.83














Minimal Common Regions (MCRs)
MCR Recurrence














# Trspt
# on U133A
Gain/Loss
Amp/Del





















#
Max Min
K
P
Total
Sig.
%
T
C
T
C
Candidates











Gain and Amplification




















1
1.55
23
47
15
2
23
2
6
2
2




2
1.05
57
164
19
8
43
1
14
0
5



3
0.97
0
0
1
0
14
1
4
1
1



4
1.36
15
3
14
1
29
1
9
0
4
NOTCH4



5
0.84
16
6
7
2
23
2
6
1
1



6
0.93
45
53
10
6
51
4
14
0
7



7
0.82
23
25
13
1
37
1
12
1
2



8
1.06
12
22
7
5
34
1
11
1
1



9
3.06
356
282
186
42
46
4
12
1
5



10
1.71
31
14
14
8
37
1
12
0
2
FGFRI




1.44
3
4
5
0
31
1
10
0
2



11
0.58
1
15
4
0
49
4
13
1
1



12
1.78
317
461
115
41
66
6
17
0
7
MYC



13
0.86
39
23
18
9
20
1
6
0
3



14
0.9
18
26
9
0
46
0
16
0
2



15
2.52
9
5
3
1
26
2
7
2
2
KRAS2



16
1.38
15
13
1
0
23
2
6
1
2



17
0.89
43
74
19
3
20
2
5
1
2



18
1.04
14
8
3
1
29
4
6
1
1



19
0.98
36
61
14
7
34
4
8
1
2




1.05
133
180
68
16
31
1
10
0
4



20
2.37
474
308
215
70
46
2
14
0
7
HER2



21
1.19
14
4
5
3
34
3
9
1
2



22
2.12
26
37
11
6
29
4
6
3
3



23
3.52
124
77
35
16
26
4
5
2
4
OZF, AKT2



24
1.59
732
396
282
65
29
3
7
1
5



25
1.26
26
31
10
1
40
3
11
1
6




1.34
129
138
57
19
46
3
13
0
7
AIB1, STK15



26
1.04
5
7
8
2
23
3
5
3
3



27
0.93
12
7
5
2
26
3
6
1
1







Loss and Deletion




















28
−0.94
14
7
2
1
46
4
12
1
2





−0.73
151
67
64
18
49
3
14
1
1



29
−0.77
28
28
17
7
37
3
10
1
2




−0.87
28
29
10
4
37
3
10
1
2



30
−1.69
5
1
0
0
49
4
13
1
5



31
−0.85
0
0
1
1
26
4
6
2
1



32
−1.03
0
12
1
0
23
2
6
1
1



33
−1
13
15
6
3
34
4
8
0
2



34
−0.93
18
39
6
2
51
4
14
0
4




−0.91
24
40
14
0
51
4
14
1
4



35
−1.28
94
194
40
8
54
6
13
0
2



36
−1
7
24
3
1
20
1
6
1
1



37
−1.11
133
181
56
21
51
5
13
0
6



38
−0.91
98
144
38
8
46
5
11
1
4



39
−0.72
22
62
8
2
46
5
11
1
3



40
−1.31
27
57
12
4
54
2
17
0
11
FEZ1




−0.95
60
131
28
16
46
1
15
0
11
NRG1



41
−1.38
15
38
8
3
60
8
13
2
9




−2.63
129
216
55
15
57
7
13
1
11
INK4A



42
−0.9
23
36
13
0
6
1
1
1
1



43
−0.73
123
129
68
18
29
2
8
1
1



44
−0.73
116
70
70
19
34
3
9
1
1




−0.69
82
69
60
4
34
2
10
1
1



45
−0.8
57
96
34
13
31
3
8
1
1



46
−1.19
63
111
27
9
37
4
9
2
4
DUSP6



47
−0.67
34
17
15
7
29
1
9
1
2



48
−0.99
18
34
14
0
57
5
15
1
5
TP53, MKK4



49
−1.19
21
94
16
5
54
6
13
0
7



50
−1.53
98
287
23
8
60
7
14
0
7



51
−1.31
55
9
12
2
31
4
7
1
2



52
−1.24
2
8
3
0
60
5
16
1
2



53
−0.9
4
3
2
2
34
5
7
0
3




−0.87
8
1
6
1
40
5
9
0
3



54
−1.32
424
350
243
65
54
5
14
0
7

















TABLE 2







List of cell lines and corresponding references used in this study









Name
Source
Reference





PA-TU-8988T
DSMZ
Elsasser et al. Virchows Arch B Cell Pathol 61(5): 295-306. 1992


PA-TU-8988S
DSMZ
Elsasser et al. Virchows Arch B Cell Pathol 61(5): 295-306. 1992


PA-TU-8902
DSMZ
Elsasser et al. Virchows Arch B Cell Pathol. 64: 201-207. 1993


DAN-G
DSMZ
Not Published


HUP-T4
DSMZ
Nishimura et al., Int. J. Pancreatol 13: 31-41. 1993


HUP-T3
DSMZ
Nishimura et al., Int. J. Pancreatol 13: 31-41. 1993


Panc 10.05
ATCC
Jaffee E M et al. Cancer J Sci Am 4: 194-203, 1998.


PL45
ATCC
Jaffee E M et al. Cancer J Sci Am 4: 194-203, 1998.


Aspc-1
ATCC
Chen et al. In Vitro. 1982 January; 18(1): 24-34.


Mpanc-96
ATCC
Peiper M et al. Int. J. Cancer 71: 993-999, 1997.


BxPC-3
ATCC
Tan M H et al. Cancer Invest. 4: 15-23, 1986.


Capan-1
ATCC
Fogh et al. JNCI 58: 209-214. 1977


Capan-2
ATCC
Fogh et al. JNCI 58: 209-214. 1977


CFPAC-1
ATCC
Schoumacher et al. PNAS 87: 4012-4016. 1990


HPAF-II
ATCC
Kim Y W et al. Pancreas 4: 353-362. 1989.


Hs766T
ATCC
Owens R. B. et al. JNCI 56: 843-849, 1976.


Panc-1
ATCC
Lieber M et al. Int J. Cancer 15: 741-747. 1975


SW1990
ATCC
Kyriazis A P et al. Cancer Res. 43: 4393-4401. 1983


MIA PaCa-2
ATCC
Yunis A A et al. Int. J. Cancer 19: 128-135. 1977


HPAC
ATCC
Gower W R et al. In vitro Cell Dev Biol. 30A: 151-161. 1994


Panc 02.03
ATCC
Jaffee E M et al. Cancer J Sci Am 4: 194-203, 1998.


Panc 02.13
ATCC
Jaffee E M et al. Cancer J Sci Am 4: 194-203, 1998.


Panc 3.27
ATCC
Jaffee E M et al. Cancer J Sci Am 4: 194-203, 1998.


Panc 08.13
ATCC
Jaffee E M et al. Cancer J Sci Am 4: 194-203, 1998.
















TABLE 3





List of High and Moderal Confidence MCRs.



















Locus
Minimal Common Regions (MCRs)
MCR Recorrance


















Locus
Peak Profits





# on

Amp/


















Cytogenetic
Boundary
Max/

Size
Max
# Trapt
U133A
Gain/Loss
Del























#
Band
(Mb)
Pos (MB)
Min
Position (MB)
(MB)
Min
K
P
Total
Sig.
%
T
C
T
C


























1
1p13.1-p12
115.53-119.49
119.07-119.2
1.55
116.83-119.48
2.65
1.55
23
47
15
2
23
2
8
2
2


2
2p11.2-p11.1
85.23-91.48
85.78-85.78
1
85.75-85.85
0.1
1
10
5
7
2
14
0
5
0
2


3
3p11.1-q12.3
87.99-102.85
95.13-102.38
0.93
90.13-99.58
9.45
0.93
18
37
5
0
29
1
9
0
2







101.35-101.63
0.48
0.93
5
7
5
5
17
0
8
0
2


4
5p15.33-p15.31
0.28-6.69
0.28-0.51
0.79
0.28-8.69
6.4
1.05
57
164
19
8
43
1
14
0
5


5
5q23.1-q31.1
115.81-132.47
131.44-132.27
1.08
115.81-132.47
16.88
1.08
110
182
44
8
28
1
8
0
1


6
5q31.1-q31.1
133.51-134.33
133.56-133.95
0.97
133.53-133.56
0.04
0.97
0
0
1
0
14
1
4
1
1


7
5q31.3-q31.3
139.21-140.22
139.48-140.22
0.65
139.21-140.04
0.83
0.65
38
16
10
0
31
2
9
0
2


9
8p22.1-p21.32
28.12-32.72
31.98-32.44
1.38
33.98-32.11
0.13
1.38
15
3
14
1
29
1
9
0
4







32.19-32.21
0.02
1.36
0
0
2
0
29
1
9
0
3


10
8p21.1-p21.1
42.91-43.19
42.98-43.15
0.54
42.91-43.03
0.12
0.64
16
8
7
2
23
2
8
1
1


11
8q24.3-q25.1
145.88-151.63
130.09-151.19
1.45
150.09-151.12
1.03
1.45
16
32
4
1
17
0
8
0
2


12
7p22.3-p22.1
0.72-4.53
0.72-2.46
0.93
0.72-2.28
1.58
0.93
46
63
10
6
51
4
14
0
7


13
7p21.3-p21.2
7.39-14.44
7.97-14.44
0.58
7.39-14.44
7.04
0.8
20
58
13
3
40
2
12
1
2


14
7p15.1-p14.3
30.12-31.58
30.5-30.81
0.82
30.12-31.58
1.44
0.82
23
25
13
1
37
1
12
1
2


15
7p13-p11.2
44.93-54.88
43.64-47.71
1.23
44.93-54.88
9.95
1.23
41
143
27
13
34
0
12
0
2


16
7p11.21-q21.11
64.54-77.18
54.95-54.95
1.06
64.95-65.65
0.9
1.08
12
22
7
5
34
1
11
1
1


17
7q21.11-q32.2
79.45-129.48
97.88-98.55
3.06
92.33-112.27
19.94
3.06
358
282
168
42
45
4
12
1
5


18
7q34-q38.1
142.7-150.22
143.26-143.38
0.94
143.38-147.82
4.44
0.94
6
38
4
0
17
0
6
0
2


19
8p23.1-p22
8.75-12.74
11.05-11.67
2.05
6.76-12.74
5.95
2.05
45
74
14
3
11
0
4
0
1


20
8p12-p11.21
37.7-41.78
37.72-38.02
1.71
37.7-28.45
0.75
1.71
31
14
14
8
37
1
12
0
2







38.68-39.52
0.84
1.44
3
4
5
0
31
1
10
0
2


21
8q12.1-q12.3
59.09-63.88
59.23-82.28
0.58
59.23-60.62
1.5
0.58
1
15
4
0
49
4
13
1
1


22
8q12.3-q13.1
83.7-67.05
65.44-88.81
0.87
63.7-67.06
3.37
0.87
20
43
6
0
43
3
12
0
1


23
8q21.3-q24.3
90.7-145.83
133.72-134.16
1.78
118.97-145.83
26.88
1.78
317
461
115
41
68
6
17
0
7


24
8p21.3-p13.2
23.88-37.87
35.68-36.56
0.88
35.8-38.56
0.96
0.85
39
23
18
9
20
1
6
0
3


25
10q25.2-q25.3
112.4-113.62
112.4-112.77
0.85
112.4-115.62
3.22
0.85
20
61
15
3
28
2
7
0
1


26
10q26.13-q28.2
124.92-129.83
126.14-126.14
0.85
124.92-126.41
1.49
0.85
9
30
7
0
23
1
7
0
2


27
11p12-q13.6
40.18-77.26
77.77.15 *
0.9
60.11-61.14
1.03
0.81
32
19
13
0
40
1
13
0
3


28
11q14.1-q14.2
78.15-80.74
82.76-85.89
0.9
83.39-86.21
2.82
0.9
18
26
9
0
46
0
16
0
2


29
12p13.33-p13.2
0.18-11.45
0.18-2.79
1.77
0.18-0.74
0.56
1.77
10
8
4
1
23
2
8
0
2







5.03-6.85
1.82
1.89
58
31
33
14
20
1
8
0
2







7.05-7.18
0.13
0.84
5
3
2
1
20
1
8
0
2


30
12p13.2-p13.1
11.95-13.06
12.18-12.77
0.8
11.95-13.03
1.11
0.95
26
23
14
8
26
3
8
0
2


31
12p12.3-q13.13
18.8-53.08
20.7-22.54
2.52
21.82-22.39
0.57
2.52
9
5
3
1
28
2
7
2
2







24.67-31.14
6.27
1.49
71
91
26
13
34
1
11
0
3


32
12q14.3-q14.3
64.81-68.08
65.95-88.08
0.93
54.61-68.03
1.46
0.93
9
19
7
0
17
1
5
0
2


33
12q15-q15
88.27-68.87
68.44-88.77
1.38
88.27-88.85
0.59
1.38
16
13
1
0
23
2
6
1
2


34
12p24.11-q24.12
108.06-110.72
108.55-110.88
0.98
108.08-110.72
0.68
0.98
59
37
28
0
20
1
6
0
3


35
12q24.31-q24.33
120.76-133.4
123.83-125.12
1.5
122.12-131.01
8.89
1.5
123
132
29
0
20
1
6
0
3


36
13q12.11-q14.15
18.88-43.95
19.21-24.22
0.89
18.88-19.21
0.53
0.89
6
6
3
0
20
2
5
0
2







20.63-24.85
4.19
0.89
43
74
19
3
20
2
5
1
2


37
13q14.2-q14.2
45.38-47.81
47.45-47.77
0.56
45.64-47.81
2.18
0.62
12
29
12
4
28
4
5
1
2


38
13q34-q34
112.84-113.08
112.85-113.01
0.82
112.84-113.06
0.23
1.04
14
8
3
1
28
4
6
1
1


39
14q11.2-q24.3
18.82-74.47
73.58-74.47 *
1.08
28.12-33.17
5.05
0.98
36
61
14
7
34
4
8
1
2







48.08-31.17
3.09
0.74
61
40
28
17
29
2
8
0
3







52.41-63.53
11.12
1.05
133
180
68
16
31
1
10
0
4


40
14q24.3-q32.11
75.23-88.88
75.25-86.96
0.93
75.23-87.87
12.83
0.83
65
138
29
9
17
0
6
0
2


41
14p32.12-q32.33
91.16-104.37
91.39-104.37
0.83
103.33-103.81
0.28
0.83
14
7
7
4
23
3
6
0
3


42
18p13.3-p13.3
0.03-4.95
3.37-3.49
1.23
1.81-1.95
0.15
0.85
7
5
4
2
11
1
3
0
2







3.27-3.34
0.26
1.23
13
13
9
3
14
0
5
0
2







3.9-4.52
0.62
0.94
11
18
6
2
14
0
5
0
2


43
16p13.3-p13.11
5.14-15.76
7.63-13.08
0.93
14.73-15.62
0.88
0.93
18
12
5
2
20
1
6
0
2


44
15p12.2-p12.1
23.7-27.3
24.03-25.18 *
1.24
23.7-27.3
3.81
1.24
41
48
0
4
28
2
7
1
0


45
17q11.2-2q11.2
29.14-30.78
30.22-30.88
0.74
30.5-30.78
0.28
0.74
7
3
1
0
28
3
6
1
1


46
17q12-q23.2
37.45-58.39
43.53-43.53
0.37
38.95-65.20
18.31
2.37
474
308
215
70
48
2
14
0
7


47
17q23.2-q25.3
59.83-79.84
82.33-82.47 *
3.81
74.02-74.22
0.2
1.19
14
4
5
3
34
3
9
1
2


48
18p11.21-q12.1
12.02-28.55
19.87-21.37
2.12
19.12-21.37
2.25
2.12
26
37
11
6
29
4
8
3
3


49
19p13.11-q13.32
23.32-50
45.03-45.39 *
3.64
41.41-44.6
3.2
3.52
124
77
35
16
26
4
5
2
4


50
19q13.32-q13.43
50.08-63.76
50.85-50.85
1.59
50.08-52.89
12.83
1.59
732
398
282
85
29
3
7
1
5


51
20p13-q13.33
0.33-63.41
58.54-83.41
1.34
25.58-31.48
5.8
1.25
28
31
10
1
40
3
11
1
5







50.87-65.38
4.71
1.16
15
69
12
4
48
3
13
0
7







38.24-83.41
5.17
1.34
129
138
57
19
48
3
13
0
7


52
22q11.1-q12.1
14.55-26.38
22.7-22.7
1.04
22.64-22.83
0.2
1.04
5
7
6
2
23
3
5
3
3


53
22q12.2-q12.3
28.73-29.85
28.96-29.83
0.93
29.38-29.83
0.61
0.93
12
7
5
2
26
3
8
1
1


54
1p38.21-p36.11
15.02-28.95
21.45-21.46
−0.94
21.06-21.56
0.48
−0.94
14
7
2
1
48
4
12
2
2







22.82-28.85
4.13
−0.73
151
67
64
18
49
3
14
1
1


55
1p35.3-p34.3
28.37-39.18
32.79-33.07 *
−0.87
28.37-31.18
2.81
−0.77
28
28
17
7
37
3
10
1
2







32.87-34.68
2.01
−0.87
28
29
10
4
37
3
10
1
2


56
1p34.2-p34.2
40.39-42.59
42.09-42.11
−0.75
40.39-42.59
2.21
−0.75
32
26
14
4
29
3
7
0
0


57
1p32.2-p31.3
56.74-60.95
56.77-59.75
−0.59
56.74-60.95
4.21
−0.67
17
48
7
2
37
3
10
1
2


58
1p21.2-p21.1
100.63-103.3
103.25-103.25
1.69
103.25-103.3
0.05
−1.69
5
1
0
0
49
4
13
1
5


59
2p25.3-p24.3
0.21-15.09
10.92-11.3 *
0.85
0.21-7.04
8.83
−0.73
36
146
14
6
34
3
9
1
3







11.3-11.32
0.02
−0.85
0
0
1
1
26
4
5
2
1


60
2p12-p11.2
75.23-69
79.31-85.51 *
−1.03
77.7-79.27
1.57
−1.03
0
12
1
0
23
2
5
1
1







86.97-89
2.03
−0.78
15
37
10
0
11
1
3
1
1


61
3p26.3-p24.3
1.33-15-8
13.65-14.52
−1
11.29-12.18
0.89
−0.78
8
8
5
2
34
5
7
1
1







13.57-14.52
0.85
−1
13
15
5
3
34
4
8
0
2


62
3p21.32-p14.1
44.51-68.3
53.69-57.41
−0.87
53.08-61.56
8.51
−0.57
68
122
25
12
43
3
12
0
4


63
4p16.3-p16.2
4.27-4.86
4.41-4.43
−0.82
4.27-4.86
0.59
−0.82
3
11
2
1
37
4
0
0
2


64
4p15.32-p15.2
17.25-26.18
24.69-25.12
−0.94
17.25-25.18
5.91
−0.94
49
107
15
5
40
4
10
0
4


65
4q13.2-q13.3
59.37-72.64
69.37-71.59
−0.64
59.37-72.84
3.47
−0.65
47
56
28
4
51
4
14
0
2


66
4q22.1-q22.1
88.79-89.29
89.13-89.13
−0.75
88.79-89.29
0.5
−0.73
5
8
6
0
48
3
13
0
2


67
4q24-q24
104.07-103.95
104.07-104.07
−0.52
104.07-103.95
−0.13
−0.62
0
0
0
0
43
2
13
0
2


68
4q27-q28.1
123.22-129.09
123.77-124.24
−0.75
123.22-129.09
5.87
−0.75
27
49
9
2
48
3
13
0
2


69
4q31.22-q32.1
148.03-158.75
149.46-153.1
−0.03
151.71-154.37
2.65
−0.93
18
39
8
2
51
4
14
0
4







155.09-156.56
3.47
−0.91
24
40
14
0
51
4
14
1
4


70
4q34.1-q35.2
174.84-188.09
175.03-157.79
−1.25
174.54-188.09
13.26
−1.28
94
194
40
8
54
5
13
0
2


71
5q23.2-q23.3
127.85-130.53
127.65-128.48
−0.62
127.55-130.53
2.98
−1
7
24
3
1
20
1
8
1
1


72
6p25.3-p12.2
0.4-62.14
0.4-2.83
−1.17
0.4-2.53
2.42
−1.17
14
50
7
1
40
6
5
0
4







25.1-27.55
1.74
−0.86
75
55
56
7
29
4
5
0
3


73
6p12.1-p12.1
56.4-57.04
56.4-58.4
−0.64
56.4-57.04
0.64
−0.72
10
7
6
1
34
5
7
1
3


74
6q21-q22.31
100.03-119.45
107.02-110.57
−1.11
105.53-119.43
12.4
−1.11
133
181
66
21
51
6
13
0
6


75
6q23.3-q24.3
135.17-146.81
137.45-138.13
−0.91
125.17-146.81
11.54
−0.91
98
144
38
8
48
5
11
1
4


76
6q26.3-q27
180.39-165.68
180.51-182.94
−0.73
160.39-165.85
5.27
−0.73
37
110
18
6
48
5
11
0
5


77
6q27-q27
168.07-170.54
168.63-170.54
−0.72
168.0-170.54
2.47
−0.72
22
82
4
2
48
5
11
1
3


78
7p21.2-p21.1
14.44-18.67
17.08-18.86
−0.9
14.44-18.87
4.43
−0.9
17
43
13
1
9
1
2
1
0


79
7q34-q35.1
142.47-147.82
143.2-143.38
−0.8
142.7-147.82
5.12
−0.8
17
83
12
0
31
3
8
2
2


80
8p23.3-p12
2.06-37.7
2.08-2.1 *
−1.84
18.07-21.78
3.68
−1.31
27
87
12
4
54
2
17
0
11







28.45-37.7
9.25
−0.95
60
131
28
16
48
1
15
0
11


81
8p12-p11.21
37.7-42.45
38.45-42.33
−1.19
37.7-39.05
1.35
−1.19
41
21
17
3
37
1
12
0
5


82
9p24.3-p21.2
0.47-27.18
20.77-21.31
−2.53
0.47-3.39
2.92
−1.38
15
38
8
3
60
6
13
2
9







5.3-23.65
18.39
−2.53
129
215
55
15
57
7
13
1
11


83
9q13-q21.11
65.19-65.51
65.26-85.51
−1.27
65.19-55.51
1.32
−1.27
11
21
0
0
40
5
9
0
2


84
9q22.33-q31.1
97.63-101.94
97.64-101.51
−0.81
97.63-101.94
4.31
−0.81
31
44
13
2
14
2
3
1
0


85
9q31.2-q31.3
105.59-109.38
106.59-107.89
−1
108.59-106.63
2.04
−1
22
19
13
8
14
1
4
0
2


86
9q34.11-q34.11
127.73-128.53
127.75-127.81
−0.91
127.73-126.53
0.8
−0.91
17
9
10
5
14
2
3
1
0


87
10p16.3-p15.3
0.29-1.17
0.29-1.08
−0.81
0.29-1.17
0.88
−0.78
12
21
7
3
40
3
11
1
4


88
10p12.33-p12.1
17.91-26.65
15.09-15.32
−3.3
17.91-26.68
6.75
−3.3
48
110
24
4
57
1
12
0
5


89
10p12.1-p11.22
27.19-33.36
27.83-32.71
−0.62
27.19-33.38
5.17
−0.78
63
94
20
9
34
2
10
1
4


90
10q22.1-q22.1
73.46-73.72
73.48-73.47
−3.23
73.48-73.72
0.28
−3.23
2
4
1
0
17
1
5
0
2


91
10q22.3-q25.3
61.01-135.27
128.41-135.27
−0.93
126.14-135.27
9.13
−0.93
92
204
34
16
20
2
7
0
2



TRUE
81.01-135.27
128.41-135.27
−0.93
126.14-135.27
0.13


92
11p15.4-p15.3
10.84-13
12.02-12.02
−3.72
10.84-13
2.16
−3.72
24
35
10
4
25
1
5
0
2


93
11p15.2-p15.1
14.56-18.47
17.38-18.43
−0.92
14.58-18.47
3.69
−0.92
38
49
27
4
29
1
9
0
3


94
11p15.1-p13
20.11-32.17
20.42-31.65
−1.2
20.11-32.17
12.05
−1.2
80
100
23
7
28
1
8
0
2


95
11q14.1-q14.2
78.15-63.59
62.78-53.39
−0.57
78.15-85.58
7.44
−0.67
25
55
11
0
8
1
1
1
1


96
11q14.2-q14.3
85.59-69.24
66.21-89.24
−0.9
85.89-59.24
3.35
−0.9
23
36
13
0
8
1
1
1
1


97
11q21-q23.2
95.74-114.13
95.74-114.13
−0.75
95.74-114.13
16.39
−0.75
188
207
75
8
17
2
4
1
1


98
11q23.5-q25
115.73-134.28
117.1-134.27
−0.7
120.33-121.54
1.21
−0.7
10
19
4
1
11
1
3
1
2


99
12q12-q13.12
40.05-49.24
41.04-49.21
−0.71
40.08-49.24
9.18
−0.73
123
129
58
18
29
2
8
1
1


100
12q13.12-q13.3
49.65-65.82
50.01-53.06
−0.7
49.85-53.06
3.41
−0.73
118
70
70
19
34
3
9
1
1







53.17-55.62
2.65
−0.69
82
69
60
4
34
2
10
1
1


101
12q14.1-q15
57.92-68.77
62.76-68.27
−0.8
52.76-68.77
6.01
−0.8
57
96
34
13
31
3
8
1
1


102
12q21.2-q24.33
77.19-133.4
51.03-55.63
−1.19
77.19-91.44
14.25
−1.19
63
111
27
9
37
4
9
2
4


103
14q24.2-q24.3
71.68-72.03
71.72-71.73
−1.1
71.68-72.03
0.35
−1.1
5
6
2
1
17
2
4
1
0


104
15p13.3-p12.2
0.03-23.97
0.76-23.7
−0.67
2.24-2.82
0.58
−0.57
34
17
15
7
29
1
9
1
2


105
15q12.1-q12.2
48.37-55.26
50.08-53.88
−1.1
48.37-53.88
5.52
−1.1
35
83
24
6
11
1
3
0
2


106
17p13.3-q11.1
0.02-25.81
8.16-14.05
−0.99
10.26-12.8
2.44
−0.99
18
34
14
0
57
5
15
1
5


107
17q11.2-q11.2
26.83-30.22
28.55-25.55
−0.83
27.5-26.97
1.17
−0.83
17
12
5
1
26
3
6
0
2


108
17q24.2-q25.3
66.73-75.58
55.98-71.88
−0.92
71.63-71.88
0.23
−0.92
10
6
2
0
23
2
6
0
3


110
18q11.2-q21.1
18.51-48.28
34.95-40.88
−1.19
34.16-43.14
8.99
−1.19
21
94
16
5
54
6
13
0
7


111
18q22.1-q23
60.4-77.83
74.45-75.84
−1.53
50.4-77.83
17.23
−1.53
96
257
23
8
50
7
14
0
7


112
19p13.3-p13.2
4.82-9.11
5.53-13.11
−0.86
6.61-6.65
0.24
−0.88
11
4
5
2
31
2
9
0
2


113
19p13.2-p13.2
12.83-13.16
12.84-13.11
−0.88
12.9-13.07
0.17
−0.88
10
3
4
3
29
2
8
0
2


114
19q13.2-q13.43
44.57-63.75
59.99-50.04
−1.31
50.65-51.57
0.92
−0.68
40
29
21
12
34
4
8
1
2







59.55-60.18
0.33
−1.31
55
9
12
2
31
4
7
1
2


115
20p13-q11.21
0.33-30.69
8.81-25.83
−1.39
5.91-25.68
19.77
−1.39
139
321
76
27
34
2
10
0
6


116
21p11.2-q11.2
9.96-13.43
9.95-10.08
−1.24
9.98-13.43
3.47
−1.24
2
8
3
0
60
5
18
1
2


117
21q22.11-q22.11
31.95-32.5
32.16-32.5
−1.18
31.95-32.5
0.55
−1.10
3
8
5
0
34
3
9
0
2


118
21q22.11-q22.12
34.19-35.52
35.05-35.42
−1.09
34.81-35.42
0.01
−1.09
5
14
4
0
40
4
10
0
3


119
21q22.13-q22.13
37.01-37.52
37.01-37.02
−1.58
37.01-37.62
0.8
−1.58
9
10
5
2
37
4
9
0
1


120
21q22.2-q22.3
39.75-46.94
41.7-41.75 *
−4.09
45.08-45.17
0.09
−0.9
4
3
2
2
34
5
7
0
3







48.77-48.94
0.17
−0.87
8
1
6
1
40
5
9
0
3


121
22q11.1-q13.2
14.49-39.40
22.7-22.7
−1.32
20.64-39.45
18.83
−1.32
424
350
243
85
54
5
14
0
7
























Affu
Affu













genes
genes



#
sign
ID

CNA
Bands

Locus
Max-Min
Max:Min_Vid
MCR_Loc
MCR_Size







 1
2
15
29
1
1p13.1-1p12
TRUE
116.83-119.49
119.07-119.2
1.85
116.83-119.49
2.65



 2
2
7
2
2
2p11.2-2p11
TRUE
85.23-91.48
95.75-65.76
1
85.75-85.65
0.1



 3
0
5
31
3
3p11.1-3q12
TRUE
87.99-102.55
95.13-102.58
0.83
90.13-99.38
9.45




5
6
32
3
3p11.1-3q12
FALSE
87.99-102.85
95.13-102.68
0.83
101.85-101.83
0.48



 4
6
18
6
4
5p15.33-8p11
TRUE
0.28-5.09
0.25-0.51
0.79
0.28-6.69
6.4



 5
6
44
77
5
5q23.1-5q31
TRUE
115.81-132.47
131.44-132.27
1.08
115.61-132.47
18.66



 6
0
1
78
6
5q31.1-6q31
TRUE
133.51-134.33
133.58-133.05
0.97
133.83-133.86
0.04



 7
0
10
113
7
5q31.3-5q31
TRUE
139.21-140.22
139.48-140.22
0.85
139.21-140.04
0.83



 9
1
14
34
8
8p22.1-6p21
TRUE
28.12-32.72
31.98-32.44
1.38
31.98-32.11
0.13




0
2
35
9
8p22.1-6p21
FALSE
28.12-32.72
31.98-32.44
1.38
32.19-32.21
0.02



10
2
7
125
10
6p21.1-8p21
TRUE
42.91-43.18
42.98-43.15
0.84
42.91-43.03
0.12



11
1
4
8
11
8q24.3-8q25
TRUE
145.88-131.63
150.09-151.19
1.45
150.09-151.12
1.03



12
6
10
38
12
7p22.3-7p22
TRUE
0.72-4.83
0.72-2.48
0.93
0.72-2.28
1.36



13
3
13
155
13
7p21.3-7p21
TRUE
7.39-14.44
7.97-14.44
0.58
7.30-14.44
7.04



14
1
13
125
14
7p13.1-7p14
TRUE
30.12-31.55
30.5-30.41
0.82
30.12-31.56
1.44



15
13
27
82
15
7p13-7p11.2
TRUE
44.93-54.88
45.64-47.71
1.23
44.93-64.88
9.95



16
5
7
9
16
7q11.21-7q2
TRUE
64.94-77.16
64.95-84.95
1.06
64.95-65.85
0.9



17
42
166
11
17
7q21.11-7q3
TRUE
79.45-129.46
97.86-95.55
3.06
92.55-112.27
19.94



18
0
4
38
18
7q34-7q36.1
TRUE
142.7-150.22
143.26-143.38
0.94
143.38-147.82
4.44



19
3
14
83
19
8p23.1-8p22
TRUE
8.76-12.74
11.06-11.67
2.05
8.78-12.74
3.98



20
6
14
84
20
8p12-8p11.2
TRUE
37.7-41.78
37.72-38.02
1.71
37.7-38.45
0.78




0
5
65
20
8p12-8p11.2
FALSE
37.7-41.78
37.72-35.02
1.71
38.68-39.52
0.64



21
0
4
98
21
8q12.1-8q12
TRUE
59.09-63.56
50.23-82.26
0.58
69.23-80.82
1.6



22
0
8
127
22
8q12.3-8q13
TRUE
83.7-67.06
85.44-88.81
0.87
83.7-87.00
3.37



23
41
115
13
23
8q21.3-8q24
TRUE
90.7-145.63
133.72-134.16
1.78
118.87-145.83
25.88



24
9
18
14
24
9p21.3-9p13
TRUE
23.88-37.87
35.85-38.56
0.88
35.6-38.58
0.98



25
3
16
15
25
10q25.2-10q
TRUE
112.4-115.62
112.4-112.77
0.88
112.4-115.62
3.22



26
0
7
74
26
10q28.13-10
TRUE
124.82-129.83
125.14-125.14
0.88
124.92-126.41
1.49



27
0
13
18
27
11p12-11q13
TRUE
40.18-77.26
77-77.15
0.9
60.11-31.14
1.03



28
0
9
87
28
11q14.1-11q
TRUE
78.16-88.74
82.78-85.89
0.9
83.39-66.21
2.82



29
1
4
99
29
12p13.33-12
TRUE
0.18-11.45
0.18-2.79
1.77
0.18-0.74
0.58




14
33
100
29
12p13.33-12
FALSE
0.18-11.45
0.18-2.79
1.77
8.03-6.88
1.82




1
2
101
29
12p13.33-12
FALSE
0.18-11.45
0.18-2.79
1.77
7.05-7.18
0.13



30
8
14
68
30
12p13.2-12p
TRUE
11.95-13.06
12.46-12.77
0.6
11.95-13.06
1.11



31
1
3
40
31
12p12.3-12q
TRUE
18.8-53.06
20.7-22.34
2.52
21.82-22.39
0.57




13
28
41
31
12p12.3-12q
FALSE
16.6-53.06
20.7-22.64
2.52
24.87-31.14
6.27



32
0
7
42
32
12q14.3-12q
TRUE
64.61-65.08
65.95-66.06
0.93
64.61-66.08
1.46



33
0
1
114
33
12q15-12q18
TRUE
66.27-68.87
68.44-88.77
1.38
88.27-68.88
0.59



34
9
28
88
34
12q24.11-12
TRUE
108.08-110.72
108.85-110.68
0.98
108.06-110.72
2.68



35
9
29
89
35
12q24.31-12
TRUE
120.76-133.4
123.83-125.12
1.6
122.12-131.01
8.89



36
0
3
81
36
13q12.11-13
TRUE
18.88-43.95
19.21-24.22
0.89
18.68-19.21
0.83




3
19
82
36
13q12.11-13
FALSE
18.63-43.95
19.21-24.22
0.89
20.65-24.85
4.19



37
4
12
90
37
13q14.2-13q
TRUE
45.55-47.61
47.45-47.77
0.68
48.64-47.81
2.18



38
1
3
67
38
13q34-13q34
TRUE
112.84-113.06
112.85-113.01
0.62
112.84-113.08
0.23



39
7
14
18
39
14q11.2-14q
TRUE
18.82-74.47
73.56-74.47
1.08
28.12-33.17
5.05




17
28
19
39
14q11.2-14q
FALSE
18.82-74.47
73.56-74.47
1.08
48.08-31.17
3.09




18
68
20
39
14q11.2-14q
FALSE
18.82-74.47
73.56-74.47
1.08
52.41-63.83
11.12



40
9
29
75
40
14q24.3-14q
TRUE
75.23-88.88
75.26-88.98
0.93
75.23-87.87
12.63



41
4
7
43
41
14q32.12-14
TRUE
91.16-104.37
91.39-104.37
0.63
103.33-103.61
0.28



42
2
4
21
42
16p13.3-16p
TRUE
0.03-4.95
3.37-3.49
1.23
1.81-1.85
0.15




3
9
22
42
16p13.3-16p
FALSE
0.03-4.95
3.37-3.49
1.23
3.27-3.84
0.25




2
6
23
42
16p13.3-16p
FALSE
0.03-4.95
3.37-3.49
1.23
3.9-4.52
0.62



43
2
5
24
43
16p13.3-16p
TRUE
5.14-15.78
7.63-15.08
0.93
14.73-15.82
0.85



44
4
9
148
44
16p12.2-18p
TRUE
23.7-27.3
24.03-25.16
1.24
23.7-27.3
3.61



45
0
1
142
45
17q11.2-17q
TRUE
29.14-30.78
30.32-30.88
0.74
30.5-30.78
0.28



46
70
215
27
46
17q12-17q23
TRUE
37.48-56.39
43.63-43.63
2.87
38.96-66.28
18.31



47
3
5
82
47
17q23.2-17q
TRUE
59.83-79.04
62.33-62.47
3.61
74.02-74.22
0.2



48
6
11
140
48
18p11.21-18
TRUE
12.02-28.55
19.67-21.37
2.12
19.12-21.37
2.25



49
18
35
45
49
19p13.11-19
TRUE
23.32-50
45.03-45.39
3.84
41.41-44.6
3.2



50
65
282
46
50
19q13.32-19
TRUE
50.06-63.76
50.88-60.05
1.59
50.05-52.89
12.83



51
1
10
47
51
20p13-20q13
TRUE
0.33-63.41
58.54-63.41
1.34
25.68-31.48
5.8




4
12
48
51
20p13-20q13
FALSE
0.33-63.41
58.54-63.41
1.34
60.87-65.58
4.71




19
87
49
51
20p13-20q13
FALSE
0.33-63.41
58.54-63.41
1.34
55.24-63.41
5.17



52
2
6
83
52
22q11.1-22q
TRUE
14.65-28.58
22.7-22.7
1.04
22.64-22.83
0.2



53
2
5
115
53
22q12.2-22q
TRUE
28.76-29.85
28.98-29.83
0.93
29.35-29.85
0.51



54
1
2
236
54
1p36.21-1p3
TRUE
15.02-28.96
21.48-21.48
−0.94
21.08-21.56
0.48




18
64
237
54
1p36.21-1p3
FALSE
15.02-28.95
21.45-21.48
−0.94
22.82-26.95
4.13



55
7
17
182
55
1p33.3-1p34
TRUE
28.37-39.18
32.79-33.04
−0.87
28.37-31.18
2.51




4
10
183
56
1p33.3-1p34
FALSE
28.37-39.18
32.79-33.04
−0.87
32.87-34.88
2.01



56
4
14
283
58
1p34.2-1p34
TRUE
40.39-42.59
42.09-42.11
−0.75
40.39-42.89
2.21



57
2
7
252
57
1p32.2-1p31
TRUE
56.74-60.95
56.77-59.75
−0.59
56.74-60.95
4.21



58
0
0
238
58
1p21.2-1p21
TRUE
100.53-103.3
100.25-103.25
−1.69
103.25-103.3
0.05



59
6
14
161
59
2p25.3-2p24
TRUE
0.21-16.09
10.92-11.3
−0.65
0.21-7.04
6.83




1
1
162
59
2p25.3-2p24
FALSE
0.21-16.09
10.92-11.3
−0.85
11.3-11.32
0.02



60
0
1
230
60
2p12-2p11.2
TRUE
75.23-89
79.31-65.51
−1.03
77.7-79.27
1.57




0
10
231
60
2p12-2p11.2
FALSE
75.23-59
79.31-65.51
−1.03
88.97-59
2.03



61
2
5
163
61
3p26.3-3p24
TRUE
1.33-18.8
13.85-14.52
−1
11.29-12.18
0.89




3
8
164
61
3p26.3-3p24
FALSE
1.33-18.5
13.65-14.52
−1
13.87-14.52
0.85



62
12
28
233
62
3p21.32-3p1
TRUE
44.51-68.3
53.69-57.41
−0.57
53.08-61.58
5.51



63
1
2
185
63
4p16.3-4p16
TRUE
4.27-4.86
4.41-4.43
−0.82
4.27-4.
0.59



64
5
15
239
64
4p15.32-4p1
TRUE
17.28-25.18
24.69-25.12
−0.94
17.28-28.18
8.91



65
4
28
265
65
4q13.2-4q13
TRUE
69.37-72.84
69.37-71.59
−0.04
69.27-72.84
3.47



66
0
8
218
66
4q22.1-4q22
TRUE
88.79-89.29
69.13-59.13
−0.75
88.79-69.29
0.5



67
0
0
219
67
4q24-4q24
TRUE
104.07-103.96
104.07-104.07
−0.62
104.07-103.95
−0.13



68
2
9
165
68
4q27-4q28.1
TRUE
123.22-129.09
123.77-124.24
−0.75
123.22-129.09
5.87



69
2
6
167
69
4q31.22-4q1
TRUE
148.03-158.75
149.48-153.1
−0.93
151.71-154.37
2.85




0
14
168
69
4q31.22-4q3
FALSE
148.03-158.78
149.48-153.1
−0.93
155.09-158.58
3.47



70
8
40
210
70
4q34.1-4q35
TRUE
174.84-168.09
175.03-187.79
−1.25
174.84-188.09
13.28



71
1
3
281
71
5q23.2-5q23
TRUE
127.55-130.53
127.85-128.48
−0.82
127.55-130.53
2.98



72
1
7
184
72
6p25.3-5p12
TRUE
0.4-52.14
0.4-2.83
−1.17
0.4-2.83
2.42




7
56
185
72
6p25.3-6p12
FALSE
0.4-52.14
0.4-2.83
−1.17
25.1-27.86
1.75



73
1
5
279
73
6p12.1-6p12
TRUE
56.4-57.04
55.4-55.4
−0.64
58.4-57.04
0.54



74
21
56
187
74
6q21-6q22.3
TRUE
106.63-119.43
107.02-118.57
−1.11
105.83-119.43
12.8



75
5
38
241
75
6q23.3-6q24
TRUE
135.17-146.81
137.40-138.13
−0.91
135.17-146.81
11.84



76
8
18
188
76
6q25.3-6q27
TRUE
160.39-165.58
180.51-152.94
−0.73
180.39-166.08
6.27



77
2
8
282
77
6q27-6q27
TRUE
158.07-170.54
168.63-170.54
−0.72
158.07-170.54
2.47



78
1
13
295
78
7p21.2-7p21
TRUE
14.44-16.87
17.08-18.88
−0.9
14.44-18.87
4.43



79
0
12
265
79
7q34-7q36.1
TRUE
142.47-147.82
143.2-143.36
−0.8
142.7-147.82
5.12



80
4
12
190
80
8p23.3-8p12
TRUE
2.08-37.7
2.08-2.1
−1.88
18.07-21.75
3.68




18
28
191
80
8p23.3-8p12
FALSE
2.08-17.7
2.08-2.1
−1.88
28.45-37.7
9.25



81
3
17
212
81
8p12-8p11.2
TRUE
37.7-42.45
38.45-42.33
−1.19
37.7-39.05
1.35



82
3
8
170
82
9p24.3-9p21
TRUE
0.47-27.18
20.77-21.31
−2.53
0.47-3.39
2.92




18
65
171
82
9p24.3-9q21.1
FALSE
0.47-27.18
20.77-21.31
−2.63
6.3-23.88
18.39



83
0
0
272
83
9q13-9q21.1
TRUE
65.19-55.51
65.26-65.51
−1.27
65.19-55.51
1.32



84
2
13
297
84
9q22.33-9q3
TRUE
97.63-101.94
97.84-101.81
−0.81
97.63-101.94
4.31



85
8
13
250
85
9q31.2-9q31
TRUE
105.59-109.28
108.59-107.89
−1
106.59-108.83
2.04



86
5
10
285
86
9q34.11-9q3
TRUE
127.73-128.55
127.75-127.81
−0.91
127.73-128.53
0.6



87
3
7
258
87
10p15.3-10p
TRUE
0.29-1.17
0.29-1.08
−0.61
0.29-1.17
0.88



88
4
24
173
88
10p12.33-10p
TRUE

7.91-25.66

18.09-18.32
−3.3
17.91-26.88
8.75



89
9
20
298
89
10p12.1-10p
TRUE
27.19-33.36
27.83-32.71
−0.62
27.19-33.88
6.17



90
0
1
174
90
10q22.1-10q
TRUE
73.46-73.72
73.46-73.47
−3.23
73.48-73.72
0.28



91
15
34
221
91
10q22.3-10q



92
4
10
175
92
11p15.4-11p
TRUE
10.84-13
12.02-12.02
−3.72
10.54-13
2.16



93
4
27
195
93
11p15.2-11p
TRUE
14.58-15.47
17.38-18.43
−0.92
14.58-18.47
3.59



94
7
23
198
94
11p15.1-11p
TRUE
20.11-32.17
20.42-31.85
−1.2
20.11-32.17
12.06



95
0
11
204
95
11q14.1-11q
TRUE
78.15-85.59
82.76-83.39
−0.87
78.15-85.69
7.44



96
0
13
222
96
11q14.2-11q
TRUE

.59-69.24

85.21-89.24
−0.9
85.59-59.24
3.35



97
8
75
223
97
11q21-11q23
TRUE
95.74-114.13
95.74-114.13
−0.75
95.74-114.13
18.30



98
1
4
178
98
11q23.3-11q
TRUE
110.73-134.28
117.1-134.27
−0.7
120.33-121.54
1.21



99
18
55
290
99
12q12-12q13
TRUE
40.08-49.24
41.04-49.21
−0.71
40.06-49.24
9.16



100 
19
70
291
100
12q13.12-12
TRUE
49.55-58.82
50.01-53.06
−0.7
49.55-63.08
3.41




4
80
292
100
12q13.12-12
FALSE
49.55-55.82
50.01-53.08
−0.7
53.17-55.82
2.85



101 
13
34
293
101
12q14.1-12q
TRUE
57.92-58.77
62.76-68.27
−0.5
62.76-68.77
0.01



102 
9
27
197
102
12q21.2-12q
TRUE
77.19-133.4
81.03-85.63
−1.19
77.19-91.44
14.25



103 
1
2
294
103
14q24.2-14q
TRUE
71.88-72.03
71.72-71.73
−1.1
71.65-72.03
0.35



104 
7
15
242
104
16p13.3-16p
TRUE
0.03-23.97
0.70-23.7
−0.67
2.24-2. 2
0.58



105 
8
24
192
105
16q12.1-16q
TRUE
48.37-55.26

.06-53.88

−1.1
48.37-53.88
5.52



106 
0
14
178
106
17p13.3-17q
TRUE
0.02-25.81
8.16-14.05
−0.99
10.36-12.8
2.44



107 
1
5
225
107
17q11.2-17q
TRUE
25.68-30.32

. -28.65

−0.83
27.8-28.97
1.17



108 
0
2
228
108
17p24.2-17q
TRUE

.73-76.58

66.96-71.88
−0.92
71.63-71.65
0.23



110 
5
16
208
110
18q11.2-18q
TRUE
18.61-48.28
34.95-40.58
−1.19
34.18-43.14
8.99



111 
8
23
213
111
18q22.1-18q
TRUE
60.4-77.53
74.48-78.84
−1.83

.4-77.63

17.23



112 
2
6
196
112
19p13.3-19p
TRUE
4.62-9.11

. - .

−0.85
6.61- .88
0.24



113 
3
4
158
113
19p13.2-19p
TRUE
12.83-13.18
12.84-13.11
−0.88
12.9-13.07
0.17



114 
12
21
159
114
19q13.2-19q
TRUE
44.87-63.76
59.99- .04
−1.31
50.65-51.57
0.92




2
12
150
114
19q13.2-19q
FALSE
44.57-63.76
59.99-60.04
−1.31
59.85-60.18
0.33



115 
27
75
194
115
20p13-20q11
TRUE
0.33-30.89
8.81-25.63
−1.39
6.91-25.68
19.77



116 
0
3
200
116
21p11.2-21q
TRUE
9.96-13.43
9.95-10.08
−1.24
9.95-13.43
3.47



117 
0
3
208
117
21q22.11-21
TRUE
31.95-32.5
32.15-32.5
−1.16
31.95-32.8
0.53



118 
0
4
214
118
21q22.11-2
TRUE
34.19-35.52
35.03-35.42
−1.09
34.81-35.42
0.61



119 
2
5
215
119
21q22.15-21
TRUE
37.01-37.52
37.01-37.02
−1.58
37.01-37.52
0.8



120 
2
2
243
120
21q22.2-21q
TRUE
39.75-45.94
41.7-41.76
−4.09
45.08-45.17
0.09




1
6
244
120
21q22.2-21q
FALSE
39.75-48.94
41.7-41.75
−4.09
48.77-46.94
0.17



121 
65
243
179
121
22q11.1-22q
TRUE
14.49-39.45
22.7-22.7
−1.32
20.64-39.45
18.83








indicates data missing or illegible when filed














TABLE 4





Markers of the invention which reside in MCRs of deletion


and display decreased expression.























Pos
Gene
Minimum






Chromosome
(Mb)
Weight
p value
Gene Description
Gene Symbol
GI
UGID#





1
21.3
−0.84
0.005
alkaline phosphatase, liver/bone/kidney
ALPL
gi: 28737
Hs.250769


1
22.8
−1.12
<.005
KIAA0601 protein
KIAA0601
gi: 3043725
Hs.348515


1
23.3
−0.38
0.037
E2F transcription factor 2
E2F2
gi: 4758225
Hs.231444


1
23.5
−0.73
<.005
lysophospholipase II
LYPLA2
gi: 9966763
Hs.413781


1
23.5
−0.39
0.049
galactose-4-epimerase, UDP-
GALE
gi: 9945333
Hs.76057


1
23.6
−0.56
0.012
3-hydroxymethyl-3-methylglutaryl-
HMGCL
gi: 4504426
Hs.444925






Coenzyme A lyase






(hydroxymethylglutaricaciduria)


1
23.6
−0.80
<.005
lysophospholipase II
LYPLA2
gi: 4376011
Hs.413781


1
23.6
−0.42
0.03
fucosidase, alpha-L-t, tissue
FUCA1
gi: 4503802
Hs.576


1
24.3
−0.48
<.005
serine/arginine repetitive matrix 1
SRRM1
gi: 5032118
Hs.18192


1
24.5
−0.64
0.021
chloride intracellular channel 4
CL1C4
gi: 4588523
Hs.25035


1
24.9
−0.61
0.005
hypothetical protein dJ465N24.2.1
DJ465N24.2.1
gi: 12005626
Hs.259412


1
25.8
−0.87
<.005
stathmin I/oncoprotein 18
STMN1
gi: 13518023
Hs.209983


1
26.1
−0.45
0.037
zinc finger protein
ZT86
gi: 7705661
Hs.102419


1
26.4
−0.44
0.021
high-mobility group nucleosomal binding
HMGN2
gi: 13277559
Hs.181163






domain 2





1
26.4
−0.86
<.005
ribosomal protein S6 kinase, 90 kDa,
RPS6KA1
gi: 4506732
Hs.149957






polypeptide 1


1
26.7
−0.61
0.015
hypothetical protein FLJ20477
FLJ20477
gi: 8923441
Hs.259605


1
26.7
−0.84
0.007
hypothetical protein FLJ20477
FLJ20477
gi: 1799134
Hs.259605


1
26.8
−2.26
0.032
hypothetical protein FLJ12455
FLJ12455
gi: 11545792
Hs.10903


1
28.5
−0.75
<.005
tRNA selenocysteine associated protein
SECP43
gi: 8923459
Hs.266935


1
28.6
−0.87
<.005
TAF12 RNA polymerase II, TATA box
TAF12
gi: 1345403
Hs.421646






binding protein (TBP)-associated factor,






20 kDa


1
28.7
−0.63
0.016
glucocorticoid modulatory element
GMEB1
gi: 13435376
Hs.4069






binding protein 1





1
28.7
−0.78
0.005
high-glucose-regulated protein 8
HGRG8
gi: 7705410
Hs.20993


1
29.1
−0.47
0.032
splicing factor, arginine/serine-rich 4
SFRS4
gi: 5032088
Hs.76122


1
29.2
−0.57
0.008
nuclear receptor binding factor 1
CGI-63
gi: 7705776
Hs.183646


1
31.1
−0.49
0.037
hypothetical protein FLJ12650
FLJ12650
gi: 13375663
Hs.436090


1
32.7
−0.67
0.006
KIAA1522 protein
KIAA1522
gi: 6588393
Hs.322735


1
32.9
−0.63
0.017
hypothetical protein FLJ90005
FLJ90005
gi: 6729581
Hs.511807


1
32.9
−0.27
<.005

Homo sapiens transcribed sequence with


gi: 6664283
Hs.416117






moderate similarity to protein






ref: NP_060265.1 (H. sapiens)






hypothetical protein FLJ20378 [Homo







sapiens]



1
33.3
−0.96
<.005
polyhomeotic-like 2 (Drosophila)
PHC2
gi: 4758241
Hs.165263


3
14.1
−0.86
<.005
hypothetical protein MGC3222
MGC3222
gi: 1384812
Hs.130330


3
14.1
−1.30
<.005
xeroderma pigmentosum,
XPC
gi: 475156
Hs.320






complementation group C


3
14.2
−0.47
0.033
LSM3 homolog, U6 small nuclear RNA
LSM3
gi: 7657314
Hs.111632






associated (S. cerevisiae)


4
153.2
−0.42
0.023
PET112-like (yeast)
PET112L
gi: 4758893
Hs.119316


4
153.8
−0.38
0.043
F-box and WD-40 domain protein 7
FBXW7
gi: 8922851
Hs.312503






(archipslago homolog, Drosophila)





4
174.8
−0.42
0.037
UDP-N-acetyl-alpha-D-
GALNT7
gi: 8393408
Hs.156856






galactosamine:polypeptide N-






acetylgalactosaminyltransferase 7






(GalNAc-T7)


4
175
−0.42
0.027

Homo sapiens clone FLB9413 PRO2532


gi: 11493455
Hs.383372






mRNA, complete cds


4
175.1
−0.40
0.021
heart and neural crest derivatives
HAND2
gi: 12545383
Hs.388245






expressed 2


4
184.5
−0.73
<.005
dCMP deaminase
DCTD
gi: 4740472
Hs.76894


4
185
−0.70
0.006
collaborates/cooperates with ARF
CARF
gi: 8923039
Hs.32922






(alternate reading frame) protein


4
186
−0.74
<.005
interferon regulatory factor 2
IRF2
gi: 4755144
Hs.83795


4
186.9
−0.38
0.05
hypothetical protein DKFZp761O0113
DKFZp761O0113
gi: 8922176
Hs.42768


4
187
−0.40
0.028
hypothetical protein FLJ11200
FLJ11200
gi: 8922937
Hs.368022


5
128.5
−2.23
0.042
CGI-111 protein
CGI-111
gi: 7705613
Hs.11085


6
106.7
−0.63
<.005
APG5 autophagy 5-like (S. cerevisiae)
APG5L
gi: 7023451
Hs.11171


6
107.1
−0.45
0.015
glutaminyl-tRNA synthase (glutamine-
QRSL1
gi: 12052881
Hs.406917






hydrolyzing)-like 1


6
107.6
−0.61
<.005
chromosome 6 open reading frame 210
C6orf210
gi: 9966852
Hs.268733


6
108.2
−0.95
<.005
SEC63-like (S. cerevisiae)
SEC63
gi: 5393231
Hs.330767


6
108.5
−0.48
0.01
sorting nexin 3
SNX3
gi: 6010168
Hs.12102


6
108.6
−0.53
0.007
sorting nexin 3
SNX3
gi: 11765691
Hs.12102


6
109
−0.24
0.014


gi: 3821018



6
109.7
−0.75
<.005
CD164 antigen, sialomucin
CD164
gi: 11943350
Hs.43910


6
109.8
−0.77
<.005
sphingomyelin phosphodiesterase 2,
SMPD2
gi: 5101461
Hs.55235






neutral membrane (neutral






sphingomyelinase)


6
109.8
−0.76
<.005
NEDD9 interacting protein with calponin
NICAL
gi: 12232438
Hs.33476






homology and LIM domains


6
109.8
−0.54
0.008
zinc finger protein 450
ZNF450
gi: 7662127
Hs.409876


6
110.1
−0.36
0.04
KIAA0274
KIAA0274
gi: 7662033
Hs.419998


6
110.5
−0.52
<.005
WAS protein family, member 1
WASF1
gi: 4507912
Hs.75850


6
110.5
−0.94
<.005
cell division cycle 40 homolog (yeast)
CDC40
gi: 7706656
Hs.116674


6
110.9
−1.03
<.005
cyclin-dependent kinase (CDC2-like) 11
CDK11
gi: 5100783
Hs.129836


6
111
−1.36
<.005
cyclin-dependent kinase (CDC2-like) 11
CDK11
gi: 5689392
Hs.129836


6
111.2
−0.41
<.005
adenosylmethionine decarboxylase 1
AMD1
gi: 178517
Hs.159118


6
111.7
−0.76
<.005
REV3-like, catalytic subunit of DNA
REV3L
gi: 4506482
Hs.232021






polymerase zeta (yeast)


6
114.3
−0.72
<.005
histone deacetylase 2
HDAC2
gi: 4557640
Hs.3352


6
116.6
−0.84
<.005
TSPY-like
TSPYL
gi: 12052783
Hs.458358


6
119.2
−0.10
<.005
ASF1 anti-silencing function 1 homolog
ASF1A
gi: 7661591
Hs.292316






A (S. cerevisiae)


6
135.3
−0.65
0.019
HBS1-like (S. cerevisiae)
HBS1L
gi: 6703779
Hs.221040


6
135.6
−0.57
0.016
Abelson helper integration stie
AHI1
gi: 8923074
Hs.273294


6
136.9
−0.29
0.024
mitogen-activated protein kinase kinase
MAP3K5
gi: 1805499
Hs.151988






kinase 5


6
137.3
−0.58
<.005
interleukin 20 receptor, alpha
IL20RA
gi: 7657690
Hs.288240


6
138.7
−0.52
0.012
heme binding protein 2
HEBP2
gi: 7657602
Hs.439081


6
139
−0.74
<.005
chromosome 6 open reading frame 80
C6orf80
gi: 12653928
Hs.44468


6
139.4
−0.45
0.024
headcase homolog (Drosophila)
HECA
gi: 7706434
Hs.6679


6
146.5
−0.76
<.005
glutamate receptor, metabotropic 1
GRM1
gi: 6006005
Hs.32945


6
169.8
−0.25
0.047
PHD finger protein 10
PHF10
gi: 11085906
Hs.435933


6
169.8
−0.61
0.011
PHD finger protein 10
PHF10
gi: 8922799
Hs.435933


8
18
−0.44
0.026
N-acetyltransferase 1 (arylamine N-
NAT1
gi: 4505334
Hs.458430






acetyltransferase)


8
18.3
−0.65
<.005
hypothetical protein DKFZp761K1423
DKFZp761K1423
gi: 8922171
Hs.236438


8
18.5
−0.68
<.005
ADP-ribosylation factor guanine
EFA6R
gi: 6085952
Hs.408177






nucleotide factor 6


8
20
−0.75
<.005
ATPase, H+ transporting, lysosomal
ATP6V1B2
gi: 4502310
Hs.295917






56/58 kDA, V1 subunit B, isoform 2


8
28.6
−0.40
0.042
exostoses (multiple)-like 3
EXTL3
gi: 2897904
Hs.9018


8
28.6
−0.49
0.02
exostoses (multiple)-like 3:exostoses
EXTL3
gi: 13623512
Hs.9018;






(multiple)-like 3


Hs.9018


8
28.7
−0.80
<.005
hypothetical protein FLJ10871
FLJ10871
gi: 8922725
Hs.15562


8
29.9
−0.99
<.005
hypothetical protein MGC8721
MGC8721
gi: 7706384
Hs.279921


8
30
−0.96
<.005
leptin receptor overlapping transcript-like 1
LEPROTL1
gi: 7662509
Hs.146585


8
30
−0.80
<.005
dynactin 6
DCTN6
gi: 5730115
Hs.158427


8
30.3
−0.22
0.034
RNA binding protein with multiple
RBPMS
gi: 5803140
Hs.195825






splicing


8
30.5
−0.56
<.005
general transcription factor IIE,
GTF2E2
gi: 4504194
Hs.77100






polypeptide 2, beta 34 kDA


8
30.6
−0.84
<.005
glutathione reductase
GSR
gi: 10835188
Hs.414334


8
30.6
−0.42
0.022
reproduction 8
D8S2298E
gi: 1913786
Hs.153678


8
30.7
−0.46
<.005
protein phosphatase 2 (formerly 2A),
PPP2CB
gi: 4758951
Hs.80350






catalytic subunit, beta isoform


8
32.6
−0.08
0.039
neuregulin 1
NRG1
gi: 7669513
Hs.172816


8
33.4
−1.04
<.005
RNA binding protein; RNA binding
LOC84549
gi: 13625185
Hs.77135;






protein


Hs.77135


8
33.4
−0.53
<.005
hypothetical protein FLJ23263
FLJ23263
gi: 13376690
Hs.288716


8
37.6
−0.75
<.005
chromosome 8 open reading frame 2;
C8orf2
gi: 10241715
Hs.125849;






chromosome 8 open reading frame 2


Hs.125849


8
37.6
−1.00
<.005
proline synthetase co-transcribed
PROSC
gi: 6005841
Hs.301959






homolog (bacterial)


8
131.3
−0.17
0.021

Homo sapiens cDNA: FLJ23601 fis,


gi: 10440343
Hs.306918






clone LNG15501


9
2.1
−0.36
0.026
SWI/SNF related, matrix associated, actin
SMARCA2
gi: 6133361
Hs.396404






dependent regulator of chromatin,









subfamily a, member 2


9
2.6
−0.47
0.013
very low density lipoprotein receptor
VLDLR
gi: 437386
Hs.370422


9
2.8
−1.06
<.005
minor histocompatibility antigen HA-8
XTP5
gi: 7661865
Hs.443866


9
5.3
−0.77
<.005
chromosome 9 open reading frame 46
C9orf46
gi: 8923931
Hs.416649


9
5.5
−0.51
0.007
programmed cell death 1 ligand 2
PDCD1LG2
gi: 13376849
Hs.61929


9
6
−1.15
<.005
RAN binding protein 6
RANBP6
gi: 3538999
Hs.167496


9
14
−0.20
<.005
nuclear factor I/B
NFIB
gi: 13410807
Hs.302690


9
14.2
−0.50
<.005
nuclear factor I/B
NFIB
gi: 4988418
Hs.302690


9
15.4
−0.56
<.005
small nuclear RNA activating complex,
SNAPC3
gi: 4507104
Hs.380092






polypeptide 3, 50 kDa


9
15.4
−0.65
<.005
PC4 and SFRS1 interacting protein 2
PSIP2
gi: 3283351
Hs.351305


9
19
−0.94
<.005
Ras-related GTP binding A
RRAGA
gi: 5729998
Hs.432330


9
19
−0.43
0.026
hypothetical protein FLJ20060
FLJ20060
gi: 8923062
Hs.54617


9
19.3
−0.91
<.005
ribosomal protein S6
RPS6
gi: 4506730
Hs.408073


9
20.8
−1.14
<.005
KIAA1797
KIAA1797
gi: 8923357
Hs.257696


9
21.3
−1.03
<.005
KIAA1354 protein
KIAA1354
gi: 2185814
Hs.147717


9
21.9
0.00
0.015
cyclin-dependent kinase inhibitor 2A
CDKN2A
gi: 4502748
Hs.421349






(melanoma, p16, inhibits CDK4)





9
22
−0.15
<.005
methylthioadenosine phosphorylase
MTAP
gi: 4378719
Hs.459541


12
42.4
−1.31
<.005
PTK9 protein tyrosine kinase 9; PTK9
PTK9
gi: 4506274
Hs.189075;






protein tyrosine kinase 9


Hs.189075


12
43.8
−1.03
0.015
putative glycolipid transfer protein
LOC51054
gi: 7705683
Hs.334649


12
44.6
−0.94
0.006
splicing factor, arginine/serine-rich 2,
SFRS2IP
gi: 4759171
Hs.210367






interacting protein


12
46.4
−1.06
0.047
hypothetical protein FLJ20489
FLJ20489
gi: 8923451
Hs.438867


12
46.5
−2.40
0.043
vitamin D (1,25-dihydroxyvitamin D3)
VDR
gi: 2824068
Hs.2062






receptor


12
47.3
−0.54
0.036
hypothetical protein FLJ20436
FLJ20436
gi: 10434303
Hs.268189


12
47.5
−0.89
0.007
calcium channel, voltage-dependent, beta
CACNB3
gi: 463890
Hs.250712






3 subunit


12
47.5
−0.82
0.007
calcium channel, voltage-dependent, beta
CACNB3
gi: 463890
Hs.250712






3 subunit


12
47.5
−0.58
0.028
DEAD (Asp-Glu-Ala-Asp) box
DDX23
gi: 2655201
Hs.130098






polypeptide 23


12
47.6
−0.72
0.023
FK506 binding protein 11, 19 kDa
FKBP11
gi: 7706130
Hs.438695


12
47.6
−0.61
0.019
ADP-ribosylation factor 3
ARF3
gi: 4502202
Hs.119177


12
47.6
−1.26
<.005
ADP-ribosylation factor 3; ADP-
ARF3
gi: 178980
Hs.119177;






ribosylation factor 3


Hs.119177


12
47.6
−0.48
0.045
protein kinase, AMP-activated, gamma 1
PRKAG1
gi: 4506060
Hs.3136






non-catalytic subunit


12
47.7
−0.58
0.028
lipocalin-interacting membrane receptor
LIMR
gi: 8922462
Hs.272838


12
48.1
−1.12
<.005
spermatogenesis associated, serine-rich 2
SPATS2
gi: 12751480
Hs.152982


12
48.2
−1.25
<.005
microspherule protein 1
MCRS1
gi: 5453693
Hs.25313


12
48.4
−1.09
<.005
testis enhanced gene transcript (BAX
TEGT
gi: 2645728
Hs.35052






inhibitor 1)


12
48.6
−0.55
0.03
Rac GTPase activating protein 1
RACGAP1
gi: 11015369
Hs.23900


12
49.6
−1.33
0.034
solute carrier family 11 (proton-coupled
SLC11A2
gi: 11015990
Hs.57435






divalent metal ion transporters), member 2


12
49.8
−2.85
0.034
transcription factor CP2
TFCP2
gi: 5032174
Hs.154970


12
49.9
−1.32
<.005
DAZ associated protein 2
DAZAP2
gi: 7661885
Hs.369761


12
49.9
−0.66
0.032
hypothetical protein from clone 643
LOC57228
gi: 13097236
Hs.206501


12
50.6
−0.44
0.043
activin A receptor, type IB
ACVR1B
gi: 12652986
Hs.371974


12
50.6
−0.58
0.035
activin A receptor, type IB
ACVR1B
gi: 5912233
Hs.371974


12
50.7
−0.57
0.035

Homo sapiens cDNA clone


gi: 7152120
Hs.444433






IMAGE: 5590288, partial cds


12
51.7
−1.36
0.012
eukaryotic translation initiation factor 4B
EIF4B
gi: 4503532
Hs.93379


12
51.8
−1.11
0.009
retinoic acid receptor, gamma
RARG
gi: 307424
Hs.1497


12
51.9
−0.78
0.04
hypothetical protein MGC11308
MGC11308
gi: 11975558
Hs.19210


12
51.9
−1.36
<.005
prefoldin 5
PFDN5
gi: 4505742
Hs.288856


12
51.9
−0.76
0.044
chromosome 12 open reading frame 10
C12orf10
gi: 11056017
Hs.400801


12
52.6
−0.56
0.042
homeo box C11
HOXC11
gi: 7657165
Hs.127562


12
52.7
−0.73
0.01
homeo box C6
HOXC6
gi: 6709275
Hs.820


12
52.8
−1.85
<.005
single-strand selective monofunctional
SMUG1
gi: 7657596
Hs.5212






uracil DNA glycosylase


12
52.9
−1.16
0.012
Homo sapiens, clone IMAGE: 5288883,

gi: 13284730
Hs.349283






mRNA


12
52.9
−1.60
0.034
heterogeneous nuclear ribonucleoprotein
HNRPA1
gi: 4504444
Hs.356721






A1





12
53
−4.54
0.034
costomer protein complex, subunit zeta 1
COPZ1
gi: 7706336
Hs.181271


12
54.3
−1.01
<.005
GCN5 general control of amino-acid
GCN5L1
gi: 4503954
Hs.94672






synthesis 5-like 1 (yeast)


12
54.4
−2.59
<.005
CD63 antigen (melanoma 1 antigen)
CD63
gi: 4502678
Hs.445570


12
54.4
−1.52
0.013
ORM1-like 2 (S. cerevisiae)
ORMDL2
gi: 7661819
Hs.13144


12
63.1
−0.90
0.005
exportin, tRNA (nuclear export receptor
XPOT
gi: 5811224
Hs.85951






for tRNAs)


12
63.1
−0.76
0.014
TANK-binding kinase 1
TBK1
gi: 7019546
Hs.432466


12
63.3
−1.23
<.005
glucosamine (N-acetyl)-6-sulfatase
GNS
gi: 10329021
Hs.334534






(Sanfilippo disease IIID)


12
63.4
−0.78
0.013
glucosamine (N-acetyl)-6-sulfatase
GNS
gi: 4504060
Hs.334534






(Sanfilippo disease IIID)


12
63.8
−1.13
0.006
integral inner nuclear membrane protein
MAN1
gi: 7706606
Hs.105234


12
64.8
−0.71
0.012
CGI-119 protein
CGI-119
gi: 7706334
Hs.126372


12
65.9
−0.73
0.005
TBP-interacting protein
TIP120A
gi: 8924259
Hs.512638


12
65.9
−0.79
<.005
TBP-interacting protein
TIP120A
gi: 4240146
Hs.512638


12
66.3
−0.70
0.006
dual-specificity tyrosine-(Y)-
DYRK2
gi: 1666065
Hs.173135






phosphorylation regulated kinase 2





12
67.3
−0.97
<.005
RAP1B, member of RAS oncogene
RAP1B
gi: 7661677
Hs.374418






family


12
67.4
−0.52
0.031
solute carrier family 35, member E2
SLC35E3
gi: 8922084
Hs.445043


12
68
−0.80
0.005
glioma-amplified sequence-41
GAS41
gi: 5729837
Hs.4029


12
68.2
−0.53
0.05
chaperonin containing TCP1, subunit 2
CCT2
gi: 5453602
Hs. 189772






(beta)


12
78.7
−0.98
<.005
protein phosphatase 1, regulatory
PPP1R12A
gi: 5436140
Hs.377908






(inhibitor) subunit 12A


12
81.2
−0.81
<.005
HSPC128 protein
HSPC128
gi: 7661789
Hs.90527


12
86.9
−0.86
<.005
hypothetical protein DKFZp434N2030
DKFZp434N2030
gi: 6708922
Hs.494204


12
87
−0.52
0.045
hypothetical protein FLJ13615
FLJ13615
gi: 12711597
Hs.288715


12
87.4
−0.47
0.03
KIT ligand
KITLG
gi: 4505174
Hs.1048


12
88.2
−0.45
0.027
dual specificity phosphatase 6
DUSP6
gi: 13111942
Hs.298654


12
88.4
−0.88
0.006
ATPase, Ca++ transporting, plasma
ATP2B1
gi: 7247996
Hs.20952






membrane 1


12
88.5
−0.73
<.005
ATPase, Ca++ transporting, plasma
ATP2B1
gi: 184269
Hs.20952






membrane 1


12
91
−0.65
0.008
B-cell translocation gene 1, anti-
BTG1
gi: 4502472
Hs.255935






proliferative


16
2.5
−0.60
0.026
ATPase, H+ transporting, lysosomal
ATP6V0C
gi: 4502312
Hs.389107






16 kDa, V0 subunit c


16
2.5
−0.62
0.019
ATPase, H+ transporting, lysosomal
ATP6V0C
gi: 189675
Hs.389107






16 kDa, V0 subunit c


16
2.5
−0.70
<.005
CGI-14 protein
CGI-14
gi: 7705595
Hs.433499


16
2.5
−0.14
0.02
3-phosphoinositide dependent protein
PDPK1
gi: 4505694
Hs.154729






kinase-1


16
2.5
−0.92
<.005
3-phosphoinositide dependent protein
PDPK1
gi: 2407612
Hs.154729






kinase-1


16
2.8
−0.15
<.005
serine/arginine repetitive matrix 2
SRRM2
gi: 4739778
Hs.433343


16
2.8
−0.61
0.01
serine/arginine repetitive matrix 2
SRRM2
gi: 4531907
Hs.433343


17
45.4
−0.10
0.011
golgi SNAP receptor complex member 2
GOSR2
gi: 12711466
Hs.432552


18
37.8
−0.70
<.005
phosphoinositide-3-kinase, class 3
PIK3C3
gi: 4505800
Hs.418150


18
41.9
−0.70
<.005
ATP synthase, H+ transporting,
ATP5A1
gi: 4573764
Hs.298280






mitochondrial F1 complex, alpha subunit,






isoform 1, cardiac muscle


18
42.6
−0.41
0.031
Msx-interacting-zinc finger
MIZ1
gi: 10720797
Hs.441069


18
42.6
−0.50
0.008
Msx-interacting-zinc finger
MIZ1
gi: 3643114
Hs.441069


18
42.9
−0.60
0.006
HSPC039 protein
HSPC039
gi: 7770186
Hs.406542


18
66.1
−0.42
0.034
suppressor of cytokine signaling 4
SOCS4
gi: 4757991
Hs.44439


18
72.8
−0.48
0.011
myelin basic protein
MBP
gi: 4505122
Hs.408543


18
75.3
−0.31
0.005
nuclear factor of activated T-cells,
NFATC1
gi: 500631
Hs.512591






cytoplasmic, calcineurin-dependent 1





38
75.5
−0.60
<.005
CTD (carboxy-terminal domain, RNA
CTDP1
gi: 4758093
Hs.4076






polymerase II; polypeptide A)









phosphatase, subunit 1


18
75.7
−0.58
<.005
hypothetical protein FLJ22378
FLJ22378
gi: 13376629
Hs.288284


18
75.8
−0.61
<.005
similar to S. pombe dim1+
DIM1
gi: 12654440
Hs.433683


18
75.8
−0.53
<.005
hypothetical protein FLJ21172
FLJ21172
gi: 13376184
Hs.444642


18
75.9
−0.28
<.005
KIAA0863 protein
KIAA0863
gi: 10434228
Hs.131915


19
51.5
−0.81
0.018
protein phosphatase 5, catalytic subunit
PPP5C
gi: 5453957
Hs.431861


19
59.3
−0.32
<.005
PRP31 pre-mRNA processing factor 31
PRPF31
gi: 7661653
Hs.312927






homolog (yeast)


19
59.9
−0.40
0.047
killer cell immunoglobulin-like receptor,
KIR2DL1
gi: 897908
Hs.512572






two domains, long cytoplasmic tail, 1


19
60.1
−0.18
0.016
natural cytotoxicity triggering receptor 1
NCR1
gi: 4758691
Hs.97084


21
45
−0.51
0.025
SMT3 suppressor of mif two 3 homolog 1
SMT3H1
gi: 5902095
Hs.85119






(yeast)


21
45.1
−0.84
<.005
pituitary tumor-transforming 1 interacting
PTTG1IP
gi: 11038670
Hs.369026






protein


21
46.9
−0.31
0.05
HMT1 hnRNP methyltransferase-like 1
HRMT1L1
gi: 4504494
Hs.154163






(S. cerevisiae)


22
21.3
−0.42
0.038
POM121 membrane glycoprotein-like 1
POM121L1
gi: 7657468
Hs.380370






(rat)


22
21.3
−0.57
0.012
immunoglobulin lambda joining 3
IGLJ3
gi: 13171335
Hs.102950


22
21.8
−0.50
0.015
RAB36, member RAS oncogene family
RAB36
gi: 6049163
Hs.369557


22
21.9
−0.64
<.005
breakpoint cluster region
BCR
gi: 11038638
Hs.446394


22
22.2
−0.65
0.009
immunoglobulin lambda-like polypeptide 1
IGLL1
gi: 13399297
Hs.348935


22
22.3
−0.52
0.013

Homo sapiens, clone IMAGE: 5728597,


gi: 292400
Hs.272302






mRNA


22
22.4
−0.45
0.03
matrix metalloproteinase 11 (stromelysin
MMP11
gi: 5177469
Hs.143751






3)


22
22.4
−1.28
<.005
SWI/SNF related, matrix associated, actin
SMARCB1
gi: 4507076
Hs.512700






dependent regulator of chromatin,






subfamily b, member 1


22
22.7
−1.44
<.005
glutathione S-transferase theta 1
GSTT1
gi: 4504184
Hs.268573


22
23.2
−0.58
0.028
small nuclear ribonucleoprotein D3
SNRPD3
gi: 4759159
Hs.356549


22
25.1
−0.48
0.014
Hermansky-Pudlak syndrome 4
HPS4
gi: 5420802
Hs.441481


22
25.1
−0.64
<.005
Hermansky-Pudlak syndrome 4
HPS4
gi: 11559920
Hs.441481


22
25.2
−0.15
<.005
tuftelin interacting protein 11
TFIP11
gi: 5262598
Hs.20225


22
25.3
−0.52
0.046
crystallin, beta A4
CRYBA4
gi: 4503058
Hs.57690


22
26.4
−0.67
0.033
meningiorna (disrupted in balanced
MN1
gi: 4505222
Hs.268515






translocation) 1


22
27.4
−0.73
0.035
CHK2 checkpoint homolog (S. pombe)
CHEK2
gi: 13278893
Hs.146329


22
28.2
−0.07
0.033
chromosome 22 open reading frame 19
C22orf19
gi: 13177658
Hs.75361


22
28.5
−0.55
0.024
ASC-1 complex subunit P100
ASC1p100
gi: 5419897
Hs.436407


22
29
−0.84
<.005
splicing factor 3a, subunit 1, 120 kDa
SF3A1
gi: 5032086
Hs.406277


22
29.1
−0.87
<.005
SEC14-like 2 (S. cerevisiae)
SEC14L2
gi: 7110714
Hs.430576


22
30
−1.48
<.005
zinc finger protein 278
ZNF278
gi: 9954374
Hs.27801


22
30.2
−0.73
<.005
KIAA0542 gene product
KIAA0542
gi: 6635200
Hs.62209


22
30.2
−1.14
<.005
KIAA0542 gene product
KIAA0542
gi: 3043607
Hs.62209


22
30.3
−0.66
<.005
phosphatidylserine decarboxylase
PISD
gi: 13489111
Hs.8128


22
30.3
−0.84
<.005
KIAA0542 gene product
KIAA0542
gi: 5596770
Hs.62209


22
30.6
−0.72
<.005
KIAA0645 gene product
KIAA0645
gi: 7662221
Hs.435022


22
30.6
−0.45
0.021
tyrosine 3-monoxygenase/tryptophan 5-
YWHAH
gi: 4507950
Hs.226755






monoxygenase activation protein, eta






polypeptide


22
31.2
−0.72
0.011

Homo sapiens, clone IMAGE: 4818531,


gi: 10030150
Hs.150167






mRNA


22
31.5
−0.50
0.027
tissue inhibitor of metalloproteinase 3
TIMP3
gi: 1519557
Hs.245188






(Sorsby fundus dystrophy,






pseudoinflammatory)


22
34
−0.37
0.047
target of myb1 (chicken)
TOM1
gi: 4885636
Hs.9482


22
34.3
−0.48
0.039
apolipoprotein L, 6
APOL6
gi: 13449280
Hs.257352


22
34.4
−0.76
<.005
RNA binding motif protein 9
RBM9
gi: 1267308
Hs.433574


22
34.8
−0.62
0.013
apolipoprotein L, 3
APOL3
gi: 7656972
Hs.241535


22
34.9
−0.66
<.005
apolipoprotein L, 2
APOL2
gi: 13325155
Hs.398037


22
34.9
−0.44
0.032
myosin, heavy polypeptide 9, non-muscle
MYH9
gi: 5448699
Hs.146550


22
35
−0.84
<.005
thioredoxin 2
TXN2
gi: 4200326
Hs.211929


22
35.1
−1.23
<.005
thioredoxin 2
TXN2
gi: 9280552
Hs.211929


22
35.1
−1.11
<.005
eukaryotic translation initiation factor 3,
EIF3S7
gi: 4503522
Hs.55682






subunit 7 zeta, 66/67 kDa


22
35.6
−0.37
0.046
thiosulfate sulfurtransferase (rhodanese)
TST
gi: 1877030
Hs.351863


22
35.6
−0.64
0.005
mercaptopyruvate sulfurtransferase
MPST
gi: 13489090
Hs.248267


22
35.6
−0.58
0.01
hypothetical protein FLJ12242
FLJ12242
gi: 13489098
Hs.94810


22
36.1
−0.53
0.033
manic fringe homolog (Drosophila)
MFNG
gi: 5175720
Hs.371768


22
36.1
−0.20
0.015
caspase recruitment domain family,
CARD10
gi: 5877877
Hs.57973






member 10


22
36.2
−1.08
<005
golgi associated, gamma adaptin ear
GGA1
gi: 9558728
Hs.405689






containing, ARF binding protein 1


22
36.2
−0.62
0.023
golgi associated, gamma adaptin ear
GGA1
gi: 5858473
Hs.405689






containing, ARF binding protein 1


22
36.2
−0.61
0.026
SH3-domain binding protein 1
SH3BP1
gi: 11545732
Hs.511954


22
36.4
−0.76
0.021
glycine C-acetyltransferase (2-amion-3-
GCAT
gi: 7657117
Hs.54609






ketobutyrate coenzyme A ligase)


22
36.5
−1.41
>.005
polymerase (RNA) II (DNA directed)
POLR2F
gi: 1309770
Hs.46405






polypeptide F


22
36.9
−1.89
0.042
casein kinase 1, epsilon
CSNK1E
gi: 6471575
Hs.355669


22
37.1
−0.88
<.005
KDEL (Lys-Asp-Glu-Leu) endoplasmic
KDELR3
gi: 8051612
Hs.250696






reticulum protein retention receptor 3





22
37.1
−0.54
0.026
DMC1 dosage suppressor of mck1
DMC1
gi: 106600
Hs.339396






homolog meiosis-specific homologens






recombination (yeast)


22
37.3
−0.48
0.028
chromosome 22 open reading frame 2
C22orf2
gi: 7656941
Hs.334911


22
37.3
−0.47
0.038
transiocase of outer mitochondrial
TOMM22
gi: 9910381
Hs.285005






membrane 22 homolog (yeast)


22
37.3
−0.75
<.005
KIAA0063 gene product
KIAA0063
gi: 7661887
Hs.3094


22
37.3
−0.41
0.036
unc-84 homolog B (C. elegans)
UNC84B
gi: 4582132
Hs.406612


22
37.3
−0.50
0.016
GTP binding protein 1
GTPBP1
gi: 1916924
Hs.283677


22
37.3
−1.04
<.005
GTP binding protein 1
GTPBP1
gi: 7661735
Hs.283677


22
37.8
−0.43
0.008
platelet-derived growth factor beta
PDGFB
gi: 11012269
Hs.1976






polypeptide (sitnian sarcoma viral (v-sis)









oncogene homolog)


22
38
−0.54
0.042
mitogen-activated protein kinase kinase
MAP3K7IP1
gi: 5174702
Hs.403927






kinase 7 interacting protein 1





22
38.1
−0.56
0.047
mannosyl (beta-1,4-)-glycoprotein beta-
MGAT3
gi: 6031184
Hs.276808






1,4-N-acetylglucosaminyltransferase


22
38.1
−0.64
0.03
hypothetical protein FLJ20232
FLJ20232
gi: 12803520
Hs.505742


22
38.1
−0.62
0.018
hypothetical protein FLJ20232
FLJ20232
gi: 1524716
Hs.505742


22
38.1
−0.48
0.041
activating transcription factor 4 (tax-
ATF4
gi: 4502264
Hs.181243






responsive enhancer element B67)





22
38.2
−0.52
0.043
mannosyl (beta-1,4-)-glycoprotein beta-
MGAT3
gi: 4914501
Hs.276808






1,4-N-acetylglucosaminyltransferase


22
39
−0.26
0.022
hypothetical protein DJ1042K10.2
DJ1042K10.2
gi: 11034850
Hs.22129























Ref Seq
SEQ ID NO.:
Ref Seq Prot
SEQ ID NO.:



Chromosome
Locus Link
Regulation
Probes
mRna ID
Nuc.
ID
A.A.







1
249
DOWN
215783_s_at
NM_000478
427
NP_000469
1178



1
23028
DOWN
212348_s_at
NM_015013
428
NP_055828
1179



1
1870
DOWN
207042_at
NM_004091
429
NP_004082
1180



1
11313
DOWN
202292_x_at
NM_007260
430
NP_009191
1181



1
2582
DOWN
202528_at
NM_000403
431
NP_000394
1182



1
3155
DOWN
202772_at
NM_000191
432
NP_000182
1183



1
11313
DOWN
215568_x_at
NM_007260
433
NP_009191
1184



1
2517
DOWN
202838_at
NM_000147
434
NP_000138
1185



1
10250
DOWN
201225_s_at
NM_005839
435
NP_005830
1186



1
25932
DOWN
201559_s_at
NM_013943
436
NP_039234
1187



1
57035
DOWN
209006_s_at
NM_020317
437
NP_064713
1188



1
3925
DOWN
200783_s_at
NM_005563
438
NP_005554
1189



1
51042
DOWN
204175_at
NM_015871
439
NP_056955
1190



1
3151
DOWN
208668_x_at
NM_005517
440
NP_005508;
1191









NP_116138



1
6195
DOWN
203379_at
NM_002953
441
NP_002944
1192



1
55650
DOWN
219238_at
NM_017837
442
NP_060307
1193



1
55650
DOWN
51146_at
NM_017837
443
NP_060307
1194



1
63906
DOWN
218895_at
NM_022078
444
NP_071361
1195



1
54952
DOWN
218977_s_at
NM_017846
446
NP_060316
1197



1
6883
DOWN
209463_s_at
NM_005644
447
NP_005635
1198



1
10691
DOWN
220938_s_at
NM_006582;
448; 449
NP_006573;
1199; 1200







NM_024482

NP_077808



1
51441
DOWN
217812_at
NM_016258
450
NP_057342
1201



1
6429
DOWN
201696_at
NM_005626
451
NP_005617
1202



1
51102
DOWN
218664_at
NM_016011
452
NP_057095
1203



1
79570
DOWN
219438_at
NM_024522
453
NP_078798
1204



1
57648
DOWN
212048_s_at
XM_036299
454
XP_036299
1205



1
127544
DOWN
213038_at
NM_153341
455
NP_699172
1206



1

DOWN
212172_at

1501 





1
1912
DOWN
200919_at
NM_004427;
456; 457
NP_004418;
1207; 1208







NM_198040

NP_932157



3
79188
DOWN
217795_s_at
NM_024334
459
NP_077310
1210



3
7508
DOWN
209375_at
NM_004628
460
NP_004619
1211



3
27258
DOWN
202209_at
NM_014463
461
NP_055278
1212



4
5188
DOWN
204300_at
NM_004564
462
NP_004555
1213



4
55294
DOWN
218751_s_at
NM_018315;
463; 464
NP_060785;
1214; 1215







NM_033632

NP_361014



4
51809
DOWN
218313_s_at
NM_017423
465
NP_059119
1216



4

DOWN
210918_at

1502 

1503



4
9464
DOWN
220480_at
NM_021973
466
NP_068808
1217



4
1635
DOWN
201571_s_at
NM_001921
467
NP_001912
1218



4
55602
DOWN
218929_at
NM_017632
468
NP_060102
1219



4
3660
DOWN
203275_at
NM_002199
469
NP_002190
1220



4
55805
DOWN
207797_s_at
NM_018409
470
NP_060879
1221



4
55325
DOWN
218449_at
NM_018359
471
NP_060829
1222



5
51015
DOWN
218170_at
NM_016048
472
NP_057132
1223



6
9474
DOWN
202511_s_at
NM_004849
473
NP_004840
1224



6
55278
DOWN
218948_at
NM_018292
474
NP_060762
1225



6
57107
DOWN
219307_at
NM_020381
475
NP_065114
1226



6
11231
DOWN
201915_at
NM_007214
476
NP_009145
1227



6
8724
DOWN
200067_x_at
NM_003795;
477; 478; 479
NP_003786;
1228; 1229; 1230







NM_152827;

NP_690040;







NM_152828

NP_690041



6
8724
DOWN
213545_x_at
NM_003795;
480; 481; 482
NP_003786;
1231; 1232; 1233







NM_152827;

NP_690040;







NM_152828

NP_690041



6

DOWN
217185_s_at







6
8763
DOWN
208654_s_at
NM_006016
483
NP_006007
1234



6
6610
DOWN
214206_at
NM_003080
484
NP_003071
1235



6
64780
DOWN
218376_s_at
NM_022765
485
NP_073602
1236



6
9841
DOWN
205340_at
XM_376525
486
XP_376525
1237



6
9896
DOWN
203656_at
NM_014845
487
NP_055660
1238



6
8936
DOWN
204165_at
NM_003931
488
NP_003922
1239



6
51362
DOWN
203377_s_at
NM_015891
489
NP_056975
1240



6
23097
DOWN
212897_at
NM_015076
490
NP_055891
1241



6
23097
DOWN
212899_at
NM_015076
491
NP_055891
1242



6
262
DOWN
201196_s_at
NM_001634
492
NP_001625
1243



6
5980
DOWN
208070_s_at
NM_002912
493
NP_002903
1244



6
3066
DOWN
201833_at
NM_001527
494
NP_001518
1245



6
7259
DOWN
221493_at
XM_371844
495
XP_371844
1246



6
25842
DOWN
203427_at
NM_014034
496
NP_054753
1247



6
10767
DOWN
209315_at
NM_006620
497
NP_006611
1248



6
54806
DOWN
220841_s_at
NM_017651
498
NP_060121
1249



6
4217
DOWN
203836_s_at
NM_005923
499
NP_005914
1250



6
53832
DOWN
219115_s_at
NM_014432
500
NP_055247
1251



6
23593
DOWN
203430_at
NM_014320
501
NP_055135
1252



6
25901
DOWN
209479_at
NM_015439
502
NP_056254
1253



6
51696
DOWN
218603_at
NM_016217
503
NP_057301
1254



6
2911
DOWN
207299_s_at
NM_000838
504
NP_000829
1255



6
55274
DOWN
221786_at
NM_018288;
505; 506
NP_060758:
1256; 1257







NM_133325

NP_579866



6
55274
DOWN
219126_at
NM_018288;
507; 508
NP_060758;
1258; 1259







NM_133325

NP_579866



8
9
DOWN
214440_at
NM_000662
509
NP_000653
1260



8
55358
DOWN
218613_at
NM_018422
510
NP_060892
1504



8
23362
DOWN
203354_s_at
NM_015310
511
NP_056125
1261



8
526
DOWM
201089_at
NM_001693
512
NP_001684
1262



8
2137
DOWN
209202_s_at
NM_001440
513
NP_001431
1263



8
2137; 2137
DOWN
211051_s_at
NM_001440
514
NP_001431
1264



8
55756
DOWN
203941_at
NM_018250
515
NP_060720
1265



8
51669
DOWN
200847_s_at
NM_016127
516
NP_057211
1266



8
23484
DOWN
202594_at
NM_015344
517
NP_056159
1267



8
10671
DOWN
203261_at
NM_006571
518
NP_006562
1268



8
11030
DOWN
207836_s_at
NM_006867
519
NP_006858
1269



8
2961
DOWN
202680_at
NM_002095
520
NP_002086
1270



8
2936
DOWN
205770_at
NM_000637
521
NP_000628
1271



8
7993
DOWN
215983_s_at
NM_005671
522
NP_005662
1272



8
5516
DOWN
201375_s_at
NM_004156
523
NP_004147
1273



8
3084
DOWN
206237_s_at
NM_004495;
524; 525; 526; 527;
NP_004486;
1274; 1275; 1276;







NM_013956;
528; 529; 530; 531;
NP_039250;
1277; 1278; 1279;







NM_013957;
532
NP_039251;
1280; 1281; 1282







NM_013958;

NP_039252;







NM_013959;

NP_039253;







NM_013960;

NP_039254;







NM_013961;

NP_039255;







NM_013962;

NP_039256;







NM_013964

NP_039258



8
84549;
DOWN
211686_s_at
NM_032509
533
NP_115898
1283




84549



8
80185
DOWN
219124_at
NM_025115
534
NP_079391
1284



8
11160;
DOWN
221543_s_at
NM_007175;
535; 536
NP_009106
1285




11160


NM_007175



8
11212
DOWN
214545_s_at
NM_007198
537
NP_009129
1286



8

DOWN
216416_at

1505 





9
6595
DOWN
212257_s_at
NM_003070;
538; 539
NP_003061;
1287; 1288







NM_139045

NP_620614



9
7436
DOWN
209822_s_at
NM_003383
540
NP_003374
1289



9
9933
DOWN
203712_at
NM_014878
541
NP_055693
1290



9
55848
DOWN
218992_at
NM_018465
542
NP_060935
1291



9
80380
DOWN
220049_s_at
NM_025239
543
NP_079515
1292



9
26953
DOWN
213019_at
NM_012416
544
XP_039701




9
4781
DOWN
213029_at
NM_005596
545
NP_005587
1293



9
4781
DOWN
209289_at
NM_005596
546
NP_005587
1294



9
6619
DOWN
204001_at
NM_003084
547
NP_003075
1295



9
11168
DOWN
209337_at
NM_021144;
548; 549
NP_066967;
1296; 1297







NM_033222

NP_150091



9
10670
DOWN
201628_s_at
NM_006570
550
NP_006561
1298



9
54801
DOWN
218602_s_at
NM_017645
551
NP_060115
1299



9
6194
DOWN
201254_x_at
NM_001010
552
NP_001001
1300



9
54914
DOWN
218503_at
NM_017794
553
NP_060264
1301



9
55958
DOWN
213233_s_at
NM_018847
554
NP_061335
1302



9
1029
DOWN
207039_at
NM_000077;
556; 557; 558;
NP_000068;
1304; 1305; 1306







NM_058195;
559
NP_478102;







NM_058196;

NP_478103;







NM_058197

NP_478104



9
4507
DOWN
211363_s_at
NM_002451
560
NP_002442
1307



12
5756; 5756
DOWN
201745_at
NM_002822;
561; 562; 563; 564
NP_002813;
1308; 1309







NM_198974;

NP_945325







NM_002822;







NM_198974



12
51054
DOWN
220157_x_at
NM_015899
565
NP_056983
1310



12
9169
DOWN
206989_s_at
NM_004719
566
NP_004710
1311



12
55652
DOWN
218417_s_at
NM_017842
567
NP_060312
1312



12
7421
DOWN
204255_s_at
NM_000376
568
NP_000367
1313



12
54934
DOWN
221821_s_at
NM_017822
569
NP_060292
1314



12
784
DOWN
209530_at
NM_000725
570
NP_000716
1315



12
784
DOWN
34726_at
NM_000725
571
NP_000716
1316



12
9416
DOWN
40465_at
NM_004818
572
NP_004809
1317



12
51303
DOWN
219117_s_at
NM_016594
573
NP_057678
1318



12
377
DOWN
200011_s_at
NM_001659
574
NP_001650
1319



12
377; 377
DOWN
211622_s_at
NM_001659
575
NP_001650
1320



12
5571
DOWN
201805_at
NM_002733
576
NP_002724
1321



12
55716
DOWN
220036_s_at
NM_018113
577
NP_060583
1322



12
65244
DOWN
218324_s_at
NM_023071
578
NP_075559
1323



12
10445
DOWN
202556_s_at
NM_006337
579
NP_006328
1324



12
7009
DOWN
200803_s_at
NM_003217
580
NP_003208
1325



12
29127
DOWN
222077_s_at
NM_013277
581
NP_037409
1326



12
4891
DOWN
203123_s_at
NM_000617
582
NP_000608
1327



12
7024
DOWN
207627_s_at
NM_005653
583
NP_005644
1328



12
9802
DOWN
200794_x_at
NM_014764
584
NP_055579
1329



12
57228
DOWN
209679_s_at
NM_020467
585
NP_065200
1330



12
91
DOWN
205209_at
NM_004302;
587; 588; 589
NP_004293;
1332; 1333; 1334







NM_020327;

NP_064732;







NM_020328

NP_064733



12
91
DOWN
213198_at
NM_004302;
590; 591; 592
NP_004293;
1335; 1336; 1337







NM_020327;

NP_064732;







NM_020328

NP_064733



12

DOWN
222304_x_at

1506 





12
1975
DOWN
211937_at
NM_001417
593
NP_001408
1338



12
5916
DOWN
217178_at
NM_000966
594
NP_000957
1339



12
84975
DOWN
212861_at
NM_032889
595
NP_116278
1340



12
5204
DOWN
207132_x_at
NM_002624;
596; 597; 598
NP_002615;
1341; 1342; 1343







NM_145896;

NP_665903;







NM_145897

NP_665904



12
60314
DOWN
218220_at
NM_021640
599
NP_067653
1344



12
3227
DOWN
206745_at
NM_014212
600
NP_055027
1345



12
3223
DOWN
206194_at
NM_004503;
601; 602
NP_004494;
1346; 1347







NM_153693

NP_710160



12
23583
DOWN
218685_s_at
NM_014311
603
NP_055126
1348



12

DOWN
212126_at

1507 





12
3178
DOWN
200016_x_at
NM_002136;
604; 605
NP_002127;
1349; 1350







NM_031157

NP_112420



12
22818
DOWN
217726_at
NM_016057
606
NP_057141
1351



12
2647
DOWN
202592_at
NM_001487
607
NP_001478
1352



12
967
DOWN
200663_at
NM_001780
608
NP_001771
1353



12
29095
DOWN
218556_at
NM_014182
609
NP_054901
1354



12
11260
DOWN
212160_at
NM_007235
614
NP_009166
1359



12
29110
DOWN
218520_at
NM_013254
615
NP_037386
1360



12
2799
DOWN
212334_at
NM_002076
616
NP_002067
1361



12
2799
DOWN
203676_at
NM_002076
617
NP_002067
1362



12
23592
DOWN
218604_at
NM_014319
618
NP_055134
1363



12
51643
DOWN
219206_x_at
NM_016056
619
NP_057140
1364



12
55832
DOWN
207483_s_at
NM_018448
620
NP_060918
1365



12
55832
DOWN
208838_at
NM_018448
621
NP_060918
1366



12
8445
DOWN
202968_s_at
NM_003583;
622; 623
NP_003574;
1367; 1368







NM_006482

NP_006473



12
5908
DOWN
200833_s_at
NM_015646
624
NP_056461
1369



12
55508
DOWN
218988_at
NM_018656
625
NP_061126
1370



12
8089
DOWN
218911_at
NM_006530
626
NP_006521
1371



12
10576
DOWN
201947_s_at
NM_006431
627
NP_006422
1372



12
4659
DOWN
201603_at
NM_002480
628
NP_002471
1373



12
29080
DOWN
218936_s_at
NM_014167
629
NP_054886
1374



12
91298
DOWN
213701_at

1508 





12
80184
DOWN
221683_s_at
NM_025114
630
NP_079390
1375



12
4254
DOWN
207029_at
NM_000899;
631; 632
NP_000890;
1376; 1377







NM_003994

NP_003985



12
1848
DOWN
208891_at
NM_001946;
633; 634
NP_001937;
1378; 1379







NM_022652

NP_073143



12
490
DOWN
212930_at
NM_001682
635
NP_001673
1380



12
490
DOWN
209281_s_at
NM_001682
636
NP_001673
1381



12
694
DOWN
200921_s_at
NM_001731
637
NP_001722
1382



16
527
DOWN
200954_at
NM_001694
638
NP_001685
1383



16
527
DOWN
36994_at
NM_001694
639
NP_001685
1384



16
51005
DOWN
219082_at
NM_015944
640
NP_057028
1385



16
5170
DOWN
204524_at
NM_002613
641
NP_002604
1386



16
5170
DOWN
32029_at
NM_002613
642
NP_002604
1387



16
23524
DOWN
208610_s_at
NM_016333
643
NP_057417
1388



16
23524
DOWN
213877_x_at
NM_016333
644
NP_057417
1389



17
9570
DOWN
210009_s_at
NM_004287;
233; 234
NP_004278;
984; 985







NM_054022

NP_473363



18
5289
DOWN
204297_at
NM_002647
645
NP_002638
1390



18
498
DOWN
213738_s_at
NM_004046
646
NP_004037
1391



18
9063
DOWN
214593_at
NM_004671;
647; 648
NP_004662;
1392; 1393







NM_173206

NP_775298



18
9063
DOWN
37433_at
NM_004671;
649; 650
NP_004662;
1394; 1395







NM_173206

NP_775298



18
51124
DOWN
211406_at
NM_016097
651
NP_057181;
1396









NP_060992



18
9306
DOWN
214462_at
NM_004232
652
NP_004223
1397



18
4155
DOWN
207323_s_at
NM_002385
653
NP_002376
1398



18
4772
DOWN
210161_at
NM_006162;
654; 655; 666; 657;
NP_006153;
1399; 1400; 1401;







NM_172387;
658
NP_765975;
1402; 1403







NM_172388;

NP_765976;







NM_172389;

NP_765977;







NM_172390

NP_765978



38
9150
DOWN
205035_at
NM_004715;
659; 660
NP_004706;
1404; 1405







NM_048368

NP_430255



18
80148
DOWN
218208_at
NM_025078
661
NP_079354
1406



18
10907
DOWN
202835_at
NM_006701
662
NP_006692
1407



18
79863
DOWN
219419_at
NM_024805
663
NP_079081
1408



18
22850
DOWN
203321_s_at
XM_377498
664
XP_377498
1409



19
5536
DOWN
201979_s_at
NM_006247
319
NP_006238
1070



19
26121
DOWN
202408_s_at
NM_015629
370
NP_056444
1121



19
3802
DOWN
210890_x_at
NM_014218
665
NP_055033
1410



19
9437
DOWN
207860_at
NM_004829
666
NP_004820
1411



21
6612
DOWN
200740_s_at
NM_006936
667
NP_008867
1412



21
754
DOWN
200677_at
NM_004339
668
NP_004330
1413



21
3275
DOWN
202098_s_at
NM_001535
669
NP_001526
1414



22
25812
DOWN
214570_x_at
NM_014348
670
NP_055163
1415



22
28831
DOWN
216846_at

1509 

1510



22
9609
DOWN
211471_s_at
NM_004914
671
NP_004905
1416



22
613
DOWN
202315_s_at
NM_004327;
672; 673
NP_004318;
1417; 1418







NM_021574

NP_067585



22
3543
DOWN
206660_at
NM_020070;
674; 675
NP_064455;
1419; 1420







NM_152855

NP_690594



22
375159
DOWN
215816_at

1511 

1512



22
4320
DOWN
203876_s_at
NM_005940
676
NP_005931
1421



22
6598
DOWN
206532_at
NM_003073
677
NP_003064
1422



22
2952
DOWN
203815_at
NM_000853
678
NP_000844
1423



22
6634
DOWN
202567_at
NM_004175
679
NP_004166
1424



22
89781
DOWN
54037_at
NM_022081;
680; 681; 682; 683;
NP_071364;
1425; 1426; 1427;







NM_152840;
684
NP_690053;
1428; 1429







NM_152841;

NP_690054;







NM_152842;

NP_690055;







NM_152843

NP_690056



22
89781
DOWN
218402_s_at
NM_022081;
685; 686; 687; 688;
NP_071364;
1430; 1431; 1432;







NM_152840;
689
NP_690053;
1433; 1434







NM_152841;

NP_690054;







NM_152842;

NP_690055;







NM_152843

NP_690056



22
24144
DOWN
202750_s_at
NM_012143
690
NP_036275
1435



22
1413
DOWN
206843_at
NM_001886
691
NP_001877
1436



22
4330
DOWN
205330_at
NM_002430
692
NP_002421
1437



22
11200
DOWN
210416_s_at
NM_007194;
693; 694;
NP_009125;
1438; 1439







NM_145862

NP_665861



22
8563
DOWN
209418_s_at

1513 

1514



22
84164
DOWN
215684_s_at
NM_032204
695
NP_115580
1440



22
10291
DOWN
201357_s_at
NM_005877
696
NP_005868
1441



22
23541
DOWN
204541_at
NM_012429
697
NP_036561
1442



22
23598
DOWN
209431_s_at
NM_014323;
698; 699; 700; 701
NP_055138;
1443; 1444; 1445;







NM_032050;

NP_114439;
1446







NM_032051;

NP_114440;







NM_032052

NP_114441



22
9814
DOWN
213431_x_at
XM_038520
702
XP_038520
1447



22
9814
DOWN
36545_s_at
XM_038520
703
XP_038520
1448



22
23761
DOWN
202392_s_at
NM_014338
704
NP_055153
1449



22
9814
DOWN
215699_x_at
XM_038520
705
XP_038520
1450



22
9681
DOWN
205223_at
XM_377498
706
XP_376007
1451



22
7533
DOWN
201020_at
NM_003405
707
NP_003396
1452



22

DOWN
215762_at

1515 





22
7078
DOWN
201149_s_at
NM_000362
708
NP_000353
1453



22
10043
DOWN
202807_s_at
NM_005488
709
NP_005479
1454



22
80830
DOWN
219716_at
NM_030641
710
NP_085144
1455



22
23543
DOWN
212104_s_at
NM_014309
711
NP_055124
1456



22
80833
DOWN
221087_s_at
NM_014349;
712; 713; 714; 715;
NP_055164;
1457; 1458; 1459;







NM_030644;
716; 717
NP_085147;
1460; 1461; 1462







NM_145639;

NP_663614;







NM_145640;

NP_663615;







NM_145641;

NP_663616;







NM_145642

NP_663617



22
23780
DOWN
221653_x_at
NM_030882;
718; 719
NP_112092;
1463; 1464







NM_145637

NP_663612



22
4627
DOWN
211926_s_at
NM_002473
720
NP_002464
1465



22
25828
DOWN
209077_at
NM_012473
721
NP_036605
1466



22
25828
DOWN
209078_s_at
NM_012473
722
NP_036605
1467



22
8664
DOWN
200005_at
NM_003753
723
NP_003744
1468



22
7263
DOWN
209605_at
NM_003312
724
NP_003303
1469



22
4357
DOWN
203524_s_at
NM_021126
725
NP_066949
1470



22
79734
DOWN
205561_at
NM_024681
726
NP_078957
1471



22
4242
DOWN
213783_at
NM_002405
727
NP_002396
1472



22
29775
DOWN
214207_s_at
NM_014550
728
NP_055365
1473



22
26088
DOWN
218114_at
NM_013365
729
NP_037497
1474



22
26088
DOWN
45572_s_at
NM_013365
730
NP_037497
1475



22
23616
DOWN
213633_at
NM_018957
731
NP_061830
1476



22
23464
DOWN
205164_at
NM_014291
732
NP_055106
1477



22
5435
DOWN
209511_at
NM_021974
733
NP_068809
1478



22
1454
DOWN
222015_at
NM_001894;
734; 735
NP_001885;
1479; 1480







NM_152221

NP_689407



22
11015
DOWN
204017_at
NM_006855;
736; 737
NP_006846;
1481; 1482







NM_016657

NP_057839



22
11144
DOWN
208382_s_at
NM_007068
738
NP_008999
1483



22
25776
DOWN
203450_at
NM_015373
739
NP_056188
1484



22
56993
DOWN
217960_s_at
NM_020243
740
NP_064628
1485



22
9929
DOWN
201751_at
NM_014876
741
NP_055691
1486



22
25777
DOWN
212144_at
NM_015374
742
NP_056189
1487



22
9567
DOWN
205274_at
NM_004286;
743
NP_004277;
1488









NP_054746



22
9567
DOWN
219357_at
NM_004286;
744
NP_004277;
1489









NP_054746



22
5155
DOWN
216061_x_at
NM_002608;
745; 746
NP_002599;
1490; 1491







NM_033016

NP_148937



22
10454
DOWN
203901_at
NM_006116;
747; 748
NP_006107;
1492; 1493







NM_153497

NP_705717



22
4248
DOWN
208058_s_at
NM_002409
749
NP_002400
1494



22
54471
DOWN
221516_s_at
NM_019008
750
NP_061881
1495



22
54471
DOWN
204593_s_at
NM_019008
751
NP_061881
1496



22
468
DOWN
200779_at
NM_001675;
752; 753
NP_001666;
1497; 1498







NM_182810

NP_877962



22
4248
DOWN
209764_at
NM_002409
754
NP_002400
1499



22
27352
DOWN
203014_x_at
NM_015705
755
NP_056520
1500

















TABLE 5





Markers of the invention which reside in MCRs of amplification


and display increased expression.























Pos
Gene
Minimum






Chromosome
(Mb)
Weight
p value
Gene Description
Gene Symbol
GI
UGID#





1
26.8
1.03
0.032
nuclear distribution gene C homolog
NUDC
gi: 5729952
Hs.263812






(A. nidulans)


1
116.9
2.41
0.038
transcription termination factor, RNA
TTF2
gi: 5733121
Hs.201774






polymerase II


1
117.8
0.85
0.029
WD repeat domain 3
WDR3
gi: 5803220
Hs.201375


2
11.3
0.06
0.035
hypothetical protein MGC33602
MGC33602
gi: 7328008
Hs.274415


5
0.2
0.69
<.005
succinate dehydrogenase complex,
SDHA
gi: 4759079
Hs.440475






subunit A, flavoprotein (Fp)


5
0.3
0.85
<.005
programmed cell death 6
PDCD6
gi: 7019484
Hs.24087


5
0.5
0.95
<.005
Sec 6 (S. cerevisiae) homolog
SEC6
gi: 3005726
Hs.448580


5
0.6
0.58
0.005
hypothetical protein FLJ10565
FLJ10565
gi: 8922520
Hs.100824


5
0.9
0.77
<.005
hypothetical protein FLJ13441
FLJ13441
gi: 12965190
Hs.449178


5
1.5
0.60
0.014
hypothetical protein FLJ12443
FLJ12443
gi: 13376233
Hs.179882


5
1.8
0.96
<.005
NADH dehydrogenase (ubiquinone) Fe—S
NDUFS6
gi: 4758791
Hs.408257






protein 6, 13 kDa (NADH-coenzyme Q






reductase)


5
5.5
0.72
0.005
KIAA0947 protein
KIAA0947
gi: 13436178
Hs.5070


6
32
0.67
0.032
tenascin XB
TNXB
gi: 8361667
Hs.411644


6
42.9
1.03
<.005
trinucleotide repeat containing 5
TNRC5
gi: 13325207
Hs.414099


6
43
0.25
0.005
protein phosphatase 2, regulatory subunit
PPP2R5D
gi: 5453953
Hs.118244






B (B56), delta isoform


7
0.8
0.47
0.017
G protein-coupled receptor 30
GPR30
gi: 1381668
Hs.113207


7
1.2
0.56
<.005
G protein-coupled receptor 30
GPR30
gi: 2656120
Hs.113207


7
1.2
0.14
<.005
DKFZP586J0619 protein
DKFZP586J0619
gi: 10809392
Hs.112184


7
1.5
0.64
<.005
MICAL-like 2
FLJ23471
gi: 13376030
Hs.376617


7
1.6
0.40
0.026
v-maf musculosponeurotic fibrosarcoma
MAFK
gi: 4505074
Hs.131953






oncogene homolog K (avian)


7
2.1
0.95
<.005
MAD1 mitotic arrest deficient-like 1
MAD1L1
gi: 4505064
Hs.7345






(yeast)


7
30.7
0.47
0.027
glycyl-tRNA synthetase
GARS
gi: 577711
Hs.293885


7
64.9
1.19
0.008
argininosuccinate lyase
ASL
gi: 4502256
Hs.442047


7
65
0.87
0.02
calcitonin gene-related peptide-receptor
RCP9
gi: 7656976
Hs.300684






component protein


7
65.5
1.17
<.005
potassium channel tetramerisation domain
KCTD7
gi: 5596067
Hs.119683






containing 7


7
65.6
1.04
<.005
RAB guanine nucleotide exchange factor
RABGEF1
gi: 7657495
Hs.187660






(GEF) 1


7
65.8
1.42
<.005
hypothetical protein FLJ10099
FLJ10099
gi: 8922228
Hs.287955


7
93.2
0.69
<.005
BET1 homolog (S. cerevisiae)
BET1
gi: 12654162
Hs.23103


7
93.7
0.37
0.05
O-acetyltransferase
CAS1
gi: 12597638
Hs.324725


7
94.6
0.43
0.032
paraoxonase 3
PON3
gi: 1333633
Hs.440967


7
94.6
1.07
<.005
paraoxonase 2
PON2
gi: 2228776
Hs.165598


7
94.8
0.50
0.015
pyruvate dehydorgenase kinase,
PDK4
gi: 4505692
Hs.8364






isoenzyme 4


7
95.4
0.57
0.017
solute carrier family 25, member 13
SLC25A13
gi: 7657580
Hs.9599






(citrin)


7
95.9
1.01
<.005
split hand/foot malformation
SHFM1
gi: 5453639
Hs.333495






(ectrodactyly) type 1


7
96.3
0.42
0.045
ACN9 homolog (S. cerevisiae)
ACN9
gi: 9910179
Hs.42785


7
97.1
0.77
<.005
asparagine synthetase
ASNS
gi: 4502258
Hs.446546


7
97.2
1.23
<.005

Homo sapiens transcribed sequence with


gi: 8008445
Hs.512431






weak similarity to protein pir: PC4369






(H. sapiens) PC4369 olfactory receptor.






HT2 —human (fragment)


7
97.3
0.89
<.005
kinase phosphatase inhibitor 2
KPI2
gi: 7662475
Hs.122708


7
98.1
0.94
<.005
transformation/transcription domain-
TRRAP
gi: 4507690
Hs.203952






associated protein


7
98.2
0.66
<.005
E3 ubiquitin ligase SMURF1
SMURF1
gi: 4738848
Hs.436249


7
98.2
0.70
<.005
transformation/transcription domain-
TRRAP
gi: 3694662
Hs.203952






associated protein


7
98.2
0.58
0.02
E3 ubiquitin ligase SMURF1
SMURF1
gi: 6446605
Hs.436249


7
98.2
0.63
0.005

Homo sapiens cDNA: FLJ21284 fis,


gi: 10437358
Hs.288218






clone COL01911


7
98.5
0.49
0.013

Homo sapiens clone 24438 mRNA


gi: 3283921
Hs.124126






sequence


7
98.5
0.99
<.005
actin related protein ⅔ complex, subunit
ARPC1A
gi: 5454077
Hs.291981






1A, 41 kDa


7
98.5
1.01
<.005
actin related protein ⅔ complex, subunit
ARPC1B
gi: 5031600
Hs.433506






1B, 41 kDa


7
98.7
0.76
<.005
zinc finger protein 95 homolog (mouse)
ZFP95
gi: 11036641
Hs.110839


7
98.8
0.60
0.005
cytochrome P450, family 3, subfamily A,
CYP3A5
gi: 945005
Hs.150276






polypeptide 5


7
98.8
0.67
<.005
cytochrome P450, family 3, subfamily A,
CYP3A5
gi: 4503230
Hs.150276






polypeptide 5


7
99.3
0.59
0.014
adaptor-related protein complex 4, mu 1
AP4M1
gi: 5442365
Hs.194703






subunit


7
99.3
0.98
<.005
TAF6 RNA polymerase II, TATA box
TAF6
gi: 5032146
Hs.289950






binding protein (TBP)-associated factor,






80 kDa


7
99.5
0.38
0.035
postmelotic segregation increased 2-like 6
PMS2L6
gi: 4175684
Hs.367667


7
99.8
0.94
0.05
guanine nucleotide binding protein (G
GNB2
gi: 4885282
Hs.185172






protein), beta polypeptide 2


7
100
0.67
0.005
solute carrier family 12
SLC12A9
gi: 9910385
Hs.437628






(potassium/chloride transporters),






member 9


7
100.4
0.94
<.005
procollagen-lysine, 2-oxoglutarate 5-
PLOD3
gi: 4505890
Hs.153357






dioxygenase 3


7
100.4
0.50
0.018
zine finger, HIT domain containing 1
ZNHIT1
gi: 5453616
Hs.211079


7
100.4
0.36
0.044
tetratricopeptide repeat domain 11
TTC11
gi: 7705631
Hs.423968


7
100.8
0.36
0.047
myosin light chain 2, precursor
MYLC2PL
gi: 12803868
Hs.247831






lymphocyte-specific


7
101.6
0.35
0.007
chromosome 7 open reading frame 19
C7orf19
gi: 12357031
Hs.289053


7
101.6
0.49
0.02
HSPC047 protein
HSPC047
gi: 7661749
Hs.512142


7
101.6
0.36
0.05
polymerase (RNA) II (DNA directed)
POLR2J
gi: 5100572
Hs.489461






polypeptide J, 13.3 kDa


7
101.7
0.38
0.038
DNA directed RNA polymerase II
POLR2J2
gi: 10036750
Hs.406505






polypeptide J-related gene


7
101.7
0.48
<.005
DNA directed RNA polymerase II
POLR2J2
gi: 5901957
Hs.406505






polypeptide J-related gene





7
105
0.84
<.005
hypothetical protein MGC33190
MGC33190
gi: 3231718
Hs.211068


7
105.3
0.36
0.03
synaptophysin-like protein
SYPL
gi: 5235354
Hs.80919


7
106.8
0.76
0.048
solute carrier family 26, member 4
SLC26A4
gi: 4505696
Hs.512611


7
106.9
0.48
0.041
Cas-Br-M (murine) ecotropic retroviral
CBLL1
gi: 13376203
Hs.458382






tranforming sequence-like 1


7
107.1
0.69
0.01
dihydrolipoamide dehydrogenase (E3
DLD
gi: 181574
Hs.74635






component of pyruvate dehydrogenase






complex, 2-oxo-glutarate complex,






branched chain keto acid dehydrogenase






complex)


7
111.1
0.50
0.017
dedicator of cytokinesis 4
DOCK4
gi: 7662263
Hs.118140


8
37.7
0.60
0.037
G protein-coupled receptor 124
GPR124
gi: 11594613
Hs.17270


8
37.7
0.72
0.029
G protein-coupled receptor 124
GPR124
gi: 4739882
Hs.17270


8
37.7
1.12
<.005
BRF2, subunit of RNA polymerase III
BRF2
gi: 11096174
Hs.274136






transcription initiation factor, BRF1-like


8
37.9
0.56
0.008
ash2 (absent, small, or homeotic)-like
ASH2L
gi: 4417209
Hs.6856






(Drosophila)


8
38
0.66
0.005
LSM1 homolog, U6 small nuclear RNA
LSM1
gi: 7657312
Hs.425311






associated (S. cerevisiae)


8
38
0.39
0.046
BCL2-associated athanogene 4
BAG4
gi: 6631074
Hs.194726


8
38.1
0.83
<.005
KIAA0725 protein
KIAA0725
gi: 3882170
Hs.434966


8
38.2
0.46
0.039
Wolf-Hirschhorn syndrome candidate 1-
WHSC1L1
gi: 13699812
Hs.415895






like 1


8
120.7
0.43
0.026
TAP2 RNA polmerase II, TATA box
TAF2
gi: 7022983
Hs.122752






binding protein (TBP)-associated factor,






150 kDa


8
121.3
0.38
0.042
mitochondrial ribosomal protein L13
MRPL13
gi: 7662495
Hs.333823


8
122.5
0.43
0.03
hysturonan synthase 2
HAS2
gi: 4885390
Hs.159226


8
123.9
0.48
0.014
hypothetical protein MGC3067
MGC3067
gi: 8924181
Hs.241576


8
123.9
0.37
0.05
hypothetical protein MGC3067
MGC3067
gi: 13236515
Hs.241576


8
124.1
0.44
0.021
unknown MGC21654 product
MGC21654
gi: 3231900
Hs.95631


8
124.3
0.55
0.008
hypothetical protein FLJ10204
FLJ10204
gi: 8922280
Hs.18029


8
124.6
0.52
0.012
annexin A13
ANXA13
gi: 4757753
Hs.181107


8
125.4
1.03
<.005
ring finger 139
RNF139
gi: 3395786
Hs.228285


8
125.9
0.66
<.005
KIAA0196 gene product
KIAA0196
gi: 7661987
Hs.437991


8
128.7
0.62
0.012
v-myc myelocytomatosis viral oncogene
MYC
gi: 12962934
Hs.202453






homolog (avian)


8
128.9
0.62
0.017

Homo sapiens cDNA FLJ26234 fis, clone


gi: 190753
Hs.459222






ADG09627


8
130.8
0.59
0.013
hypothetical protein BM-009
BM-009
gi: 7705303
Hs.369973


8
133.7
0.43
0.022
CGI-72 protein
CGI-72
gi: 7705782
Hs.44159


8
134.2
0.47
0.023
N-myc downstream regulated gene 1
NDRG1
gi: 5174656
Hs.318567


8
141.8
0.37
0.048
PTK2 protein tyrosine kinase 2
PTK2
gi: 5406748
Hs.434281


8
143.8
0.41
0.034
mescochymal stem cell protein DSCD75
LOC51337
gi: 7706199
Hs.25237


8
144.6
0.59
0.009
KIAA0150 protein
KIAA0150
gi: 1469881
Hs.370491


8
144.7
0.37
0.037
hypothetical protein FLJ12150
FLJ12150
gi: 13376057
Hs.118983


8
144.7
0.37
0.032
eukaryotic translation elongation factor 1
EEF1D
gi: 4622550
Hs.334798






delta (guanine nucleotide exchange






protein)


8
144.8
0.73
<.005
tissue specific transplantation antigen
TSTA3
gi: 6598326
Hs.404119






P35B


8
144.8
0.80
<.005
tissue specific transplantation antigen
TSTA3
gi: 1381178
Hs.404119






P35B


8
144.8
0.61
0.01
KIAA0628 gene product
KIAA0628
gi: 7662213
Hs.43133


8
144.9
0.46
0.022
scribble
SCRIB
gi: 4331493
Hs.436329


8
145.2
0.61
<.005
5-oxoprolinase (ATP-hydrolysing)
OPLAH
gi: 5838792
Hs.305882


8
145.2
0.49
0.014
exosome complex exonuclease RRP41
RRP41
gi: 4534672
Hs.343589


8
145.2
0.55
0.012
exosome complex exonuclease RRP41
RRP41
gi: 9506688
Hs.343589


8
145.2
0.46
0.021
exosome complex exonuclease RRP41
RRP41
gi: 54085 17
Hs.343589


8
145.2
0.37
0.031
GPAA1P anchor attachment protein 1
GPAA1
gi: 6031166
Hs.4742






homolog (yeast)


8
145.2
0.51
0.009
GPAA1P anchor attachment protein 1
GPAA1
gi: 13623546
Hs.4742;






homolog (yeast); GPAA1P anchor


Hs.4742






attachment protein 1 homolog (yeast)


8
145.2
0.45
0.014
GPAA1P anchor attachment protein 1
GPAA1
gi: 7018511
Hs.4742






homolog (yeast)


8
145.2
0.43
0.021
cytochrome c-1
CYC1
gi: 4503184
Hs.289271


8
145.2
0.39
0.03
hypothetical protein DKFZp434N1923;
DKFZP434N1923
gi: 13569949
Hs.295866;






hypothetical protein DKFZp434N1923


Hs.295866


8
145.3
0.31
0.014
brain protein 16
LOC51236
gi: 13124772
Hs.300224


8
145.5
0.33
0.048
diacylglycerol O-acyltransferase homolog
DGAT1
gi: 7382489
Hs.512810






1 (mouse)


8
145.5
0.67
<.005
putative G-protein coupled receptor
FLJ11856
gi: 13375681
Hs.6459






GPCR41


8
145.6
0.43
0.039
cleavage and polyadenylation specific
CPSF1
gi: 10037183
Hs.83727






factor 1, 160 kDa


8
145.6
0.51
0.01
solute carrier family 39 (zinc transporter),
SLC39A4
gi: 8923304
Hs.411274






member 4


8
145.6
0.43
0.031
vacuolar protein sorting 28 (yeast)
VPS28
gi: 7705884
Hs.418175


8
145.6
0.44
0.036

Homo sapiens mRNA, chromosome 1


gi: 7150895
Hs.459379






specific transcript KIAA0496.


9
21.9
0.02
<.005
methylthioadenosine phosphorylase
MTAP
gi: 6006025
Hs.446152


9
35.6
0.48
<.005
tropomyosin 2 (beta)
TPM2
gi: 1219494
Hs.300772


9
35.7
0.52
0.013
talin 1
TLN1
gi: 5454129
Hs.375001


9
35.7
0.89
<.005
cAMP responsive element binding protein 3
CREB3
gi: 2599559
Hs.287921


9
35.7
0.84
<.005
KIAA0258
KIAA0258
gi: 7662029
Hs.47313


9
35.7
0.11
0.032
natriuretic peptide receptor B/guanylate
NPR2
gi: 2337354
Hs.78518






cyclase B (atrionatriuretic peptide






receptor B)


9
35.8
0.67
0.006
nasopharyngeal carcinoma related protein
NGX6
gi: 7706546
Hs.440953


9
36.1
1.33
<.005
clathrin, light polypeptide (Lca)
CLTA
gi: 6005992
Hs.207052


9
36.2
0.20
0.005
clathrin, light polypeptide (Lca)
CLTA
gi: 704460
Hs.207052


9
36.3
0.49
0.021
ring finger protein 38
RNF38
gi: 12232470
Hs.333503


12
22.1
0.62
0.006
cytidine monophosphate N-
CMAS
gi: 8923899
Hs.311346






acetylneuraminic acid synthetase


12
50
0.00
0.005
elastase 1, pancreatic
ELA1
gi: 4503546
Hs.348395


12
54.8
0.77
0.033
myosin, light polypeptide 6, alkali,
MYL6
gi: 2078957
Hs. 77385






smooth muscle and non-muscle


13
22.8
0.84
0.011
ADP-ribosyltransferase (NAD+; poly
ADPRTL1
gi: 11496990
Hs. 437959






(ADP-ribose) polymerase)-like 1


13
23.6
0.67
0.035
myorubularin related protein 6
MTMR6
gi: 1669390
Hs. 79877


13
24.5
2.81
0.042
ring finger protein (C3H2C3 type) 6
RNF6
gi: 12656362
Hs.136885


13
112.9
0.32
0.011
UPF3 regulator of nonsense transcripts
UPF3A
gi: 12620405
Hs.399740






homolog A (yeast)


14
29.1
0.62
0.006
chromosome 14 open reading frame 163
C14orf163
gi: 4240322
Hs.27023


14
29.5
0.41
0.038
adaptor-related protein complex 4, sigma
AP4S1
gi: 12654832
Hs.496614






1 subunit


14
30.1
0.80
<.005
chromosome 14 open reading frame 127
C14orf127
gi: 13376746
Hs.288981


14
30.6
0.46
0.032
Rho GTPase activating protein 5
ARHGAP5
gi: 5905160
Hs.409546


14
31.8
0.56
0.043
neuronal PAS domain protein 3
NPAS3
gi: 11545846
Hs.243209


14
32.9
1.12
0.007
chromosome 14 open reading frame 11
C14orf11
gi: 8922092
Hs.433269


14
33
0.98
<.005
sorting nexin 6
SNX6
gi: 13027619
Hs.283443


14
52.4
1.17
0.013
bone morphogenetic protein 4; bone
BMP4
gi: 576934
Hs.68879;






morphogenetic protein 4


Hs.68879


14
53.2
0.58
0.021
sterile alpha motif domain containing 4
SAMD4
gi: 5689442
Hs.98259


14
53.4
0.48
0.025
WD repeat and HMG-box DNA binding
WDHD1
gi: 7704203
Hs.385998






protein 1


14
53.5
1.36
<.005
chromosome 14 open reading frame 32
C14orf32
gi: 10438139
Hs.406401


14
53.6
0.51
0.013
discs, large homolog 7 (Drosophila)
DLG7
gi: 7661851
Hs.77695


14
53.8
0.42
0.03
F-box only protein 34
FBXO34
gi: 8923650
Hs.15467


14
54
0.29
0.025
kinectin 1 (kinesin receptor)
KTN1
gi: 11681348
Hs. 368212


14
55.6
0.62
0.006
SEC10-like 1 (S. cerevisiae)
SEC10L1
gi: 5730036
Hs. 365863


14
55.7
0.68
<.005
chromosome 14 open reading frame 108
C14orf108
gi: 8922687
Hs.106210


14
56.6
0.65
0.008
actin-related protein 10 homolog
ACTR10
gi: 10433604
Hs.248569






(S. cerevisiae)


14
58.5
0.65
0.035
chromosome 14 open reading frame 135
C14orf135
gi: 11968054
Hs.413671


14
59.8
0.62
0.041
protein kinase C, eta
PRKCH
gi: 5453971
Hs.315366


14
60.1
0.76
0.006
hypoxia-inducible factor 1, alpha subunit
HIF1A
gi: 4504384
Hs.412416






(basic helix-loop-helix transcription






factor)


14
60.2
0.43
0.03
small nuclear RNA activating complex,
SNAPC1
gi: 4507100
Hs. 179312






polypeptide 1, 43 kDa


14
62.9
0.49
0.021
zinc finger and BTB domain containing 1
ZBTB1
gi: 7662437
Hs. 511938


14
63.2
0.03
0.011
KIAA0599
KIAA0599
gi: 13279160
Hs. 198037


14
39.1
0.66
<.005
SWI/SNF related, matrix associated, actin
SMARCE1
gi: 13045953
Hs.437546






dependent regulator of chromatin,






subfamily e, member 1


17
39.3
1.06
<.005
keratin 10 (epidermolytic hyperkeratosis;
KRT10
gi: 4557696
Hs.99936






keratosis palmaris et plantaris)


17
40.4
1.74
<.005
ATP citrate lyase
ACLY
gi: 5768107
Hs.387567


17
40.5
0.97
0.007
I-kappa-B-interacting Ras-like protein 2
KBRAS2
gi: 8922150
Hs.502910


17
40.6
1.17
0.011
RAB5C, member RAS occogene family
RAB5C
gi: 7672664
Hs.479


17
40.7
0.83
0.033
signal transducer and activator of
STAT5B
gi: 9970172
Hs.434992






transcription 5B


17
41
1.01
<.005
ATPase, H+ transporting, lysosomal V0
ATP6V0A1
gi: 4885084
Hs.267871






subunit a isoform 1


17
41
0.69
0.007
transcription factor-like 4
TCFL4
gi: 11761691
Hs.383019


17
41.1
0.89
0.009
GT198, complete ORF
HUMGT198A
gi: 1164152
Hs.279032


17
41.1
1.06
0.009
hypothetical protein LOC162427
LOC162427
gi: 12779367
Hs.432850


17
41.2
1.30
0.005
enhancer of zeste homolog 1 (Drosophila)
EZH1
gi: 2224716
Hs.194669


17
41.2
1.08
<.005
enhancer of zeste homolog 1 (Drosophila)
EZH1
gi: 2224716
Hs.194669


17
41.3
1.62
<.005
HSPC009 protein
HSPC009
gi: 7661731
Hs.16059


17
41.3
1.15
<.005
beclin 1 (coiled-coil, myosin-like BCL2
BECN1
gi: 4502394
Hs.12272






interacting protein)


17
41.3
1.56
<.005
proteasome (prosome, macropain)
PSME3
gi: 5031996
Hs.152978






activator subunit 3 (PA28 gamma; Ki)





17
41.3
1.09
<.005
amine oxidase, copper containing 2
AOC2
gi: 6806881
Hs.143102






(retina-specific)


17
41.4
1.74
<.005
hypothetical protein MGC2744
MFC2744
gi: 3432163
Hs.317403


17
41.5
1.86
<.005
ribosomal protein L27
RPL27
gi: 4506622
Hs.405528


17
41.8
0.44
0.05

Homo sapiens cDNA FLJ35853 fis, clone


gi: 5935951
Hs.277721






TESTt2007078, highly similar to






MEMBRANE COMPONENT,






CHROMOSOME 17, SURFACE






MARKER 2.


17
42.3
0.45
<.005
dual specificity phosphatase 3 (vaccinia
DUSP3
gi: 12803692
Hs.181046






virus phosphatase VH1-related)


17
42.3
1.16
<.005
membrane protein, palmitoylated 3
MPP3
gi: 4505238
Hs.396566






(MAGUK p55 subfamily member 3)


17
42.4
0.63
<.005
membrane protein, palmitoylated 2
MPP2
gi: 6991687
Hs.436326






(MAGUK p55 subfamily member 2)


17
42.6
1.57
<.005
glucose-6-phosphatase catalytic subunit 3
G6PC3
gi: 12951784
Hs.294005


17
42.6
1.39
<.005
glucose-6-phosphatase catalytic subunit 3
G6PC3
gi: 4834429
Hs.294005


17
42.7
1.55
<.005
hypothetical protein MGC3123
MGC3123
gi: 13129117
Hs.181391


17
42.9
0.92
<.005
granulin
GRN
gi: 4504150
Hs.180577


17
42.9
0.45
0.006
KIAA0553 protein
KIAA0553
gi: 11008117
Hs.396047


17
43.6
0.55
0.018
N-myristoyltransferase 1
NMT1
gi: 2760893
Hs.346743


17
43.7
0.34
0.031
HMBA-inducible
HIS1
gi: 7457641
Hs.15299


17
44
1.09
<.005
hypothetical protein FLJ10120
FLJ10120
gi: 8922238
Hs.378860


17
44.8
0.51
0.012
ARF protein
LOC51326
gi: 7770214
Hs.500496


17
44.8
1.09
<.005
KIAA0563 gene product
KIAA0563
gi: 3647821
Hs.38861


17
45.2
0.92
<.005
N-ethylmaleimide-sensitive factor
NSF
gi: 11079227
Hs.431279


17
45.3
0.42
0.041
wingless-type MMTV integration site
WNT3
gi: 13540476
Hs.224667;






family, member 3; wingless-type MMTV


Hs.224667






integration site family, member 3


17
45.7
1.06
<.005

Homo sapiens transcribed sequence with


gi: 1126567
Hs.514263






weak similarity to protein sp: P30260






(H. sapiens) CC27_HUMAN Protein






CD27Hs (Cell division cycle protein 27






homolog) (H-NUC)


17
46.1
0.55
0.01
aminopeptidase poromycin sensitive
NPEPPS
gi: 4210725
Hs.293007


17
46.2
0.31
<.005
karyopherin (importin) beta 1
KPNB2
gi: 13097743
Hs.439683


17
46.4
0.87
0.01
pyridoxine-5′-phosphate oxidase
PNPO
gi: 8922497
Hs.327335


17
46.6
1.27
<.005
chromobox homolog 1 (HP1 beta
CBX1
gi: 5803075
Hs.77254






homolog Drosophila)


17
46.6
0.81
0.008
sorting nexin 11
SNX11
gi: 7019538
Hs.15827


17
46.6
0.78
0.01
sorting nexin 11
SNX11
gi: 4827951
Hs.15827


17
47.4
0.86
<.005
ATP synthase, H+ transporting,
ATP5G1
gi: 5262506
Hs.80986






mitochondrial P0 complex, subunit c






(subunit 9), isoform 1


17
47.4
0.60
0.017
hypothetical protein FLJ13855
FLJ13855
gi: 12751494
Hs.369120


17
47.4
1.17
<.005
EAP30 subunit of ELL complex
EAP30
gi: 6005754
Hs.127249


17
47.8
0.49
0.021
KIAA0924 protein
KIAA0924
gi: 7662383
Hs.190386


17
47.9
1.15
<.005
prohibitin
PHB
gi: 6031190
Hs.75323


17
48.1
1.06
<.005
specide-type POZ protein
SPOP
gi: 4507182
Hs.129951


17
48.2
1.61
<.005
solute carrier family 35, member B1
SLC35B1
gi: 5032212
Hs: 154073


17
48.2
0.82
<.005
C/EBP-induced protein; C/EBP-induced
LOC81558
gi: 13540589
Hs.9851;






protein


Hs.9851


17
48.3
1.18
<.005
MYST histone acetyltransferase 2
MYST2
gi: 5901961
Hs.21907


17
48.6
0.45
0.026
integrin, alpha 3 (antigen CD49C, alpha 3
ITGA3
gi: 4504746
Hs.265829






subunit of VLA-3 receptor)





17
48.6
0.37
0.047
pyruvate dehydrogenase kinase,
PDK2
gi: 5544583
Hs.92261






isoenzyme 2


17
48.9
0.45
0.03
xylosyltransferase II
XYLT2
gi: 11545913
Hs.32117


17
48.9
0.91
<.005
hypothetical protein PRO1855
PRO1855
gi: 10437822
Hs.370927


17
49
0.79
<.005
hypothetical protein FLJ20920
FLJ20920
gi: 13376740
Hs.288959


17
49
0.57
0.017
hypothetical protein FLJ11164
FLJ11164
gi: 8922910
Hs.8033


17
49
0.60
0.016
epsin 3
EPN3
gi: 8923677
Hs.165904


17
49.1
0.57
0.018
hypothetical protein FLJ21347
FLJ21347
gi: 12383067
Hs.103147


17
49.2
0.93
<.005
hypothetical protein MGC15396;
MGC15396
gi: 13543385
Hs.351247;






hypothetical protein MGC15396


Hs.351247


17
49.2
0.42
0.034
cisplatin resistance-associated
LUC7A
gi: 7706534
Hs.130293






overexpressed protein


17
49.3
0.40
0.041
cisplatin resistance-associated
LUC7A
gi: 5174618
Hs.130293






overexpressed protein


17
49.5
0.69
0.017
sperm associated antigen 9
SPAG9
gi: 4504524
Hs.500367


17
49.7
1.32
<.005
non_metastatic cells 1, protein (NM23A)
NME1
gi: 4557796
Hs.118638






expressed in





17
49.7
1.75
<.005
non_metastatic cells 2, protein (NM23B)
NME2
gi: 4505408
Hs.433416






expressed in


17
49.8
1.01
0.005
CGI-48 protein
CGI-48
gi: 3179644
Hs.441503


17
53.5
0.61
0.005
COXII homolog, cytochrome c oxidase
COXII
gi: 4186577
Hs.436988






assembly protein (yeast)


17
53.8
0.23
0.01
hepatic leukemia factor
HLF
gi: 184223
Hs.250692


17
54.3
0.64
0.005
phosphatidylcholine transfer protein
PCTP
gi: 10864026
Hs.285218


17
74
0.63
0.028
KIAA0195 gene product
KIAA0195
gi: 7661985
Hs.301132


17
74
1.07
0.009
CASK interacting protein 2
CASKIN2
gi: 5766922
Hs.274408


17
74
0.76
0.014
CASK interacting protein 2
CASKIN2
gi: 5928096
Hs.274408


18
19.3
0.56
0.022
RIO kinase 3 (yeast)
RIOK3
gi: 4507298
Hs.209061


18
19.3
0.79
0.006
RIO kinase 3 (yeast)
RIOK3
gi: 5855068
Hs.209061


18
19.3
1.02
<.005
colon cancer-associated protein Mic1
MIC1
gi: 7019454
Hs.287633


18
19.3
0.95
<.005
Niemann-Pick disease, type C1
NPC1
gi: 4557802
Hs.404930


18
19.9
0.48
0.024
calcium-binding tyrosine-(Y)-
CABYR
gi: 6912377
Hs.511983






phosphorylation regulated (fibrousheathin









2)


18
20.1
0.48
0.022
oxysterol binding protein-like 1A;
OSBPL1A
gi: 13877169
Hs.415753;






oxysterol binding protein-like 1A;


Hs.415753


19
41.4
1.18
<.005
zinc finger protein 146
ZNF146
gi: 6005965
Hs.301819


19
42.9
0.71
0.012
hypothetical protein FLJ30921
FLJ30921
gi: 9722561
Hs.290703


19
43.3
0.53
0.028
D4, zinc and double PHD fingers family 1
DPF1
gi: 4758797
Hs.389057


19
43.5
1.08
<.005
proteasome (prosome, macropain) 26S
PSMD8
gi: 4506232
Hs.78466






subunit, non-ATPase, 8


19
43.7
0.54
0.027
mitogen-activated protein kinase kinase
MAP4K1
gi: 9970929
Hs.95424






kinase kinase 1


19
43.8
1.41
<.005
eukaryotic translation initiation factor 3
eIF3k
gi: 5114050
Hs.143773






subunit k


19
43.8
1.43
<.005
eukaryotic translation initiation factor 3
eIF3k
gi: 6038285
Hs.143773






subunit k


19
44
0.43
0.013
heterogeneous nuclear ribonucleoprotein L
HNRPL
gi: 5935937
Hs.446623


19
44
1.01
<.005
sirtutin (silent mating type information
STRT2
gi: 13775599
Hs.375214






regulation 2 homolog) 2 (S. cerevisiae)





19
44
0.70
<.005
nuclear factor of kappa light polypeptide
NFKBIB
gi: 4505384
Hs.9731






gene enhancer in B-cells inhibitor, beta


19
44.1
0.86
<.005
seryl-tRNA synthetase 2
SARS2
gi: 8923420
Hs.14220


19
44.1
0.12
<.005
mitochondrial ribosomal protein S12
MRPS12
gi: 2252149
Hs.411125


19
44.1
0.56
0.019
F-box only protein 26
FBXO26
gi: 13376364
Hs.425352


19
44.3
0.40
<.005
p21(CDKNIA)-activated kinase 4
PAK4
gi: 7382497
Hs.20447


19
44.3
0.83
<.005
p21(CDKNIA)-activated kinase 4
PAK4
gi: 4101586
Hs.20447


19
44.5
1.16
<.005
hypothetical protein F23149_1
PD2
gi: 9506582
Hs.152894


19
50.5
3.52
0.045
RelA-associated inhibitor
RAI
gi: 5730000
Hs.324051


19
50.6
1.27
<.005
CD3-epsilon-associated protein; antisense
ASE-1
gi: 6912245
Hs.446684






to ERCC-1


19
50.7
3.60
0.045
optic atrophy 3 (autosomal recessive, with
OPA3
gi: 13376716
Hs.123473






chorea and spastic parsplegia)


19
50.8
3.04
0.045
echinoderm microtubulo associated
EML2
gi: 4568182
Hs.24178






protein like 2


19
50.8
0.98
0.018
gastric inhibitory polypeptide receptor
GIPR
gi: 4503998
Hs.251412


19
50.8
1.00
0.044
small nuclear ribonucleoprotein D2
SNRPD2
gi: 7242206
Hs.424327






polypeptide 16.5 kDa





19
50.9
0.65
0.012

Homo sapiens cDNA FLJ90345 fis, clone


gi: 7151592
Hs.43314






NT2RP2002974, highly similar to






HOMEOBOX PROTEIN SIX5.


19
50.9
0.49
<.005
dystrophis myotonica-protein kinase
DMPK
gi: 189038
Hs.898


19
51.8
0.71
0.005
protein kinase D2
PRKD2
gi: 12659006
Hs.205431


19
51.8
0.65
0.009
protein kinase D2
PRKD2
gi: 4884153
Hs.205431


19
51.9
1.42
0.008
striatin, calmodulin binding protein 4
STRN4
gi: 7019572
Hs.406918


19
51.9
0.90
0.009
solute carrier family 1 (neutral amino acid
SLC1A5
gi: 4191561
Hs.183556






transporter), member 5


19
52
1.66
<.005
adaptor-related protein complex 2, sigma
AP2S1
gi: 11038644
Hs.119591






1 subunit


19
52
1.35
<.005
adaptor-related protein complex 2, sigma
AP2S1
gi: 13623468
Hs.119591;






1 subunit; adaptor-related protein


Hs.119591






complex 2, sigma 1 subunit


19
52.1
1.87
0.014
glucocorticoid receptor DNA binding
GRLF1
gi: 4758481
Hs.102548






factor 1


19
52.5
1.05
0.036
complement component 5 receptor 1 (C5a
C5R1
gi: 4502508
Hs.2161






ligand)


19
52.6
2.86
<.005
N-ethylmaleimide-sensitive factor
NAPA
gi: 4505328
Hs.75932






attachment protein, alpha


19
52.9
3.79
0.045
EH-domain containing 2
EHD2
gi: 7657053
Hs.325650


19
52.9
5.29
0.045
EH-domain containing 2
EHD2
gi: 4261421
Hs.325650


19
52.9
4.35
0.045
EH-domain containing 2
EHD2
gi: 4261421
Hs.325650


19
53.3
2.23
0.045
liase 1, DNA, ATP-dependent
LIG1
gi: 4557718
Hs.1770


19
53.5
1.97
0.045
KDEL (Lys-Asp-Glu-Leu) endoplasmic
KDELR1
gi: 5803047
Hs.78040






reticulum protein retention receptor 1


19
53.6
3.06
0.045
glutamate-rich WD repeat containing 1
GRWD1
gi: 13274610
Hs.400625


19
53.8
1.25
0.041
ribosomal protein L18
RPL18
gi: 4834123
Hs.409634


19
53.8
1.82
0.013
D site of albumin promoter (albumin D-
DBP
gi: 460704
Hs.414480






box) binding protein


19
53.9
0.88
0.033
fucosyltransferase 1 (galactoside 2-alpha-
FUT1
gi: 4503804
Hs.69747






L-fucosyltransferase)


19
53.9
0.93
0.013
fucosyltransferase 1 (galactoside 2-alpha-
FUT1
gi: 6739499
Hs.69747






L-fucosyltransferase)


19
54
0.58
0.036
pleckstrin homology domain containing,
PLEKHA4
gi: 10190743
Hs.9469






family A (phosphoinositide binding






specific) member 4


19
54.1
0.43
0.044
nucleobindin 1
NUCB1
gi: 5453817
Hs.172609


19
54.1
0.63
0.032
BCL2-associated X protein
BAX
gi: 4751837
Hs.159428


19
54.1
1.36
<.005
glycogen synthase 1 (muscle)
GYS1
gi: 4504232
Hs.386225


19
54.1
1.13
<.005
RuvB-like 2 (E. coli)
RUVBL2
gi: 5730022
Hs.6455


19
54.2
0.94
0.011
luteinizing hormone beta polypeptide
LHB
gi: 4504988
Hs.154704


19
54.2
2.38
<.005
small nuclear ribonucleoprotein 70 kDa
SNRP70
gi: 4507118
Hs.174051






polypeptide (RNP antigen)


19
54.5
0.95
0.011
soggy-1 gene
DKKL1-
gi: 7657553
Hs.124021







pending


19
54.6
0.51
0.034
ribosomal protein L13a
RPL13A
gi: 12653484
Hs.449070


19
54.7
0.92
0.029
reticulocalbin 3, EF-hand calcium binding
RCN3
gi: 10257434
Hs.439184






domain


19
54.7
0.99
0.012
nitric oxide synthase interacting protein
NOSIP
gi: 7705715
Hs.7236


19
54.8
0.65
0.017
interferon regulatory factor 3
IRF3
gi: 4504724
Hs.75254


19
54.8
2.10
0.045
HMT1 hnRNP methyltransferase-like 2
HRMT1L2
gi: 4504496
Hs.20521






(S. cerevisiae)


19
55
1.21
0.014
prostate tumor overexpressed gene 1
PTOV1
gi: 4884338
Hs.227429


19
55.1
2.89
0.045
nucleoporin 62 kDa
NUP62
gi: 7705354
Hs.437023


19
55.5
0.48
0.024
nuclear receptor subfamily 1, group H,
NR1H2
gi: 11321629
Hs.432976






member 2


19
56.2
0.11
0.007
kallikrein 13
KLK13
gi: 4884461
Hs.165296


19
58.7
1.51
<.005
zinc finger protein 331
ZNF331
gi: 10092612
Hs.147644


19
59.3
1.45
<.005
CCR4-NOT transcription complex,
CNOT3
gi: 7657386
Hs.343571






subunit 3


19
59.3
0.87
0.013
leukocyte receptor cluster (LRC) member 4
LENG4
gi: 13236521
Hs.78768


16
59.3
0.65
0.013
leukocyte receptor cluster (LRC) member 5
LENG5
gi: 13129061
Hs.15580


19
59.4
0.73
0.019
ribosomal protein S9
RPS9
gi: 4506744
Hs.139876


19
60.3
0.50
0.018
synaptotagmin V
SVT5
gi: 4763527
Hs.23179


19
60.8
1.68
0.01
LDL induced EC protein
LOC51157
gi: 7705880
Hs.94392


19
60.8
0.88
0.019
U2 (RNU2) small nuclear RNA auxiliary
U2AF2
gi: 5396722
Hs.297629






factor 2


19
60.8
1.08
0.027
epsin 1
EPN1
gi: 7019368
Hs.279953


19
61.2
1.42
<.005
hypothetical protein LOC126208
LOC126108
gi: 1496048
Hs.397153


19
61.3
0.55
0.039
zinc finger protein 444
ZNF444
gi: 8922893
Hs.24545


19
61.3
0.62
0.027
zinc finger protein 444
ZNF444
gi: 3916863
Hs.24545


19
62.4
0.69
0.022
zinc finger protein 264
ZNF264
gi: 4585642
Hs.426358


19
62.4
0.79
<.005
zinc finger protein 272
ZNF272
gi: 498733
Hs.99971


19
62.5
0.44
0.043
zinc finger protein 304
ZNF304
gi: 10190695
Hs.287374


19
62.6
0.66
0.008
hypothetical protein FLJ23233
FLJ23233
gi: 13375967
Hs.98593


19
62.6
0.60
0.023
hypothetical protein FLJ23233
FLJ23233
gi: 2112716
Hs.98593


19
62.8
0.60
0.022
zinc finger protein 134 (clone pHZ-15)
ZNF134
gi: 4507982
Hs.449971


19
62.8
1.08
<.005
zinc finger protein 211
ZNF211
gi: 5454175
Hs.449970


20
30.9
0.62
0.008
inhibitor of DNA binding 1, dominant
ID1
gi: 464181
Hs.410900






negative helix-loop-helix protein


20
58.2
0.70
<.005
ATP synthase, H+ transporting,
ATP5E
gi: 5901895
Hs.177530






mitochondrial F1 complex, epsilon






subunit


20
58.2
0.65
<.005
chromosome 20 open reading frame 45
C20orf45
gi: 7705609
Hs.3945


20
59.2
0.68
<.005
protein phosphatase 1, regulatory subunit
PPP1R3D
gi: 6806895
Hs.504920






3D


20
61.2
0.00
0.025
TAF4 RNA polymerase II, TATA box
TAF4
gi: 5112317
Hs.24644






binding protein (TBP)-associated factor.






135 kDa


20
61.4
0.45
0.015
synovial sarcoma translocation gene on
SS18L1
gi: 3327199
Hs.154429






chromosome 18-like 1


20
61.4
0.38
0.048
proteasome (prosome, macropain)
PSMA7
gi: 9408092
Hs.233952






subunit, alpha type, 7


20
61.5
0.59
<.005
oxysterol binding protein-like 2
OSBPL2
gi: 12653062
Hs.15519


20
61.5
0.37
0.049
laminin, alpha 5
LAMA5
gi: 13097167
Hs.11669


20
61.6
0.06
<.005
ribosomal protein S21
RPS21
gi: 4506698
Hs.372960


20
62.2
0.49
0.018
transcription factor-llke 5 (basic helix-
TCFL5
gi: 5730082
Hs.30696






loop-helix)


20
62.2
0.39
0.03
chromosome 20 open reading frame 11
C20orf11
gi: 8923556
Hs.103808


20
62.5
0.82
<.005
chromosome 20 open reading frame 21
C20orf21
gi: 9663381
Hs.11747


20
63
0.46
0.018
ADP-ribosylation factor related protein 1
ARFRP1
gi: 8246778
Hs.389277


20
63
0.41
0.032
KIAA1847
KIAA1847
gi: 8246778
Hs.11900


20
63
0.37
0.03
ADP-ribosylation factor related protein 1
ARFRP1
gi: 4507448
Hs.389277


20
63
0.48
0.016
KIAA1847
KIAA1847
gi: 2279318
Hs.11900


20
63.2
0.77
<.005
tumor protein D52-like 2
TPD52L2
gi: 4507642
Hs.154718


20
63.3
0.73
<.005
uridine kinase-like 1
URKL1
gi: 8923486
Hs.504998


20
63.3
0.75
<.005
chromosome 20 open reading frame 14
C20orf14
gi: 13401215
Hs.31334


22
22.6
0.59
0.031
D-dopachrome tautomerase
DDT
gi: 5453630
Hs.433902


22
22.7
0.42
0.018
glutathione S-transferase theta 1
GSTT1
gi: 4504184
Hs.268573


22
29.6
0.46
0.015
hypothetical protein FLJ20618
FLJ20618
gi: 7021031
Hs.52184


22
29.6
0.65
<.005
hypothetical protein FLJ20618
FLJ20618
gi: 4899032
Hs.52184





















Ref Seq
SEQ ID NO.:
Ref Seq
SEQ ID NO.:


Chromosome
Locus Link
Regulation
Probes
mRna ID
Nuc.
Prot ID
A.A.





1
10726
UP
201173_x_at
NM_006600
445
NP_006591
1196


1
8458
UP
204407_at
NM_003594
1
NP_003585
756


1
10885
UP
218882_s_at
NM_006784
2
NP_006775
757


2
130814
UP
216458_at
NM_152391
458
NP_689604
1209


5
6389
UP
201093_x_at
NM_004168
3
NP_004159
758


5
10016
UP
203415_at
NM_013232
4
NP_037364
759


5
11336
UP
212630_at
NM_007277
5
NP_009208
760


5
55722
UP
219531_at
NM_018140
6
NP_060610
761


5
65980
UP
220155_s_at
NM_023924
7
NP_076413
762


5
79888
UP
201818_at
NM_024830
8
NP_079106
763


5
4726
UP
203606_at
NM_004553
9
NP_004544
764


5
23379
UP
209654_at
XM_029101
10
XP_029101
765


6
7148
UP
213451_x_at
NM_019105;
11; 12
NP_061978;
766; 767






NM_032470

NP_115859


6
10695
UP
217931_at
NM_006586;
13; 14
NP_006577;
768; 769






NM_183010

NP_898828


6
5528
UP
202513_s_at
NM_006245;
15; 16; 17
NP_006236;
770; 771; 772






NM_180976;

NP_851307;






NM_180977

NP_851308


7
2852
UP
211829_s_at
NM_001505
18
NP_001496
773


7
2852
UP
210640_s_at
NM_001505
19
NP_001496
774


7
26173
UP
212212_s_at
XM_291222
20
XP_291222
775


7
79778
UP
219332_at
NM_024723;
21; 22
NP_078999;
776; 777






NM_182924

NP_891554


7
7975
UP
206750_at
NM_002360
23
NP_002351
778


7
8379
UP
204857_at
NM_003550
24
NP_003541
779


7
2617
UP
208693_s_at
NM_002047
25
NP_002038
780


7
435
UP
204608_at
NM_000048
26
NP_000039
781


7
27297
UP
203899_s_at
NM_014478
27
NP_055293
782


7
154881
UP
213474_at
NM_153033
28
NP_694578
783


7
27342
UP
218310_at
NM_014504
29
NP_055319
784


7
55069
UP
218008_at
NM_017994
30
NP_060464
785


7
10282
UP
202710_at
NM_005868
31
NP_005859
786


7
64921
UP
219342_at
NM_022900
32
NP_075051
787


7
5446
UP
213695_at
NM_000940
33
NP_000931
788


7
5445
UP
210830_s_at
NM_000305
34
NP_000296
789


7
5166
UP
205960_at
NM_002612
35
NP_002603
790


7
10165
UP
203775_at
NM_014251
36
NP_055066
791


7
7979
UP
202276_at
NM_006304
37
NP_006295
792


7
57001
UP
218981_at
NM_020186
38
NP_064571
793


7
440
UP
205047_s_at
NM_001673;
39; 40; 41
NP_001664;
794; 795; 796






NM_133436;

NP_597680;






NM_183356

NP_899199


7

UP
217499_x_at

1516


7
22853
UP
206223_at
NM_014916
42
NP_055731
797


7
8295
UP
20642_s_at
NM_003496
43
NP_003487
798


7
57154
UP
212668_at
NM_020429;
44; 45
NP_065162;
799; 800






NM_181349

NP_851994


7
8295
UP
214908_a_at
NM_003496
46
NP_003487
801


7
57154
UP
215458_s_at
NM_020429;
47; 48
NP_065162;
802; 803






NM_181349

NP_851994


7

UP
215589_at

1517




7

UP
215457_at

1518




7
10552
UP
200950_at
NM_006409
49
NP_006400
804


7
10095
UP
201954_at
NM_005720
50
NP_005711
805


7
23600
UP
203731_s_at
NM_014569;
51; 52
NP_055384;
806; 807






NM_145102

NP_659570


7
1577
UP
214234_s_at
NM_000777
53
NP_000768
808


7
1577
UP
205765_at
NM_000777
54
NP_000768
809


7
9179
UP
209837_at
NM_004722
55
NP_004713
810


7
6878
UP
203572_s_at
NM_005641;
56; 57; 58; 59
NP_005632;
811; 812; 813; 814






NM_139122;

NP_620834;






NM_139123;

NP_620835;






NM_139315

NP_647476


7
5384
UP
215667_x_at

1519




7
2783
UP
200852_x_at
NM_005273
60
NP_005264
815


7
56996
UP
220371_s_at
NM_020246
61
NP_064631
816


7
8985
UP
202185_at
NM_001084
62
NP_001075
817


7
10467
UP
201541_s_at
NM_006349
63
NP_006340
818


7
51024
UP
218034_at
NM_016068
64
NP_057152
819


7
93408
UP
221659_s_at
NM_138403
65
NP_612412
820


7
80228
UP
218811_at
NM_032831
66
NP_079432;
821








NP_116220


7
29060
UP
220692_at
NM_014147
67
NP_054866
822


7
5439
UP
212707_s_at
NM_006234
68
NP_006225
823


7
246721
UP
214740_at
NM_032958;
69; 70; 71
NP_116580;
824; 825; 826






NM_032959;

NP_116581;






NM_145325

NP_663165


7
246721
UP
208534_s_at
NM_032958;
72; 73; 74
NP_116580;
827; 828; 829






NM_032959;

NP_116581;






NM_145325

NP_663165


7
222255
UP
214342_at
NM_152749
75
NP_689962
830


7
6856
UP
201259_s_at
NM_006754;
76; 77
NP_006745;
831; 832






NM_182715

NP_874384


7
5172
UP
206529_x_at
NM_000441
78
NP_000432
833


7
79872
UP
220018_at
NM_024814
79
NP_079090
834


7
1738
UP
209095_at
NM_000108
80
NP_000099
835


7
9732
UP
205003_at
NM_014705
81
NP_055520
836


8
25960
UP
221814_at
NM_032777
82
NP_116166
837


8
25960
UP
65718_at
NM_032777
83
NP_116166
838


8
55290
UP
218954_s_at
NM_018310
84
NP_060780
839


8
9070
UP
209517_s_at
NM_004674
85
NP_004665
840


8
27257
UP
203534_at
NM_014462
86
NP_055277
841


8
9530
UP
219624_at
NM_004874
87
NP_004865
842


8
23259
UP
212690_at
NM_291291
88
XP_291291
843


8
54904
UP
218173_s_at
NM_017778;
89; 90
NP_060248;
844; 845






NM_023034

NP_075447


8
6873
UP
209523_at
NM_003184
91
NP_003175
846


8
28998
UP
218049_s_at
NM_014078
92
NP_054797
847


8
3037
UP
206432_at
NM_005328
93
NP_005319
848


8
79139
UP
218172_s_at
NM_024295
94
NP_061100;
849








NP_077271


8
79139
UP
219402_s_at
NM_024295
95
NP_061100;
850








NP_077271


8
93594
UP
214061_at
NM_145647
96
NP_663622
851


8
55093
UP
219060_at
NM_018024
97
NP_060494
852


8
312
UP
208323_s_at
NM_004306
98
NP_004297
853


8
11236
UP
209510_at
NM_007218
99
NP_009149
854


8
9897
UP
201985_at
NM_014846
100
NP_055661
855


8
4609
UP
202431_s_at
NM_002467
101
NP_002458
856


8
375682
UP
216240_at

1520




8
51571
UP
217916_s_at
NM_016623
102
NP_057707
857


8
51105
UP
219606_at
NM_016018;
103; 104; 105; 106
NP_057102;
858; 859; 860; 861






NM_024878;

NP_079154;






NM_032205;

NP_115581;






NM_198513

NP_940915


8
10397
UP
200632_s_at
NM_006096
107
NP_006087
862


8
5747
UP
208820_at
NM_005607;
108; 109
NP_005598;
863; 864






NM_153831

NP_722560


8
51337
UP
218500_at
NM_016647
110
NP_057731
865


8
23144
UP
213445_at
NM_015117
111
NP_055932
866


8
79792
UP
218154_at
NM_024736
112
NP_079012
867


8
1936
UP
214394_x_at
NM_001960;
113; 114
NP_001951;
868; 869






NM_032378

NP_115754


8
7264
UP
201644_at
NM_003313
115
NP_003304
870


8
7264
UP
36936_at
NM_003313
116
NP_003304
871


8
9831
UP
206188_at

1521

1522


8
23513
UP
212556_at
NM_015356;
117
NP_056171;
872; 873






NM_182706

NP_874365


8
26873
UP
222025_s_at
XM_291266
118
XP_291266
874


8
54512
UP
91682_at
NM_019037
119
NP_061910
875


8
54512
UP
218695_at
NM_019037
120
NP_061910
876


8
54512
UP
58696_at
NM_019037
121
NP_061910
877


8
8733
UP
201618_x_at
NM_003801
122
NP_003792
878


8
8733; 8733
UP
211060_x_at
NM_003801
123
NP_003792
879


8
8733
UP
215690_x_at
NM_003801
124
NP_003792
880


8
1537
UP
201066_at
NM_001916
125
NP_001907
881


8
81858;
UP
220973_s_at
NM_030974
126
NP_112236
882



81858


8
51236
UP
219071_x_at
NM_016458
127
NP_057542
883


8
8694
UP
203669_s_at
NM_012079
128
NP_036211
884


8
79581
UP
218151_x_at
NM_024531
129
NP_078807
885


8
29894
UP
201638_s_at
NM_013291
130
NP_037423
886


8
55630
UP
219215_s_at
NM_017767;
131; 132
NP_060237;
887; 878






NM_130849

NP_570901


8
51160
UP
218679_s_at
NM_016208;
133; 134
NP_057292;
879; 880






NM_183057

NP_898880


8

UP
213681_at

1523




9
4507
UP
204956_at
NM_002451
555
NP_002442
1303


9
7169
UP
212654_at
NM_003289
135
NP_003280
891


9
7094
UP
203254_s_at
NM_006289
136
NP_006280
892


9
10488
UP
209432_s_at
NM_006368
137
NP_006359
893


9
9827
UP
203169_at
XM_376830
138
XP_376830
894


9
4882
UP
214066_x_at
NM_000907;
139; 140
NP_000898;
895






NM_003995

NP_003986


9
51754
UP
207839_s_at
NM_016446
141
NP_057530
896


9
1211
UP
200960_x_at
NM_001833;
142; 143
NP_001824;
897; 898






NM_007096

NP_009027


9
1211
UP
216293_at
NM_001833;
144; 145
NP_001824;
899; 900






NM_007096

NP_009027


9
152006
UP
218528_s_at
NM_022781;
146; 147; 148; 149;
NP_073618;
901; 902; 903; 904;






NM_194328;
150; 151
NP_919309;
905; 906






NM_194329;

NP_919310;






NM_194330;

NP_919311;






NM_194331;

NP_919312;






NM_194332

NP_919313


12
55907
UP
218111_s_at
NM_018686
152
NP_061156
907


12
1990
UP
206446_s_at
NM_001971
586
NP_001962
1331


12
4637
UP
214002_at
NM_021019;
610; 611; 612; 613
NP_066299;
1355; 1356; 1357;






NM_079423;

NP_524147;
1358






NM_079424;

NP_524148;






NM_079425

NP_524149


13
143
UP
202239_at
NM_006437
153
NP_006428
908


13
9107
UP
214429_at
NM_004685
154
NP_004676
909


13
6049
UP
210931_at
NM_005977;
155; 156; 157; 158
NP_005968;
910; 911; 912; 913






NM_183043;

NP_898864;






NM_183044;

NP_898865;






NM_183045

NP_898866


13
65110
UP
206958_s_at
NM_023011;
159; 160
NP_075387;
914; 915






NM_080687

NP_542418


14
23256
UP
215548_s_at
NM_016106;
161; 162
NP_057190;
916; 917






NM_182835

NP_878255


14
11154
UP
210952_at
NM_007077
163
NP_009008
918


14
80224
UP
220176_at
NM_025152
164
NP_079428
919


14
394
UP
217936_at
NM_001173
165
NP_001164
920


14
64067
UP
220316_at
NM_022123;
166; 167
NP_071406;
921; 922






NM_173159

NP_775182


14
55837
UP
202623_at
NM_018453
168
NP_060923
923


14
58533
UP
217789_at
NM_021249;
169; 170
NP_067072;
924; 925






NM_152233

NP_689419


14
652; 652
UP
211518_s_at
NM_001202;
171; 172; 173; 174;
NP_001193;
926; 927; 928






NM_130850;
175; 176
NP_570911;






NM_130851;

NP_570912






NM_001202;






NM_130850;






NM_130851


14
23034
UP
212845_at
NM_015589
177
NP_056404
929


14
11169
UP
204727_at
NM_007086
178
NP_009017
930


14
93487
UP
212499_s_at
NM_144578
179
NP_653179
931


14
9787
UP
203764_at
NM_014750
180
NP_055565
932


14
55030
UP
218539_at
NM_017943
181
NP_060413
933


14
3895
UP
200914_x_at
NM_182926
182
NP_891556
934


14
10640
UP
218748_s_at
NM_006544
183
NP_006535
935


14
55745
UP
218139_s_at
NM_018229
184
NP_060699
936


14
55860
UP
222230_s_at
NM_018477
185
NP_060947
937


14
64430
UP
219972_s_at
NM_022495
186
NP_071940
938


14
5583
UP
206099_at
NM_006255;
187
NP_006246;
939








NP_076969


14
3091
UP
200989_at
NM_001530;
188; 189
NP_001521;
940; 941






NM_181054

NP_851397


14
6617
UP
205443_at
NM_003082
190
NP_003073
942


14
22890
UP
205092_x_at
XM_375086
191
XP_375086
943


14
26030
UP
217044_s_at

1524

1525


14
6605
UP
211988_at
NM_003079
192
NP_003070
944


17
3858
UP
207023_x_at
NM_000421
193
NP_000412
945


17
47
UP
201127_s_at
NM_001096;
194; 195
NP_001087;
946






NM_198830

NP_942127


17
28511
UP
218240_at
NM_017595
196
NP_060065
947


17
5878
UP
201156_s_at
NM_004583;
197; 198
NP_004574;
948; 949






NM_201434

NP_958842


17
6777
UP
212549_at
NM_012448
199
NP_036580
950


17
535
UP
205095_s_at
NM_005177
200
NP_005168
951


17
6945
UP
210752_s_at
NM_170607;
201; 202; 203
NP_733752;
952; 953; 954






NM_198204;

NP_937847;






NM_198205

NP_937848


17
29893
UP
213708_s_at
NM_013290;
204; 205
NP_037422;
955; 956






NM_016556

NP_057640


17
162427
UP
212697_at
NM_178126
206
NP_835227
957


17
2145
UP
203249_at
NM_001991
207
NP_001982
958


17
2145
UP
32259_at
NM_001991
208
NP_001982
959


17
28958
UP
218026_at
NM_014019
209
NP_054738
960


17
8678
UP
208945_s_at
NM_003766
210
NP_003757
961


17
10197
UP
200988_s_at
NM_005789;
211; 212
NP_005780;
962; 963






NM_176863

NP_789839


17
314
UP
207064_s_at
NM_001158;
213; 214
NP_001149;
964; 965






NM_009590

NP_0033720


17
80755
UP
222064_s_at
NM_025267
215
NP_079543
966


17
6155
UP
200025_s_at
NM_000988
216
NP_000979
967


17

UP
201383_s_at

1526


17
1845
UP
201537_s_at
NM_004090
217
NP_004081
968


17
4356
UP
206186_at
NM_001932
218
NP_001923
969


17
4355
UP
213270_at
NM_005374
219
NP_005365
970


17
92579
UP
221759_at
NM_138387
220
NP_612396
971


17
92579
UP
44654_at
NM_138387
221
NP_612396
972


17
79089
UP
218419_s_at
NM_024107;
222; 223
NP_077012;
973; 974






NM_17741

NP_803190


17
2896
UP
200678_x_at
NM_002087
224
NP_002078
975


17
23131
UP
212485_at
XM_290758
225
XP_290758
976


17
4836
UP
201157_s_at
NM_021079
226
NP_066565
977


17
10614
UP
214188_at
NM_006460
227
NP_006451
978


17
55073
UP
220219_s_at
NM_018001
228
NP_060471
979


17
51326
UP
210718_s_at
NM_016632
229
NP_057716
980


17
9884
UP
221740_x_at
NM_014834
230
NP_055649
981


17
4905
UP
202395_at
NM_006178
231
NP_006169
982


17
7473; 7473
UP
221455_s_at
NM_030753
232
NP_110380
983


17

UP
217880_at

1527




17
9520
UP
201455_s_at
NM_006310
235
NP_006301
986


17
3837
UP
208974_x_at
NM_002265
236
NP_002256
987


17
55163
UP
218511_s_at
NM_018129
237
NP_060599
988


17
10951
UP
201518_at
NM_006807
238
NP_006798
989


17
29916
UP
220140_s_at
NM_013323;
239; 240
NP_037455;
990; 991






NM_152244

NP_689450


17
29916
UP
53912_at
NM_013323;
241; 242
NP_689450
992; 993






NM_152244

NP_689450


17
516
UP
208972_s_at
NM_005175
243
NP_005166
994


17
65264
UP
217750_s_at
NM_023079
244
NP_075567
995


17
11267
UP
218391_at
NM_007241
245
NP_009172
996


17
22834
UP
205594_at
XM_375471
246
XP_375471
997


17
5245
UP
200659_s_at
NM_002634
247
NP_002625
998


17
8405
UP
204640_s_at
NM_003563
248
NP_003554
999


17
10237
UP
202433_at
NM_005827
249
NP_005818
1000


17
81558;
UP
221249_s_at
NM_030802
250
NP_110429
1001



81558


17
11143
UP
200049_at
NM_007067
251
NP_008998
1002


17
3675
UP
201474_s_at
NM_002204;
252; 253
NP_002195;
1003; 1004






NM_005501

NP_005492


17
5164
UP
213724_s_at
NM_002611
254
NP_002602
1005


17
64132
UP
219401_at
NM_022167
255
NP_071450
1006


17
55379
UP
222231_s_at
NM_018509
256
NP_060979
1007


17
80221
UP
218844_at
NM_025149
257
NP_079425
1008


17
55316
UP
218307_at
NM_018346
258
NP_060816
1009


17
55040
UP
220318_at
NM_017957
259
NP_060427
1010


17
64847
UP
218164_at
NM_022827
260
NP_073738
1011


17
91369;
UP
211717_at
NM_052855
261
NP_443087
1012



91369


17
51747
UP
220044_x_at
NM_016424
262
NP_057508
1013


17
51747
UP
203804_s_at
NM_016424
263
NP_057508
1014


17
9043
UP
206748_s_at
NM 003971,
264; 265
NP_003962;
1015; 1016






NM_172345

NP_758853


17
4830
UP
201577_at
NM_000269;
266; 267
NP_000260;
1017; 1018






NM_198175

NP_937818


17
4831
UP
201268_at
NM_002512
268
NP_002503
1019


17
51096
UP
222038_s_at
NM_016001
269
NP_057085
1020


17
1353
UP
214277_at
NM_004375
270
NP_004366
1021


17
3131
UP
204755_x_at
NM_002126
271
NP_002117
1022


17
58488
UP
218676_s_at
NM_021213
272
NP_067036
1023


17
9772
UP
202650_s_at
NM_014738
273
NP_055553
1024


17
57513
UP
221846_s_at
NM_020753
274
NP_065804
1025


17
57513
UP
61297_at
NM_020753
275
NP_065804
1026


18
8780
UP
202131_s_at
NM_003831;
276; 277
NP_003822;
1027; 1028






NM_145906

NP_665913


18
8780
UP
202129_s_at
NM_003831;
278; 279
NP_003822;
1029; 1030






NM_145906

NP_665913


18
29919
UP
221190_s_at
NM_013326
280
NP_037458
1031


18
4864
UP
202679_at
NM_000271
281
NP_000262
1032


18
26256
UP
219928_s_at
NM_012189;
282; 283; 284; 285;
NP_036321;
1033; 1034; 1035;






NM_138643;
286; 287
NP_619584;
1036; 1037; 1038






NM_138644;

NM_619585;






NM_153768;

NP_722452;






NM_153769;

NP_722453;






NM_153770

NP_722454


18
114876;
UP
208158_s_at
NM_018030;
288; 289; 290
NP_060500;
1039; 1040; 1041



114876


NM_080597;

NP_542164;






NM_133268

NP_579802


19
7705
UP
200050_at
NM_007145
291
NP_009076
1042


19
126231
UP
217627_at
NM_152360
292
NP_689573
1043


19
8193
UP
206531_at
NM_004647
293
NP_004638
1044


19
5714
UP
200820_at
NM_002812
294
NP_002803
1045


19
11184
UP
214219_x_at
NM_007181
295
NP_009112
1046


19
27335
UP
221494_x_at
NM_013234
296
NP_037366
1047


19
27335
UP
212716_s_at
NM_013234
297
NP_037366
1048


19
3191
UP
221860_at
NM_001533
298
NP_001524
1049


19
22933
UP
220605_s_at
NM_012237;
299; 300
NP_036369;
1050; 1051






NM_030593

NP_085096


19
4793
UP
214448_x_at
NM_002503
301
NP_002494
1052


19
54938
UP
218702_at
NM_017827
302
NP_060297
1053


19
6183
UP
210008_s_at
NM_021107;
303; 304; 305
NP_066930;
1054; 1055; 1056






NM_033362;

NP_203526;






NM_033363

NP_203527


19
115290
UP
220233_at
NM_024907;
306; 307
NP_079183;
1057; 1058






NM_148169

NP_680474


19
10298
UP
203154_s_at
NM_005884
308
NP_005875
1059


19
10298
UP
33814_at
NM_005884
309
NP_005875
1060


19
54623
UP
202093_s_at
NM_019088
310
NP_061961
1061


19
10848
UP
218849_s_at
NM_006663
311
NP_006654
1062


19
10849
UP
205264_at
NM_012099
312
NP_036231
1063


19
80207
UP
206357_at
NM_025136
313
NP_079412
1064


19
24139
UP
204399_s_at
NM_012155
314
NP_036287
1065


19
2696
UP
208105_at
NM_000164
315
NP_000155
1066


19
6633
UP
200826_at
NM_004597;
316; 317
NP_004588;
1067; 1068






NM_177542

NP_808210


19

UP
217661_x_at

1528


19
1760
UP
217062_at
NM_004409
318
NP_004400
1069


19
25865
UP
209282_at
NM_016457
320
NP_057541
1071


19
25865
UP
38269_at
NM_016457
321
NP_057541
1072


19
29888
UP
217903_at
NM_013403
322
NP_037535
1073


19
6510
UP
208916_at
NM_005628
323
NP_005619
1074


19
1175
UP
202120_x_at
NM_004069;
324; 325
NP_004060;
1075; 1976






NM_021575

NP_067586


19
1175; 1175
UP
211047_x_at
NM_004069;
326; 327
NP_004060;
1077; 1078






NM_021575

NP_067586


19
2909
UP
202046_s_at
NM_004491;
328; 329
NP_004482;
1079; 1080






NM_024342

NP_077318


19
728
UP
220088_at
NM_001736
330
NP_001727
1081


19
8775
UP
206491_s_at
NM_003827
331
NP_003818
1082


19
30846
UP
205341_at
NM_014601
332
NP_055416
1083


19
30846
UP
221870_at
NM_014601
333
NP_055416
1084


19
30846
UP
45297_at
NM_014601
334
NP_055416
1085


19
3978
UP
202726_at
NM_000234
335
NP_000225
1086


19
10945
UP
200922_at
NM_006801
336
NP_006792
1087


19
83743
UP
221549_at
NM_031485
337
NP_113673
1088


19
6141
UP
214335_at
NM_000979
338
NP_000970
1089


19
1628
UP
209783_at
NM_001352
339
NP_001343
1090


19
2523
UP
206109_at
NM_000148
340
NP_000139
1091


19
2523
UP
211411_at
NM_000148
341
NP_000139
1092


19
57664
UP
219011 _at
NM_020904
342
NP_065955
1093


19
4924
UP
200646_s_at
NM_006184
343
NP_006175
1094


19
581
UP
208478_s_at
NM_004324;
344; 345; 346; 347;
NP_004315;
1095; 1096; 1097;






NM_138761;
348; 349
NP_620116;
1098; 1099; 1100






NM_138762;

NP_620117;






NM_138763;

NP_620118;






NM_138764;

NP_620119;






NM_138765

NP_620120


19
2997
UP
201673_s_at
NM_002103
350
NP_002094
1101


19
10856
UP
201459_at
NM_006666
351
NP_006657
1102


19
3972
UP
214471_x_at
NM_000894
352
NP_000885
1103


19
6625
UP
201221_s_at
NM_003089
353
NP_003080
1104


19
27120
UP
220284_at
NM_014419
354
NP_055234
1105


19
23521
UP
200715_x_at
NM_012423
355
NP_036555
1106


19
57333
UP
219102_at
NM_020650
356
NP_065701
1107


19
51070
UP
217950_at
NM_015953
357
NP_057037
1108


19
3661
UP
202621_at
NM_001571
358
NP_001562
1109


19
3276
UP
206445_s_at
NM_001536:
359; 360; 361
NP_001527;
1110; 1111; 1112






NM_198318;

NP_938074;






NM_198319

NP_938075


19
53635
UP
213690_s_at
NM_017432
362
NP_059128
1113


19
23636
UP
202153_s_at
NM_012346;
363; 364; 365; 366
NP_036478;
1114; 1115; 1116;






NM_016553;

NP_057637;
1117






NM_153718:

NP_714940:






NM_153719

NP_714941


19
7376
UP
218215_s_at
NM_007121
367
NP_009052
1118


19
26085
UP
216670_at
NM_015596
368
NP_056411
1119


19
55422
UP
219228_at
NM_018555
369
NP_061025
1120


19
4849
UP
203239_s_at
NM_014516
371
NP_055331
1122


19
79143
UP
205634_x_at
NM_024298
372
NP_077274
1123


16
79042
UP
218132_s_at
NM_024075
373
NP_076980
1124


19
6203
UP
217747_s_at
NM_001013
374
NP_001004
1125


19
6861
UP
206161_s_at
NM_003180
375
NP_003171
1126


19
51157
UP
220748_s_at
NM_016202
376
NP_057286
1127


19
11338
UP
214171_s_at
NM_007279
377
NP_009210
1128


19
29924
UP
221141_x_at
NM_013333
378
NP_037465
1129


19
126208
UP
213402_at
XM_058999
379
XP_058999
1130


19
55311
UP
218707_at
NM_018337
380
NP_060807
1131


19
55311
UP
50376_at
NM_018337
381
NP_060807
1132


19
9423
UP
205917_at
XM_375660
382
XP_375660
1133


19
10794
UP
216273_at
NM_006635
383
NP_006626
1134


19
57343
UP
207753_at
NM_020657
384
NP_065708
1135


19
79744
UP
219826_at
NM_024691
385
NP_078967
1136


19
79744
UP
58367_s_at
NM_024691
386
NP_078967
1137


19
7693
UP
206182_at
NM_003435
387
NP_003426
1138


19
10520
UP
205437_at
NM_006385:
388; 389
NP_0063763
1139; 1140






NM_198855

NP_942152


20
3397
UP
208937_s_at
NM_002165;
390; 391
NP_002156;
1141, 1142






NM_181353

NP_851998


20
514
UP
217801_at
NM_006886
392
NP_008817
1143


20
51012
UP
217851_s_at
NM_016045
393
NP_057129
1144


20
5509
UP
204555_s_at
NM_006242
394
NP_006233
1145


20
6874
UP
213090_s_at
NM_003185
395
NP_003176
1146


20
26039
UP
213140_s_at
NM_015558;
396; 397
NP_056373;
1147; 1148






NM_198935

NP_945173


20
5688
UP
216088_s_at
NM_002792;
398; 399
NP_002783;
1149; 1150






NM_152255

NP_689468


20
9885
UP
209222_s_at
NM_014835;
400; 401
NP_055650;
1151; 1152






NM_144498

NP_653081


20
3911
UP
210150_s_at
NM_005560
402
NP_005551
1153


20
6227
UP
200834_s_at
NM_001024
403
NP_001015
1154


20
10732
UP
204849_at
NM_006602
404
NP_006593
1155


20
54994
UP
218448_at
NM_017896
405
NP_060366
1156


20
54915
UP
221741_s_at
NM_017798
406
NP_060268
1157


20
10139
UP
215984_s_at
NM_003224
407
NP_003215
1158


20
84619
UP
221848_at
NM_032527;
408; 409; 410
NP_115916;
1159; 1160; 1161






NM_181484;

NP_852149;






NM_181485

NP_852130


20
10139
UP
203174_s_at
NM_003224
411
NP_003215
1162


20
84619
UP
57539_at
NM_032527;
412; 413; 414
NP_115916;
1163; 1164; 1165






NM_181484;

NP_852149;






NM_181485

NP_852150


20
7165
UP
201379_s_at
NM_003288;
415; 416; 417; 418;
NP_003279;
1166; 1167; 1168;






NM_199359;
419; 420
NP_955391;
1169; 1170; 1171






NM_199360;

NP_955392;






NM_199361;

NP_955393;






NM_199362;

NP_955394;






NM_199363;

NP_955395


20
54963
UP
218533_s_at
NM_017859
421
NP_060329
1172


20
24148
UP
208879_x_at
NM_012469
422
NP_036601
1173


22
1652
UP
202929_s_at
NM_001355
423
NP_001346
1174


22
2952
UP
203815_at
NM_000853
424
NP_000844
1175


22
55000
UP
222244_s_at
NM_017903
425
NP_060373
1176


22
55000
UP
212337_at
NM_017903
426
NP_060373
1177








Claims
  • 1. A method of assessing whether a subject is afflicted with cancer or at risk for developing cancer, the method comprising comparing the copy number of a minimal common region (MCR) in a subject sample to the normal copy number of the MCR, wherein said MCR is selected from the group consisting of the MCRs listed in Table 1, and wherein an altered copy number of the MCR in the sample indicates that the subject is afflicted with cancer or at risk for developing cancer.
  • 2. The method of claim 1, wherein the copy number is assessed by fluorescent in situ hybridization (FISH).
  • 3. The method of claim 1, wherein the copy number is assessed by quantitative PCR (qPCR).
  • 4. The method of claim 1, wherein the normal copy number is obtained from a control sample.
  • 5. A method of assessing whether a subject is afflicted with cancer or at risk for developing cancer, the method comprising comparing: a) the amount, structure, and/or activity of a marker in a subject sample, wherein the marker is a marker which resides in an MCR listed in Table 1; andb) the normal amount, structure, and/or activity of the of the marker,wherein a significant difference between the amount, structure, and/or activity of the marker in the sample and the normal amount, structure, and/or activity is an indication that the subject is afflicted with cancer or at risk for developing cancer.
  • 6. The method of claim 5, wherein the marker is selected from the group consisting of the markers listed in Tables 4 or 5.
  • 7. The method of claim 5, wherein the amount of a marker is compared.
  • 8. The method of claim 5, wherein the structure of a marker is compared.
  • 9. The method of claim 5, wherein the activity of a marker is compared.
  • 10. The method of claim 7, wherein amount of the marker is determined by determining the level of expression of the marker.
  • 11. The method of claim 5, wherein amount of the marker is determined by determining copy number of the marker.
  • 12. The method of claim 5, wherein the normal amount/structure, and/or activity is obtained from a control sample.
  • 13. The method of claims 1 or 5, wherein the sample is selected from the group consisting of tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue.
  • 14. The method of claim 1 or 11, wherein the copy number is assessed by comparative genomic hybridization (CGH).
  • 15. The method of claim 14, wherein said CGH is performed on an array.
  • 16. The method of claim 10, wherein the level of expression of the marker in the sample is assessed by detecting the presence in the sample of a protein corresponding to the marker.
  • 17. The method of claim 16, wherein the presence of the protein is detected using a reagent which specifically binds with the protein.
  • 18. The method of claim 17, wherein the reagent is selected from the group consisting of an antibody, an antibody derivative, and an antibody fragment.
  • 19. The method of claim 10, wherein the level of expression of the marker in the sample is assessed by detecting the presence in the sample of a transcribed polynucleotide or portion thereof, wherein the transcribed polynucleotide comprises the marker.
  • 20. The method of claim 19, wherein the transcribed polynucleotide is an mRNA.
  • 21. The method of claim 19, wherein the transcribed polynucleotide is a cDNA.
  • 22. The method of claim 19, wherein the step of detecting further comprises amplifying the transcribed polynucleotide.
  • 23. The method of claim 10, wherein the level of expression of the marker in the sample is assessed by detecting the presence in the sample of a transcribed polynucleotide which anneals with the marker or anneals with a portion of a polynucleotide wherein the polynucleotide comprises the marker, under stringent hybridization conditions.
  • 24. A method for monitoring the progression of cancer in a subject, the method comprising: a) detecting in a subject sample at a first point in time, the amount and/or activity of a marker, wherein the marker is a marker which resides in an MCR listed in Table 1;b) repeating step a) at a subsequent point in time; andc) comparing the amount and/or activity detected in steps a) and b), and therefrom monitoring the progression of cancer in the subject.
  • 25. The method of claim 24, wherein the marker is selected from the group consisting of the markers listed in Tables 4 or 5.
  • 26. The method of claim 24, wherein the sample is selected from the group consisting of tissue, whole blood, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, bile, pancreatic juice, and pancreatic tissue.
  • 27. The method of claim 24, wherein the activity of a marker is determined.
  • 28. The method of claim 24, wherein the amount of a marker is determined.
  • 29. The method of claim 28, wherein amount of the marker is determined by determining the level of expression of the marker.
  • 30. The method of claim 28, wherein the level of expression of the marker in the sample is assessed by detecting the presence in the sample of a protein corresponding to the marker.
  • 31. The method of claim 30, wherein the presence of the protein is detected using a reagent which specifically binds with the protein.
  • 32. The method of claim 31, wherein the reagent is selected from the group consisting of an antibody, an antibody derivative, and an antibody fragment.
  • 33. The method of claim 29, wherein the level of expression of the marker in the sample is assessed by detecting the presence in the sample of a transcribed polynucleotide or portion thereof, wherein the transcribed polynucleotide comprises the marker.
  • 34. The method of claim 33, wherein the transcribed polynucleotide is an mRNA.
  • 35. The method of claim 33, wherein the transcribed polynucleotide is a cDNA.
  • 36. The method of claim 33, wherein the step of detecting further comprises amplifying the transcribed polynucleotide.
  • 37. The method of claim 29, wherein the level of expression of the marker in the sample is assessed by detecting the presence in the sample of a transcribed polynucleotide which anneals with the marker or anneals with a portion of a polynucleotide wherein the polynucleotide comprises the marker, under stringent hybridization conditions.
  • 38. The method of claim 24, wherein the sample comprises cells obtained from the subject.
  • 39. The method of claim 24, wherein between the first point in time and the subsequent point in time, the subject has undergone treatment for cancer, has completed treatment for cancer, and/or is in remission.
  • 40. A method of assessing the efficacy of a test compound for inhibiting cancer in a subject, the method comprising comparing: a) the amount and/or activity of a marker in a first sample obtained from the subject and maintained in the presence of the test compound, wherein the marker is a marker which resides in an MCR listed in Table 1; andb) the amount and/or activity of the marker in a second sample obtained from the subject and maintained in the absence of the test compound,wherein a significantly higher amount and/or activity of a marker in the first sample residing in an MCR which is deleted in cancer, relative to the second sample, is an indication that the test compound is efficacious for inhibiting cancer, and wherein a significantly lower amount and/or activity of a marker in the first sample residing in an MCR which is amplified in cancer, relative to the second sample, is an indication that the test compound is efficacious for inhibiting cancer in the subject.
  • 41. The method of claim 40, wherein the marker is selected from the group consisting of the markers listed in Tables 4 or 5.
  • 42. The method of claim 40, wherein the first and second samples are portions of a single sample obtained from the subject.
  • 43. The method of claim 40, wherein the first and second samples are portions of pooled samples obtained from the subject.
  • 44. A method of assessing the efficacy of a therapy for inhibiting cancer in a subject, the method comprising comparing: a) the amount and/or activity of a marker in the first sample obtained from the subject prior to providing at least a portion of the therapy to the subject, wherein the marker is a marker which resides in an MCR listed in Table 1, andb) the amount and/or activity of the marker in a second sample obtained from the subject following provision of the portion of the therapy,wherein a significantly higher amount and/or activity of the marker in the first sample residing in an MCR which is deleted in cancer, relative to the second sample, is an indication that the test compound is efficacious for inhibiting cancer and wherein a significantly lower amount and/or activity of the marker in the first sample residing in an MCR which is amplified in cancer, relative to the second sample, is an indication that the therapy is efficacious for inhibiting cancer in the subject.
  • 45. The method of claim 44, wherein the marker is selected from the group consisting of the markers listed in Tables 4 or 5.
  • 46. A method of selecting a composition capable of modulating cancer, the method comprising: a) obtaining a sample comprising cancer cells;b) contacting said cells with a test compound; andc) determining the ability of the test compound to modulate the amount and/or activity of a marker, wherein the marker is a marker which resides in an MCR listed in Table 1,thereby identifying a modulator of cancer.
  • 47. The method of claim 46, wherein the marker is selected from the group consisting of the markers listed in Tables 4 or 5.
  • 48. The method of claim 46, wherein said cells are isolated from an animal model of cancer.
  • 49. The method of claim 46, wherein said cells are from a cancer cell line.
  • 50. The method of claim 46, wherein said cells are from a subject suffering from cancer.
  • 51. The method of claim 49, wherein said cells are from a pancreatic cancer cell line originating from a pancreatic tumor.
  • 52. A method of selecting a composition capable of modulating cancer, the method comprising: a) contacting a marker which resides in an MCR listed in Table iwith a test is compound; andb) determining the ability of the test compound to modulate the amount and/or activity of a marker which resides in an MCR listed in Table 1,thereby identifying a composition capable of modulating cancer.
  • 53. The method of claim 52, wherein the marker is selected from the group consisting of the markers listed in Tables 4 or 5.
  • 54. The method of claim 46 or 52, further comprising administering the test compound to an animal model of cancer.
  • 55. The method of claim 46 or 52, wherein the modulator inhibits amount and/or activity of a gene or protein corresponding to a marker selected from the markers listed in Table 5.
  • 56. The method of claim 46 or 52, wherein the modulator increases the amount and/or activity of a gene or protein corresponding to a marker selected from the markers listed in Table 4.
  • 57. A kit for assessing the ability of a compound to inhibit cancer, the kit comprising a reagent for assessing the amount, structure, and/or activity of a marker which resides in an MCR listed in Table 1.
  • 58. A kit for assessing whether a subject is afflicted with cancer, the kit comprising a reagent for assessing the copy number of an MCR selected from the group consisting of the MCRs listed in Table 1.
  • 59. A kit for assessing whether a subject is afflicted with cancer, the kit comprising a reagent for assessing the amount, structure, and/or activity of a markerwhich resides in an MCR listed in Table 1.
  • 60. A kit for assessing the presence of human cancer cells, the kit comprising an antibody or fragment thereof, wherein the antibody or fragment thereof specifically binds with a protein corresponding to a marker which resides in an MCR listed in Table 1.
  • 61. A kit for assessing the presence of cancer cells, the kit comprising a nucleic acid probe wherein the probe specifically binds with a transcribed polynucleotide corresponding to a marker which resides in an MCR listed in Table 1.
  • 62. The kit of claim 61, wherein the nucleic acid probe is a molecular beacon probe.
  • 63. The kit of any one of claims 57, 59, 60, or 61, wherein the marker is selected from the group consisting of the markers listed in Tables 4 or 5.
  • 64. A method of treating a subject afflicted with cancer comprising administering to the subject a modulator of amount and/or activity of a gene or protein corresponding to a marker which resides in an MCR listed in Table 1, thereby treating a subject afflicted with cancer.
  • 65. The method of claim 64, wherein the marker is selected from the group consisting of the markers listed in Tables 4 or 5.
  • 66. A method of treating a subject afflicted with cancer comprising administering to the subject a compound which inhibits the amount and/or activity of a gene or protein corresponding to a marker which resides in an MCR listed in Table 1 which is amplified in cancer, thereby treating a subject afflicted with cancer.
  • 67. The method of claim 66, wherein said compound is administered in a pharmaceutically acceptable formulation.
  • 68. The method of claim 66, wherein said compound is an antibody or an antigen binding fragment thereof, which specifically binds to a protein corresponding to said marker.
  • 69. The method of claim 68, wherein said antibody is conjugated to a toxin.
  • 70. The method of claim 68, wherein said antibody is conjugated to a chemotherapeutic agent.
  • 71. The method of claim 66, wherein said compound is an RNA interfering agent which inhibits expression of a gene corresponding to said marker.
  • 72. The method of claim 71, wherein said RNA interfering agent is an siRNA molecule or an shRNA molecule.
  • 73. The method of claim 66, wherein said compound is an antisense oligonucleotide complementary to a gene corresponding to said marker.
  • 74. The method of claim 66, wherein said compound is a peptide or peptidomimetic.
  • 75. The method of claim 66, wherein said compound is a small molecule which inhibits activity of said marker.
  • 76. The method of claim 75, wherein said small molecule inhibits a protein-protein interaction between a marker and a target protein.
  • 77. The method of claim 66, wherein said compound is an aptamer which inhibits expression or activity of said marker.
  • 78. The method of claim 66, wherein the marker is selected from the group consisting of the markers listed in Table 5.
  • 79. A method of treating a subject afflicted with cancer comprising administering to the subject a compound which increases expression or activity of a gene or protein corresponding to a marker which resides in an MCR listed in Table 1 which is deleted in cancer, thereby treating a subject afflicted with cancer.
  • 80. A method of treating a subject afflicted with cancer comprising administering to the subject a protein corresponding to a marker which resides in an MCR listed in Table 1 which is deleted in cancer, thereby treating a subject afflicted with cancer.
  • 81. The method of any one of claims 79 or 80, wherein the marker is selected from the group consisting of the markers listed in Table 4.
  • 82. The method of claim 80, wherein the protein is provided to the cells of the subject, by a vector comprising a polynucleotide encoding the protein.
  • 83. The method of claim 79, wherein said compound is administered in a pharmaceutically acceptable formulation.
  • 84. An isolated protein, or fragment thereof, corresponding to a marker selected from the markers listed in Table 4 or Table 5.
  • 85. An isolated nucleic acid molecule, or fragment thereof, corresponding to a marker selected from the markers listed in Table 4 or Table 5.
  • 86. An isolated antibody, or fragment thereof, which specifically binds to a protein corresponding to a marker selected from the markers listed in Table 4 or Table 5.
  • 87. An isolated nucleic acid molecule, or fragment thereof, contained within an MCR selected from the MCRs listed in Table 1, wherein said nucleic acid molecule has an altered amount, structure, and/or activity in cancer.
  • 88. An isolated polypeptide encoded by the nucleic acid molecule of claim 87.
  • 89. A method for identifying a marker associated with cancer, said method comprising: a) performing profiling of the genome of cancer cells;b) performing segmentation analysis of profiles identified in step a);c) identifying loci;d) prioritizing said identified loci; ande) interrogating genes in the identified loci,to thereby identify a marker associated with cancer.
  • 90. A method for identifying a locus associated with cancer, said method comprising the steps of: a) performing profiling of the genome of cancer cells;b) performing segmentation analysis of profiles identified in step a);c) identifying loci; andd) prioritizing said identified loci,to thereby identify a locus associated with cancer.
  • 91. The method of claim 89, wherein said interrogation of genes in the identified loci is based on gene expression data.
  • 92. The method of claim 89, wherein said interrogation of genes in the identified loci is based on in vitro screening assays.
  • 93. The method of claim 89 or 90, wherein said profiling is performed using comparative genomic hybridization (CGH).
  • 94. The method of claim 89 or 90, wherein said cancer cells are derived from a cancer cell line or a tumor.
  • 95. A marker identified by the method of claim 89.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application No. 60/575,795, which was filed on May 28, 2004 and U.S. Provisional Application No. 60/580,337, which was filed on Jun. 15, 2004, both of which are hereby incorporated by refemce in there entirety.

GOVERNMENT FUNDING

Work described herein was supported, at least in part, by National Institutes of Health (NIH) under grant R01CA99041, R01CA86379, R01CA84628, and T32 CA09382. The government may therefore have certain rights to this invention.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US05/18850 5/27/2005 WO 00 6/16/2009
Provisional Applications (2)
Number Date Country
60575795 May 2004 US
60580337 Jun 2004 US