Grading of breast cancer

Information

  • Patent Application
  • 20030198972
  • Publication Number
    20030198972
  • Date Filed
    August 01, 2002
    22 years ago
  • Date Published
    October 23, 2003
    20 years ago
Abstract
Methods and compositions for the identification of breast cancer grade signatures are provided. The signature profiles are identified based upon multiple sampling of reference breast tissue samples from independent cases of breast cancer and provide a reliable set of molecular criteria for identification of cells as being in one or more particular stages and/or grades of breast cancer.
Description


FIELD OF THE INVENTION

[0002] The invention relates to the identification and use of gene expression profiles, or patterns, involved in breast cancer progression. In particular, the invention provides the identities of genes that may be used to identify different grades of breast cancer within and between stages thereof. The gene expression profiles, whether embodied in nucleic acid expression, protein expression, or other expression formats, are used in the study and/or diagnosis of cells and tissue during breast cancer progression as well as for the study and/or determination of prognosis of a patient. When used for diagnosis or prognosis, the profiles are used to predict the status and/or phenotype of cells and tissues relative to breast cancer and the treatment thereof.



BACKGROUND OF THE INVENTION

[0003] Breast cancer is by far the most common cancer among women. Each year, more than 180,000 and 1 million women in the U.S. and worldwide, respectively, are diagnosed with breast cancer. Breast cancer is the leading cause of death for women between ages 50-55, and is the most common non-preventable malignancy in women in the Western Hemisphere. An estimated 2,167,000 women in the United States are currently living with the disease (National Cancer Institute, Surveillance Epidemiology and End Results (NCI SEER) program, Cancer Statistics Review (CSR), www-seer.ims.nci.nih.gov/Publications/CSR1973 (1998)). Based on cancer rates from 1995 through 1997, a report from the National Cancer Institute (NCI) estimates that about 1 in 8 women in the United States (approximately 12.8 percent) will develop breast cancer during her lifetime (NCI's Surveillance, Epidemiology, and End Results Program (SEER) publication SEER Cancer Statistics Review 1973-1997). Breast cancer is the second most common form of cancer, after skin cancer, among women in the United States. An estimated 250,100 new cases of breast cancer are expected to be diagnosed in the United States in 2001. Of these, 192,200 new cases of more advanced (invasive) breast cancer are expected to occur among women (an increase of 5% over last year), 46,400 new cases of early stage (in situ) breast cancer are expected to occur among women (up 9% from last year), and about 1,500 new cases of breast cancer are expected to be diagnosed in men (Cancer Facts & Figures 2001 American Cancer Society). An estimated 40,600 deaths (40,300 women, 400 men) from breast cancer are expected in 2001. Breast cancer ranks second only to lung cancer among causes of cancer deaths in women. Nearly 86% of women who are diagnosed with breast cancer are likely to still be alive five years later, though 24% of them will die of breast cancer after 10 years, and nearly half (47%) will die of breast cancer after 20 years.


[0004] Every woman is at risk for breast cancer. Over 70 percent of breast cancers occur in women who have no identifiable risk factors other than age (U.S. General Accounting Office. Breast Cancer, 1971-1991: Prevention, Treatment and Research. GAO/PEMD-92-12; 1991). Only 5 to 10% of breast cancers are linked to a family history of breast cancer (Henderson I C, Breast Cancer. In: Murphy G P, Lawrence W L, Lenhard R E (eds). Clinical Oncology. Atlanta, Ga: American Cancer Society; 1995:198-219).


[0005] Each breast has 15 to 20 sections called lobes. Within each lobe are many smaller lobules. Lobules end in dozens of tiny bulbs that can produce milk. The lobes, lobules, and bulbs are all linked by thin tubes called ducts. These ducts lead to the nipple in the center of a dark area of skin called the areola. Fat surrounds the lobules and ducts. There are no muscles in the breast, but muscles lie under each breast and cover the ribs. Each breast also contains blood vessels and lymph vessels. The lymph vessels carry colorless fluid called lymph, and lead to the lymph nodes. Clusters of lymph nodes are found near the breast in the axilla (under the arm), above the collarbone, and in the chest.


[0006] Breast tumors can be either benign or malignant. Benign tumors are not cancerous, they do not spread to other parts of the body, and are not a threat to life. They can usually be removed, and in most cases, do not come back. Malignant tumors are cancerous, and can invade and damage nearby tissues and organs. Malignant tumor cells may metastasize, entering the bloodstream or lymphatic system. When breast cancer cells metastasize outside the breast, they are often found in the lymph nodes under the arm (axillary lymph nodes). If the cancer has reached these nodes, it means that cancer cells may have spread to other lymph nodes or other organs, such as bones, liver, or lungs.


[0007] Major and intensive research has been focussed on early detection, treatment and prevention. This has included an emphasis on determining the presence of precancerous or cancerous ductal epithelial cells. These cells are analyzed, for example, for cell morphology, for protein markers, for nucleic acid markers, for chromosomal abnormalities, for biochemical markers, and for other characteristic changes that would signal the presence of cancerous or precancerous cells. This has led to various molecular alterations that have been reported in breast cancer, few of which have been well characterized in human clinical breast specimens. Molecular alterations include presence/absence of estrogen and progesterone steroid receptors, HER-2 expression/amplification (Mark H F, et al. HER-2/neu gene amplification in stages I-IV breast cancer detected by fluorescent in situ hybridization. Genet Med; 1(3):98-103 1999), Ki-67 (an antigen that is present in all stages of the cell cycle except G0 and used as a marker for tumor cell proliferation, and prognostic markers (including oncogenes, tumor suppressor genes, and angiogenesis markers) like p53, p27, Cathepsin D, pS2, multi-drug resistance (MDR) gene, and CD31.


[0008] Examination of cells by a trained pathologist has also been used to establish whether ductal epithelial cells are normal (i.e. not precancerous or cancerous or having another noncancerous abnormality), precancerous (i.e. comprising hyperplasia, atypical ductal hyperplasia (ADH)) or cancerous (comprising ductal carcinoma in situ, or DCIS, which includes low grade ductal carcinoma in situ, or LG-DCIS, and high grade ductal carcinoma in situ, or HG-DCIS) or invasive (ductal) carcinoma (IDC). Pathologists may also identify the occurrence of lobular carcinoma in situ (LCIS) or invasive lobular carcinoma (ILC). Breast cancer progression may be viewed as the occurrence of abnormal cells, such as those of ADH, DCIS, IDC, LCIS, and/or ILC, among normal cells.


[0009] It remains unclear whether normal cells become hyperplastic (such as ADH) and then progressing on to become malignant (DCIS, IDC, LCIS, and/or ILC) or whether normal cells are able to directly become malignant without transitioning through a hyperplastic stage. It has been observed, however, that the presence of ADH indicates a higher likelihood of developing a malignancy. This has resulting in treatment of patients with ADH to begin treatment with an antineoplastic/antitumor agent such as tamoxifen. This is in contrast to the treatment of patients with malignant breast cancer which usually includes surgical removal.


[0010] The rational development of preventive, diagnostic and therapeutic strategies for women at risk for breast cancer would be aided by a molecular map of the tumorigenesis process. Relatively little is known of the molecular events that mediate the transition of normal breast cells to the various stages of breast cancer progression. Similarly, little is known of the molecular events that mediate the transition of cells from one stage of breast cancer to another.


[0011] Molecular means of identifying the differences between normal, non-cancerous cells and cancerous cells (in general) have been the focus of intense study. The use of cDNA libraries to analyze differences in gene expression patterns in normal versus tumorigenic cells has been described (U.S. Pat. No. 4,981,783). DeRisi et al. (1996) describe the analysis of gene expression patterns between two cell lines: UACC-903, which is a tumorigenic human melanoma cell line, and UACC-903(+6), which is a chromosome 6 suppressed non-tumorigenic form of UACC-903. Labeled cDNA probes made from mRNA from these cell lines were applied to DNA microarrays containing 870 different cDNAs and controls. Genes that were preferentially expressed in one of the two cell lines were identified.


[0012] Golub et al. (1999) describe the use of gene expression monitoring as means to cancer class discovery and class prediction between acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). Their approach to class predictors used a neighborhood analysis followed by cross-validation of the validity of the predictors by withholding one sample and building a predictor based only on the remaining samples. This predictor is then used to predict the class of the withheld sample. They also used cluster analysis to identify new classes (or subtypes) within the AML and ALL.


[0013] Gene expression patterns in human breast cancers have been described by Perou et al. (1999), who studied gene expression between cultured human mammary epithelia cells (HMEC) and breast tissue samples by use of microarrays comprising about 5000 genes. They used a clustering algorithm to identify patterns of expression in HMEC and tissue samples. Perou et al. (2000) describe the use of clustered gene expression profiles to classify subtypes of human breast tumors. Hedenfalk et al. describe gene expression profiles in BRCA1 mutation positive, BRCA2 mutation positive, and sporadic tumors. Using gene expression patterns to distinguish breast tumor subclasses and predict clinical implications is described by Sorlie et al. and West et al.


[0014] All of the above described approaches, however, utilize heterogeneous populations of cells found in culture or in a biopsy to obtain information on gene expression patterns. The use of such populations may result in the inclusion or exclusion of multiple genes from the patterns. For this and the lack of statistical robustness reasons, the gene expression patterns observed by the above described approaches provide little confidence that the differences in gene expression may be meaningfully associated with the stages of breast cancer.


[0015] Citation of documents herein is not intended as an admission that any is pertinent prior art. All statements as to the date or representation as to the contents of documents is based on the information available to the applicant and does not constitute any admission as to the correctness of the dates or contents of the documents.



SUMMARY OF THE INVENTION

[0016] The present invention relates to the identification and use of gene expression patterns (or profiles or “signatures”) which are correlated with (and thus able to discriminate between) cells in various stages and/or grades of breast cancer. Broadly defined, these stages are non-malignant versus malignant, but may also be viewed as normal versus atypical (optionally including reactive and pre-neoplastic) versus cancerous. Another definition of the stages is normal versus precancerous (e.g. atypical ductal hyperplasia (ADH) or atypical lobular hyperplasia (ALH)) versus cancerous (e.g. carcinoma in situ such as DCIS and/or LCIS) versus invasive (e.g. carcinomas such as IDC and/or ILC). The invention may also be applied to discriminations between normal and non-normal (including cancerous and other non-normal cells).


[0017] The invention also relates to the identification and use of gene expression patterns (or profiles or “signatures”) which are correlated with (and thus able to discriminate between) cells in various grades (within one or more stage) of breast cancer. Grading of breast cancer is normally done for cases of invasive ductal carcinoma (IDC), and may be done for invasive lobular carcinoma (ILC) as well, where cytological criteria such as the Nottingham BSR, nuclear morphology, tissue architecture, proliferation index (such as assays for PCNA or Ki67), and extent of differentiation are used to assign a grade of I, II or III to particular breast cancer samples. Grade I is usually where the cells are still well differentiated and are usually positive for the estrogen receptor (ER). Grade III is usually where the cells are poorly differentiated and usually negative for ER. Grade II is generally where the cells have characteristics intermediate between grades I and III and can make up approximately 60% of all samples assayed. This is rather unfortunate because determination of grade in IDC is used directly for decisions on patient care.


[0018] Grading of cases of ductal carcinoma in situ (DCIS) is also possible, but is not routine in current clinical practice. Grading of lobular carcinoma in situ (LCIS) is also possible. In addition to grades I to III, conventional grading schemes may use the terms “low grade” and/or “high grade”.


[0019] The present invention provides a non-subjective means for the identification of grades of various stages of cancer by assaying for the expression patterns associated with particular grades. Thus where subjective interpretation is used in grade assessment by pathologists using cytological criteria, the present invention provides objective gene expression patterns, which may optionally be performed in the absence of grading by histomorphological or cytological criteria, that are correlated with grades I-III (or low to high grade) to provide a more accurate assessment of breast cancer progression. The expression patterns of the invention thus provide a means to determine breast cancer prognosis. Furthermore, the expression patterns can also be used as a means to assay small, node negative tumors that are not readily graded by conventional means.


[0020] The gene expression patterns comprise one or more than one gene capable of discriminating between various stages and/or grades of breast cancer with significant accuracy. The gene(s) are identified as correlated with various stages and/or grades of breast cancer such that the levels of their expression are relevant to a determination of the stage and/or grade of breast cancer of a cell. Thus in one aspect, the invention provides a method to determine the stage and/or grade of breast cancer of a subject afflicted with, or suspected of having, breast cancer by assaying a cell containing sample from said subject for expression of one or more than one gene disclosed herein as correlated with one or more stages and/or grades of breast cancer.


[0021] Gene expression patterns of the invention are identified by analysis of gene expression in multiple samples of each stage and/or grade to be studied. The overall gene expression profile of a sample is obtained through quantifying the expression levels of mRNA corresponding to approximately 12000 genes. This overall profile is then analyzed to identify genes that are positively, or negatively, correlated, with a stage and/or grade of breast cancer. An expression profile of a subset of human genes may then be identified by the methods of the present invention as correlated with a particular stage and/or grade of breast cancer. The use of multiple samples increases the confidence which a gene may be believed to be correlated with a particular stage and/or grade. Without sufficient confidence, it remains unpredictable whether a particular gene is actually correlated with a stage and/or grade of breast cancer and also unpredictable whether a particular gene may be successfully used to identify the stage and/or grade of an unknown breast cancer cell sample.


[0022] A profile of genes that are highly correlated with one stage and/or grade relative to another may be used to assay an sample from a subject afflicted with, or suspected of having, breast cancer to identify the stage and/or grade of breast cancer to which the sample belongs. Such an assay may be used as part of a method to determine the therapeutic treatment for said subject based upon the stage(s) and/or grade(s) of breast cancer identified. The present invention thus also provides for the advantageous ability to determine grade of a stage of breast cancer in combination with stage information to provide more detailed information in diagnosing and treating breast cancer. This has not always been possible in the diagnosis and treatment of breast cancer using previous protocols, where it was often only possible to determine stage with grade being only occasionally determinable.


[0023] The correlated genes may be used singly with significant accuracy or in combination to increase the ability to accurately discriminate between various stages and/or grades of breast cancer. The present invention thus provides means for correlating a molecular expression phenotype with a physiological (cellular) stage or state. This correlation provides a way to molecularly diagnose and/or monitor a cell's status in comparison to different cancerous versus non-cancerous phenotypes as disclosed herein. Additional uses of the correlated gene(s) are in the classification of cells and tissues; determination of diagnosis and/or prognosis; and determination and/or alteration of therapy.


[0024] The ability to discriminate is conferred by the identification of expression of the individual genes as relevant and not by the form of the assay used to determine the actual level of expression. An assay may utilize any identifying feature of an identified individual gene as disclosed herein as long as the assay reflects, quantitatively or qualitatively, expression of the gene. Identifying features include, but are not limited to, unique nucleic acid sequences used to encode (DNA), or express (RNA), said gene or epitopes specific to, or activities of, a protein encoded by said gene. All that is required is the identity of the gene(s) necessary to discriminate between stages and/or grades of breast cancer and an appropriate cell containing sample for use in an expression assay.


[0025] In one aspect, the invention provides for the identification of the gene expression patterns by analyzing global, or near global, gene expression from single cells or homogenous cell populations which have been dissected away from, or otherwise isolated or purified from, contaminating cells beyond that possible by a simple biopsy. Because the expression of numerous genes fluctuate between cells from different patients as well as between cells from the same patient sample, multiple individual gene expression patterns are used as reference data to generate models which in turn permit the identification of individual gene(s) that are most highly correlated with particular breast cancer stages, and/or grades, and/or have the best the ability to discriminate cells of one stage and/or grade from another.


[0026] Use of the present invention has resulted in the identification of two major changes in gene expression, one of which is associated with the transition of normal breast cells to ADH (and persisting in a majority of DCIS and IDC cells), and the second is associated with tumor grade progression. The invention also provides the identification of a subset of genes that differ quantitatively in expression between DCIS and IDC cells.


[0027] In another aspect, the invention provides physical and methodological means for detecting the expression of gene(s) identified by the models generated by individual expression patterns. These means may be directed to assaying one or more aspect of the DNA template(s) underlying the expression of the gene(s), of the RNA used as an intermediate to express the gene(s), or of the proteinaceous product expressed by the gene(s).


[0028] In a further aspect, the gene(s) identified by a model as capable of discriminating between breast cancer stages and/or grades may be used to identify the cellular state of an unknown sample of cell(s) from the breast. Preferably, the sample is isolated via non-invasive means. The expression of said gene(s) in said unknown sample may be determined and compared to the expression of said gene(s) in reference data of gene expression patterns from the various stages and/or grades of breast cancer. Optionally, the comparison to reference samples may be by comparison to the model(s) constructed based on the reference samples.


[0029] One advantage provided by the present invention is that contaminating, non-breast cells (such as infiltrating lymphocytes or other immune system cells) are not present to possibly affect the genes identified or the subsequent analysis of gene expression to identify the status of suspected breast cancer cells. Such contamination is present where a biopsy is used to generate gene expression profiles.


[0030] While the present invention has been described mainly in the context of human breast cancer, it may be practiced in the context of breast cancer of any animal known to be potentially afflicted by breast cancer. Preferred animals for the application of the present invention are mammals, particularly those important to agricultural applications (such as, but not limited to, cattle, sheep, horses, and other “farm animals”) and for human companionship (such as, but not limited to, dogs and cats).







BRIEF DESCRIPTION OF THE FIGURES

[0031]
FIG. 1. Laser capture microdissection. Phenotypically normal breast epithelium and phenotypically abnormal epithelium from atypical ductal hyperplasia (ADH), ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC) from a single breast specimen (case 79) were captured from hematoxylin and eosin-stained sections (8 m in thickness). Panels A, B and C show the images of pre-capture, post-capture, and the captured epithelial compartments, respectively.


[0032]
FIGS. 2

a
and 2b. Expression profiles of breast cancer progression. 2a. Data matrix of 1940 genes by breast cancer samples of different pathological stages. Columns represent samples of tissues identified as ADH; grades I, II, or III of DCIS; and grades I, II, or III of IDC. Rows represent genes. Color scale shown at left bottom. Genes are ordered by hierarchical clustering, and samples are ordered by pathological stage and tumor grade. 2b. Examples of interesting clusters I, II and III.


[0033]
FIG. 3. Two-dimensional clustering of 62 samples and 200 genes correlated with tumor grade. Genes (columns) and samples (rows) were clustered independently using a hierarchical clustering algorithm. Red dots indicate ADH samples and green dots indicate grade II samples (DCIS or IDC). Three main clusters (down regulated, Grade III signature, and Grade I signature) are highlighted by color bars. See FIG. 2A for color scale.


[0034]
FIG. 4. Genes with increased expression in IDC relative to DCIS. Two dimensional clustering was applied to 1688 genes and 24 IDC samples and a portion of the data matrix is shown to highlight a cluster of genes with higher expression in IDC than its corresponding DCIS from the same patient. Expression values are expressed as log-ratios of expression in IDC to that in DCIS. Color scheme shown at left bottom.


[0035]
FIG. 5. Breast cancer progression model Breast cancer initiates within normal epithelium evolving into ADH, which progresses into grade I DCIS. A simultaneous 2-dimensional process drives tumor grade progression from I to II to III and stage progression from DCIS to IDC.







DETAILED DESCRIPTION OF THE SPECIFIC EMBODIMENTS

[0036] Definitions of Terms as Used Herein:


[0037] A gene expression “pattern” or “profile” or “signature” refers to the relative expression of a gene between two or more stages of breast cancer which is correlated with being able to distinguish between said stages.


[0038] A “gene” is a polynucleotide that encodes a discrete product, whether RNA or proteinaceous in nature. It is appreciated that more than one polynucleotide may be capable of encoding a discrete product. The term includes alleles and polymorphisms of a gene that encodes the same product, or a functionally associated (including gain, loss, or modulation of function) analog thereof, based upon chromosomal location and ability to recombine during normal mitosis.


[0039] A “stage” or “stages” (or equivalents thereof) of breast cancer refer to a physiologic state of a breast cell as defined by known cytological or histological (including immunohistology, histochemistry, and immunohistochemistry) procedures and are readily known to skilled in the art. Non-limiting examples include normal versus abnormal, non-cancerous versus cancerous, the different stages described herein (e.g. hyperplastic, carcinoma, and invasive), and grades within different stages (e.g. grades I, II, or III or the equivalents thereof within cancerous stages).


[0040] The terms “correlate” or “correlation” or equivalents thereof refer to an association between expression of one or more genes and a physiologic state of a breast cell to the exclusion of one or more other stages and/or identified by use of the methods as described herein. A gene may be expressed at higher or lower levels and still be correlated with one or more breast cancer stages.


[0041] A “polynucleotide” is a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. This term refers only to the primary structure of the molecule. Thus, this term includes double- and single-stranded DNA and RNA. It also includes known types of modifications including labels known in the art, methylation, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, and intemucleotide modifications such as uncharged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), as well as unmodified forms of the polynucleotide.


[0042] The term “amplify” is used in the broad sense to mean creating an amplification product can be made enzymatically with DNA or RNA polymerases. “Amplification,” as used herein, generally refers to the process of producing multiple copies of a desired sequence, particularly those of a sample. “Multiple copies” mean at least 2 copies. A “copy” does not necessarily mean perfect sequence complementarity or identity to the template sequence.


[0043] By corresponding is meant that a nucleic acid molecule shares a substantial amount of sequence identity with another nucleic acid molecule. Substantial amount means at least 95%, usually at least 98% and more usually at least 99%, and sequence identity is determined using the BLAST algorithm, as described in Altschul et al. (1990), J. Mol. Biol. 215:403-410 (using the published default setting, i.e. parameters w=4, t=17). Methods for amplifying mRNA are generally known in the art, and include reverse transcription PCR (RT-PCR) and those described in U.S. Patent Application (number to be assigned) entitled “Nucleic Acid Amplification” filed on Oct. 25, 2001 as attorney docket number 485772002900 as well as U.S. Provisional Patent Applications 60/298,847 (filed Jun. 15, 2001) and 60/257,801 (filed Dec. 22, 2000), all of which are hereby incorporated by reference in their entireties as if fully set forth. Another method which may be used is quantitative PCR (or Q-PCR). Alternatively, RNA may be directly labeled as the corresponding cDNA by methods known in the art.


[0044] A “microarray” is a linear or two-dimensional array of preferably discrete regions, each having a defined area, formed on the surface of a solid support such as, but not limited to, glass, plastic, or synthetic membrane. The density of the discrete regions on a microarray is determined by the total numbers of immobilized polynucleotides to be detected on the surface of a single solid phase support, preferably at least about 50/cm2, more preferably at least about 100/cm2, even more preferably at least about 500/cm2, but preferably below about 1,000/cm2. Preferably, the arrays contain less than about 500, about 1000, about 1500, about 2000, about 2500, or about 3000 immobilized polynucleotides in total. As used herein, a DNA microarray is an array of oligonucleotides or polynucleotides placed on a chip or other surfaces used to hybridize to amplified or cloned polynucleotides from a sample. Since the position of each particular group of primers in the array is known, the identities of a sample polynucleotides can be determined based on their binding to a particular position in the microarray.


[0045] Because the invention relies upon the identification of genes that are over- or under-expressed, one embodiment of the invention involves determining expression by hybridization of mRNA, or an amplified or cloned version thereof, of a sample cell to a polynucleotide that is unique to a particular gene sequence. Preferred polynucleotides of this type contain at least about 20, at least about 22, at least about 24, at least about 26, at least about 28, at least about 30, or at least about 32 consecutive basepairs of a gene sequence that is not found in other gene sequences. The term “about” as used in the previous sentence refers to an increase or decrease of 1 from the stated numerical value. Even more preferred are polynucleotides of at least or about 50, at least or about 100, at least about or 150, at least or about 200, at least or about 250, at least or about 300, at least or about 350, or at least or about 400 basepairs of a gene sequence that is not found in other gene sequences. The term “about” as used in the preceding sentence refers to an increase or decrease of 10% from the stated numerical value. Such polynucleotides may also be referred to as polynucleotide probes that are capable of hybridizing to sequences of the genes, or unique portions thereof, described herein. Preferably, the sequences are those of mRNA encoded by the genes, the corresponding cDNA to such mRNAs, and/or amplified versions of such sequences. In preferred embodiments of the invention, the polynucleotide probes are immobilized on an array, other devices, or in individual spots that localize the probes.


[0046] Alternatively, and in another embodiment of the invention, gene expression may be determined by analysis of expressed protein in a cell sample of interest by use of one or more antibodies specific for one or more epitopes of individual gene products (proteins) in said cell sample. Such antibodies are preferably labeled to permit their easy detection after binding to the gene product.


[0047] The term “label” refers to a composition capable of producing a detectable signal indicative of the presence of the labeled molecule. Suitable labels include radioisotopes, nucleotide chromophores, enzymes, substrates, fluorescent molecules, chemiluminescent moieties, magnetic particles, bioluminescent moieties, and the like. As such, a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.


[0048] The term “support” refers to conventional supports such as beads, particles, dipsticks, fibers, filters, membranes and silane or silicate supports such as glass slides.


[0049] As used herein, a “breast tissue sample” or “breast cell sample” refers to a sample of breast tissue or fluid isolated from an individual suspected of being afflicted with, or at risk of developing, breast cancer. Such samples are primary isolates (in contrast to cultured cells) and may be collected by any non-invasive means, including, but not limited to, ductal lavage, fine needle aspiration, needle biopsy, the devices and methods described in U.S. Pat. No. 6,328,709, or any other suitable means recognized in the art. Alternatively, the “sample” may be collected by an invasive method, including, but not limited to, surgical biopsy.


[0050] “Expression” and “gene expression” include transcription and/or translation of nucleic acid material.


[0051] As used herein, the term “comprising” and its cognates are used in their inclusive sense; that is, equivalent to the term “including” and its corresponding cognates.


[0052] Conditions that “allow” an event to occur or conditions that are “suitable” for an event to occur, such as hybridization, strand extension, and the like, or “suitable” conditions are conditions that do not prevent such events from occurring. Thus, these conditions permit, enhance, facilitate, and/or are conducive to the event. Such conditions, known in the art and described herein, depend upon, for example, the nature of the nucleotide sequence, temperature, and buffer conditions. These conditions also depend on what event is desired, such as hybridization, cleavage, strand extension or transcription.


[0053] Sequence “mutation,” as used herein, refers to any sequence alteration in the sequence of a gene disclosed herein interest in comparison to a reference sequence. A sequence mutation includes single nucleotide changes, or alterations of more than one nucleotide in a sequence, due to mechanisms such as substitution, deletion or insertion. Single nucleotide polymorphism (SNP) is also a sequence mutation as used herein. Because the present invention is based on the relative level of gene expression, mutations in non-coding regions of genes as disclosed herein may also be assayed in the practice of the invention.


[0054] “Detection” includes any means of detecting, including direct and indirect detection of gene expression and changes therein. For example, “detectably less” products may be observed directly or indirectly, and the term indicates any reduction (including the absence of detectable signal). Similarly, “detectably more” product means any increase, whether observed directly or indirectly.


[0055] Unless defined otherwise all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs.


[0056] Specific Embodiments


[0057] The present invention relates to the identification and use of gene expression patterns (or profiles or “signatures”) which discriminate between (or are correlated with) cells in various stages and/or grades of breast cancer. Such patterns may be determined by the methods of the invention by use of a number of reference cell or tissue samples, such as those reviewed by a pathologist of ordinary skill in the pathology of breast cancer, which reflect various stages and/or grades of breast cancer. Because the overall gene expression profile differs from person to person, cancer to cancer, and cancer cell to cancer cell, correlations between certain cell states and genes expressed or underexpressed may be made as disclosed herein to identify genes that are capable of discriminating between different breast cancer states.


[0058] The present invention may be practiced with any number of genes believed, or likely to be, differentially expressed in breast cancer cells. Approximately 12,000 genes were used to identify hundreds of genes capable of discriminating between various stages and/or grades of breast cancer as shown in the following Examples. The identification may be made by using expression profiles of various homogenous normal and breast cancer cell populations, which were isolated by microdissection, such as, but not limited to, laser capture microdissection (LCM) of 100-1000 cells. Each gene of the expression profile may be assigned weights based on its ability to discriminate between two or more stages and/or grades of breast cancer. The magnitude of each assigned weight indicates the extent of difference in expression between the two groups and is an approximation of the ability of expression of the gene to discriminate between the two groups (and thus stages and/or grades). The magnitude of each assigned weight also approximates the extent of correlation between expression of individual gene(s) and particular breast cancer stages and/or grades.


[0059] It should be noted that merely high levels of expression in cells from a particular stage or grade does not necessarily mean that a gene will be identified as having a high absolute weight value.


[0060] Genes with top ranking weights (in absolute terms) may be used to generate models of gene expressions that would maximally discriminate between the two groups. Alternatively, genes with top ranking weights (in absolute terms) may be used in combination with genes with lower weights without significant loss of ability to discriminate between groups. Such models may be generated by any appropriate means recognized in the art, including, but not limited to, cluster analysis, supported vector machines, neural networks or other algorithm known in the art. The models are capable of predicting the classification of a unknown sample based upon the expression of the genes used for discrimination in the models. “Leave one out” cross-validation may be used to test the performance of various models and to help identify weights (genes) that are uninformative or detrimental to the predictive ability of the models. Cross-validation may also be used to identify genes that enhance the predictive ability of the models.


[0061] The gene(s) identified as correlated with particular breast cancer stages and/or grades by the above models provide the ability to focus gene expression analysis to only those genes that contribute to the ability to identify a cell as being in a particular stage and/or grade of breast cancer relative to another stage or grade. The expression of other genes in a breast cancer cell would be relatively unable to provide information concerning, and thus assist in the discrimination of, different stages of breast cancer. For example, the cysteine-rich protein 1 (intestinal), identified by I.M.A.G.E. Consortium CloneID 1323448 (“The I.M.A.G.E. Consortium: An Integrated Molecular Analysis of Genomes and their Expression,” Lennon et al., 1996, Genomics 33:151-152; see also image.llnl.gov) has been found to be useful in discriminations between normal and ADH cells (with persistence through DCIS and IDC). Thus expression of this gene would be utilized in models to discriminate between the above listed stages but not for discerning between other stages. This type of analysis is readily incorporated into algorithms used to generate models with reference gene expression data.


[0062] As will be appreciated by those skilled in the art, the models are highly useful with even a small set of reference gene expression data and can become increasingly accurate with the inclusion of more reference data although the incremental increase in accuracy will likely diminish with each additional datum. The preparation of additional reference gene expression data using genes identified and disclosed herein for discriminating between different stages and/or grades of breast cancer is routine and may be readily performed by the skilled artisan to permit the generation of models as described above to predict the status of an unknown sample based upon the expression levels of those genes.


[0063] To determine the (increased or decreased) expression levels of genes in the practice of the present invention, any method known in the art may be utilized. In one preferred embodiment of the invention, expression based on detection of RNA which hybridizes to the genes identified and disclosed herein is used. This is readily performed by any RNA detection or amplification+detection method known or recognized as equivalent in the art such as, but not limited to, reverse transcription-PCR, the methods disclosed in U.S. Patent Application (number to be assigned) entitled “Nucleic Acid Amplification” filed on Oct. 25, 2001 as attorney docket number 485772002900 as well as U.S. Provisional Patent Applications 60/298,847 (filed Jun. 15, 2001) and 60/257,801 (filed Dec. 22, 2000), and methods to detect the presence, or absence, of RNA stabilizing or destabilizing sequences.


[0064] Alternatively, expression based on detection of DNA status may be used. Detection of the DNA of an identified gene as methylated or deleted may be used for genes that have decreased expression in correlation with a particular breast cancer stage and/or grade. This may be readily performed by PCR based methods known in the art, including, but not limited to, Q-PCR. Conversely, detection of the DNA of an identified gene as amplified may be used for genes that have increased expression in correlation with a particular breast cancer stage and/or grade. This may be readily performed by PCR based, fluorescent in situ hybridization (FISH) and chromosome in situ hybridization (CISH) methods known in the art.


[0065] Expression based on detection of a presence, increase, or decrease in protein levels or activity may also be used. Detection may be performed by any immunohistochemistry (IHC) based, blood based (especially for secreted proteins), antibody (including autoantibodies against the protein) based, ex foliate cell (from the cancer) based, mass spectroscopy based, and image (including used of labeled ligand) based method known in the art and recognized as appropriate for the detection of the protein. Antibody and image based methods are additionally useful for the localization of tumors after determination of cancer by use of cells obtained by a non-invasive procedure (such as ductal lavage or fine needle aspiration), where the source of the cancerous cells is not known. A labeled antibody or ligand may be used to localize the carcinoma(s) within a patient.


[0066] A preferred embodiment using a nucleic acid based assay to determine expression is by immobilization of one or more of the genes identified herein on a solid support, including, but not limited to, a solid substrate as an array or to beads or bead based technology as known in the art. Alternatively, solution based expression assays known in the art may also be used. The immobilized gene(s) may be in the form of polynucleotides that are unique or otherwise specific to the gene(s) such that the polynucleotide would be capable of hybridizing to a DNA or RNA corresponding to the gene(s). These polynucleotides may be the full length of the gene(s) or be short sequences of the genes (up to one nucleotide shorter than the full length sequence known in the art by deletion from the 5′ or 3′ end of the sequence) that are optionally minimally interrupted (such as by mismatches or inserted non-complementary basepairs) such that hybridization with a DNA or RNA corresponding to the gene(s) is not affected.


[0067] The immobilized gene(s) may be used to determine the state of nucleic acid samples prepared from sample breast cell(s) for which the pre-cancer or cancer status is not known or for confirmation of a status that is already assigned to the sample breast cell(s). Without limiting the invention, such a cell may be from a patient suspected of being afflicted with, or at risk of developing, breast cancer. The immobilized polynucleotide(s) need only be sufficient to specifically hybridize to the corresponding nucleic acid molecules derived from the sample. While even a single correlated gene sequence may to able to provide adequate accuracy in discriminating between two breast cancer cell stages and/or grades, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, or eleven or more of the genes identified herein may be used as a subset capable of discriminating may be used in combination to increase the accuracy of the method. The invention specifically contemplates the selection of more than one, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, or eleven or more of the genes disclosed in the tables and figures herein for use as a subset in the identification of whether an unknown or suspicious breast cancer sample is normal or is in one or more stages and/or grades of breast cancer. Optionally, the genes used will not include CloneID 809507, which is also known as GenBank accession number AA454563, described as an EST with high similarity to CD63 but of unknown function.


[0068] In embodiments where only one or a few genes are to be analyzed, the nucleic acid derived from the sample breast cancer cell(s) may be preferentially amplified by use of appropriate primers such that only the genes to be analyzed are amplified to reduce contaminating background signals from other genes expressed in the breast cell. Alternatively, and where multiple genes are to be analyzed or where very few cells (or one cell) is used, the nucleic acid from the sample may be globally amplified before hybridization to the immobilized polynucleotides. Of course RNA, or the cDNA counterpart thereof may be directly labeled and used, without amplification, by methods known in the art.


[0069] The above assay embodiments may be used in a number of different ways to identify or detect the breast cancer stage and/or grade, if any, of a breast cancer cell sample from a patient. In many cases, this would reflect a secondary screen for the patient, who may have already undergone mammography or physical exam as a primary screen. If positive, the subsequent needle biopsy, ductal lavage, fine needle aspiration, or other analogous methods may provide the sample for use in the above assay embodiments. The present invention is particularly useful in combination with non-invasive protocols, such as ductal lavage or fine needle aspiration, to prepare a breast cell sample. The current analysis of ductal lavage samples is by cytological examination by a trained pathologist who classifies the samples in terms that are at least partly subjective: unsatisfactory (too few cells), benign (including fibrocystic change), atypical (or mild atypia), suspicious (or marked atypia), or malignant.


[0070] The present invention provides a more objective set of criteria, in the form of gene expression profiles of a discrete set of genes, to discriminate (or delineate) between meaningful stages and/or grades (or classes) of breast cancer cells. In particularly preferred embodiments of the invention, the assays are used to discriminate between the three grades (I, II, III) of carcinomas in situ as well as invasive carcinomas. With the use of alternative algorithms, such as neural networks, comparisons that discriminate between multiple (more than pairwise) classes may also be performed.


[0071] In one embodiment of the invention, the isolation and analysis of a breast cancer cell sample may be performed as follows:


[0072] (1) Ductal lavage or other non-invasive procedure is performed on a patient to obtain a sample.


[0073] (2) Sample is prepared and coated onto a microscope slide. Note that ductal lavage results in clusters of cells that are cytologically examined as stated above.


[0074] (3) Pathologist or image analysis software scans the sample for the presence of non-normal and/or atypical cells.


[0075] (4) If non-normal and/or atypical cells are observed, those cells are harvested (e.g. by microdissection such as LCM).


[0076] (5) RNA is extracted from the harvested cells.


[0077] (6) RNA is purified, amplified, and labeled.


[0078] (7) Labeled nucleic acid is contacted with a microarray containing polynucleotides of the genes identified herein as correlated to discriminations between two or more stages of breast cancer under hybridization conditions, then processed and scanned to obtain a pattern of intensities of each spot (relative to a control for general gene expression in cells) which determine the level of expression of the gene(s) in the cells.


[0079] (8) The pattern of intensities is analyzed by comparison to the expression patterns of the genes in known samples of normal and breast cancer cells (relative to the same control).


[0080] A specific example of the above method would be performing ductal lavage following a primary screen, observing and collecting non-normal and/or atypical cells for analysis. The comparison to known expression patterns, such as that made possible by a model generated by an algorithm (such as, but not limited to nearest neighbor type analysis, SVM, or neural networks) with reference gene expression data for the different breast cancer stages and/or grades, identifies the cells as being most likely grade III IDC.


[0081] Alternatively, the sample may permit the collection of both normal as well as non-normal and/or atypical cells for analysis. The gene expression patterns for each of these two samples will be compared to each other as well as the model and the normal versus individual abnormal comparisons therein based upon the reference data set. This approach can be significantly more powerful that the non-normal and/or atypical cells only approach because it utilizes significantly more information from the normal cells and the differences between normal and non-normal/atypical cells (in both the sample and reference data sets) to determine the status of the non-normal and/or atypical cells from the sample.


[0082] By appropriate selection of the genes used in the analysis, identification of the relative amounts of non-normal and/or atypical cells may also be possible, although in most clinical settings, the identification of the highest grade of breast cancer with confidence makes identification of lower grades less important. Stated differently, the identification of invasive cancer determines the clinical situation regardless of the presence of carcinoma in situ or hyperplastic cells, or the identification of carcinoma in situ makes determines the clinical situation regardless of the presence of hyperplastic cells. Similarly, the identification of a higher grade of cancer cells determines the clinical situation regardless of the presence of lower grades of cancer cells.


[0083] With use of the present invention, skilled physicians may prescribe treatments based on non-invasive samples that they would have prescribed for a patient which had previously received a diagnosis via a solid tissue biopsy.


[0084] The above discussion is also applicable where a palpable lesion is detected followed by fine needle aspiration or needle biopsy of cells from the breast. The cells are plated and reviewed by a pathologist or automated imaging system which selects cells for analysis as described above. This again provides a means of linking visual to molecular cytology and provides a less subjective means of identifying the physiological state of breast cancer cells without the need for invasive solid tissue biopsies.


[0085] The present invention may also be used, however, with solid tissue biopsies. For example, a solid biopsy may be collected and prepared for visualization followed by determination of expression of one or more genes identified herein to determine the stage of breast cancer, if any. One preferred means is by use of in situ hybridization with polynucleotide or protein identifying probe(s) for assaying expression of said gene(s).


[0086] In an alternative method, the solid tissue biopsy may be used to extract molecules followed by analysis for expression of one or more gene(s). This provides the possibility of leaving out the need for visualization and collection of only those cells suspected of being non-normal and/or atypical. This method may of course be modified such that only cells suspected of being non-normal and/or atypical are collected and used to extract molecules for analysis. This would require visualization and selection as an prerequisite to gene expression analysis.


[0087] In a further modification of the above, both normal cells and cells suspected of being non-normal and/or atypical are collected and used to extract molecules for analysis of gene expression. The approach, benefits and results are as described above using non-invasive sampling.


[0088] In a further alternative to all of the above, the gene(s) identified herein may be used as part of a simple PCR or array based assay simply to determine the presence of non-normal and/or atypical cells in a sample from a non-invasive sampling procedure. This is simple to perform and utilizes genes identified to be the best discriminators of normal versus abnormal cells without the need for any cytological examination. If no non-normal and/or atypical cells are identified, no cytological examination is necessary. If non-normal and/or atypical cells are identified, cytological examination follows, and a more comprehensive analysis, as described above, may follow.


[0089] The genes identified herein may be used to generate a model capable of predicting the breast cancer stage and/or grade (if any) of an unknown breast cell sample based on the expression of the identified genes in the sample. Such a model may be generated by any of the algorithms described herein or otherwise known in the art as well as those recognized as equivalent in the art using gene(s) (and subsets thereof) disclosed herein for the identification of whether an unknown or suspicious breast cancer sample is normal or is in one or more stages and/or grades of breast cancer. The model provides a means for comparing expression profiles of gene(s) of the subset from the sample against the profiles of reference data used to build the model. The model can compare the sample profile against each of the reference profiles or against model defining delineations made based upon the reference profiles. Additionally, relative values from the sample profile may be used in comparison with the model or reference profiles.


[0090] In a preferred embodiment of the invention, breast cell samples identified as normal and non-normal and/or atypical from the same subject may be analyzed for their expression profiles of the genes used to generate the model. This provides an advantageous means of identifying the stage of the abnormal sample based on relative differences from the expression profile of the normal sample. These differences can then be used in comparison to differences between normal and individual abnormal reference data which was also used to generate the model.


[0091] The detection of gene expression from the samples may be by use of a single microarray able to assay gene expression from all pairwise comparisons disclosed herein for convenience and accuracy.


[0092] Other uses of the present invention include providing the ability to identify breast cancer cell samples as being those of a particular stage and/or grade of cancer for further research or study. This provides a particular advantage in many contexts requiring the identification of breast cancer stage and/or grade based on objective genetic or molecular criteria rather than cytological observation. It is of particular utility to distinguish different grades of a particular breast cancer stage for further study, research or characterization because no objective criteria for such delineation was previously available.


[0093] The materials for use in the methods of the present invention are ideally suited for preparation of kits produced in accordance with well known procedures. The invention thus provides kits comprising agents for the detection of expression of the disclosed genes for identifying breast cancer stage. Such kits optionally comprising the agent with an identifying description or label or instructions relating to their use in the methods of the present invention, is provided. Such a kit may comprise containers, each with one or more of the various reagents (typically in concentrated form) utilized in the methods, including, for example, pre-fabricated microarrays, buffers, the appropriate nucleotide triphosphates (e.g., dATP, dCTP, dGTP and dTTP; or rATP, rCTP, rGTP and UTP), reverse transcriptase, DNA polymerase, RNA polymerase, and one or more primer complexes of the present invention (e.g., appropriate length poly(T) or random primers linked to a promoter reactive with the RNA polymerase). A set of instructions will also typically be included.


[0094] The methods provided by the present invention may also be automated in whole or in part. All aspects of the present invention may also be practiced such that they consist essentially of a subset of the disclosed genes to the exclusion of material irrelevant to the identification of breast cancer stages in a cell containing sample.


[0095] Gene Expression Profiles of Pathological Stage and Histological Grade Progression of Human Breast Cancer


[0096] To identify gene expression changes that occur during breast cancer progression, isolation via LCM phenotypically of abnormal epithelium from ADH, DCIS and IDC and phenotypically normal epithelium (henceforth referred to as normal) from 36 breast cancer patients and 3 healthy mammoplasty reduction patients (FIG. 1A and Table 1) was performed. The resulting 300 independently microdissected samples were used to interrogate a microarray containing approximately 12,000 human genes. Genes showing significant differences in the pair-wise comparisons of normal vs. ADH, normal vs. DCIS and normal vs. IDC were selected by linear discriminant analysis, resulting in a total of 1940 unique genes for further exploration.
1TABLE 1Patient and tumor characteristics of clinical samples in this studyCaseIDStages MicrodissectedAgeERPRHER2Nodea8DCIS (III), IDC (III)48PosPosPosPos14N, DCIS (I), IDC (I)44PosPosNDPos22ADH, DCIS (I)44NDNDNDPos25DCIS (I), IDC (II)81PosNegNDND30N, DCIS (III), IDC (III)47NegNegNegPos41N, DCIS (II), IDC (II)55PosPosNDNeg43N, DCIS (II), IDC (II)53PosNegNegPos44N, DCIS (III), IDC (III)28PosPosNegNeg45N, DCIS (I)36PosNegNegNegb57N, ADH, DCIS (I)34NDNDNDNeg65N, DCIS (III), IDC (III)39PosPosNegNeg78MPR4679N, ADH, DCIS (I), IDC54PosPosNegPos(I)88N, DCIS (III), IDC (III)35PosPosNDPos95MPR1696N, DCIS (III), IDC (III)31NegNegNegPos97DCIS (III), IDC (III)79NegNegPosPos102N, DCIS (I), IDC (I)55PosNegNegPos112N, DCIS (III), IDC (III)31NegPosNegPos121N, DCIS (II), IDC (II)45PosPosPosPos130N, DCIS (II), IDC (II)54PosPosNegPos131N, ADH, DCIS (II), IDC37PosPosPosPos(II)133N, DCIS (III), IDC (III)44NegNegPosPos148N, DCIS (II), IDC (II)42PosPosNegPosb152N, DCIS (III)55NDNDNDNeg153N, IDC (I)46PosPosPosPos169N, DCIS (II), IDC (II)34PosPosNegPos170N, DCIS (II), IDC (II)44PosPosPos-PosFISH173N, DCIS (I), IDC (I)52PosPosNegNeg178N, DCIS (III), IDC (III)43PosPosPosPos179N, DCIS (III), IDC (III)37NegNegPos-PosFISH180N, ADH, DCIS (I), IDC46PosPosNegPos(I)183N, DCIS (II)46NDNDNDPos191N, ADH, DCIS (II)43NDNDND193N, ADH, DCIS (I), IDC45PosPosNegPos(I)198N, DCIS (II), IDC (II)30PosPosNegNegb210N, ADH, DCIS (I)62NDNDNDNegb213N, ADH45NDNDNDNeg215MPR30aNodal status. Tumor grades indicated by roman numerals in parenthesis after the pathological stage of the specimen. Abbreviations used for pathological stages: N, normal; ADH, atypical ductal hyperplasia; DCIS, ductal carcinoma in situ; IDC, invasive ductal carcinoma; MPR, mammoplasty reduction. Abbreviations used for tumor marker status: ND, not determined; Pos, positive; Neg, negative; Pos-Fish, HER2-positivity by fluorescent in situ hybridization (FISH). bIndividuals with pre-invasive breast cancer only.


[0097] One important advantage of LCM is the ability to procure both normal and diseased cell populations from the same biopsy. Therefore, the expression level of each gene in a disease state (ADH or DCIS or IDC) is represented as the ratio to the patient-matched normal, which highlights differences due to disease state as opposed to the genetic background of a particular patient. Unsupervised hierarchical clustering of the 1940 genes based on the resulting data across all samples reveals two main clusters (See FIG. 2a). One cluster demonstrates increased expression in a majority of the diseased samples, and another cluster shows a relatively uniform decrease in expression across all samples. Importantly, most of these alterations (both increases and decreases) occur in the earliest stage of progression (ADH) and persist throughout later stages of DCIS and IDC. In addition, closer examination of this global view suggests that some of these genes increase their expression in DCIS/IDC of higher tumor grade. See Example II below.


[0098] Three example clusters of genes, further illustrate these points (FIG. 2b). Cluster I consists of genes whose expression levels increase in ADH and persist in a majority of DCIS and IDC samples. The gene CRIP1 is especially prominent and thus may be a potential biomarker for the detection of breast cancer including the pre-malignant stage of ADH. The genes of Cluster I along with their I.M.A.G.E. Consortium CloneID number and descriptive identifiers are listed in Table 2.
2TABLE 2IMAGE CloneIDDescription729975MGEA5 | meningioma expressed antigen 5 (hyaluronidase)241043Human clone 137308 mRNA, partial cds1556859ESTs, Weakly similar to I38022 hypothetical protein [H. sapiens]1911343RAB26 | RAB26, member RAS oncogene family589232FLJ11506 | hypothetical protein FLJ11506138189WFS1 | Wolfram syndrome 1 (wolframin)1323448CRIP1 | cysteine-rich protein 1 (intestinal)488202Homo sapiens cDNA FLJ31235 fis, clone KIDNE2004681, moderately similar toMus musculus peroxisomal long chain acyl-CoA thioesterase Ib (Pte1b) gene256619HSD17B7 | hydroxysteroid (17-beta) dehydrogenase 7810063GFER | growth factor, erv1 (S. cerevisiae)-like (augmenter of liver regeneration)824879MGC11275 | hypothetical protein MGC11275


[0099] Genes in cluster II display an expression pattern that correlate with tumor grade with the highest expression in grade III DCIS/IDC. Cluster II includes several genes important in the cell cycle (CENPA, HEC, UBE2C and PLK), and their elevated expression in grade III DCIS/IDC may reflect the higher proliferative index of high-grade tumors. The genes of Cluster II along with their I.M.A.G.E. Consortium CloneID number and descriptive identifiers are listed in Table 3.
3TABLE 3IMAGECloneIDDescription66406ESTs, Highly similar to T47163 hypothetical protein DKFZp762E1312.1 [H. sapiens]1517595KIAA0175 | likely ortholog of maternal embryonic leucine zipper kinase2017415CENPA | centromere protein A (17 kD)345787HEC | highly expressed in cancer, rich in leucine heptad repeats504308FLJ10540 | hypothetical protein FLJ10540769921UBE2C | ubiquitin-conjugating enzyme E2C128711ANLN | anillin (Drosophila Scraps homolog), actin binding protein744047PLK | polo (Drosophia)-like kinase128695Homo sapiens, Similar to RIKEN cDNA 1810054O13 gene, clone IMAGE: 3845933,mRNA, partial cds


[0100] Genes in cluster III demonstrate decreased expression in all three pathological stages. The epithelium-specific transcription factor ELF5 is noteworthy since it maps to chromosome 11 p13-15, a region subject to frequent loss of heterozygosity and rearrangement in multiple carcinoma including breast cancer (Zhou, J. et al. (1998a)). Therefore, loss of expression of ELF5 in ADH may be an important first step in the initiation of breast malignancy. Taken together, these results demonstrate that the normal to ADH transition is associated with extensive gene expression alterations and support the notion that ADH is a direct precursor to DCIS and IDC. The genes of Cluster III along with their I.M.A.G.E. Consortium CloneID number and descriptive identifiers are listed in Table 4.
4TABLE 4IMAGE CloneIDDescription768007CL683 | weakly similar to glutathione peroxidase 2877621| nGAP-like protein1570670IL22RA2 | class II cytokine receptor1881774KIAA1678 | KIAA16781686766| Rag D protein505864RGL | RalGDS-like gene1569187HS3ST4 | heparan sulfate (glucosamine) 3-O-sulfotransferase 4755881AQP5 | aquaporin 51864302ELF5 | E74-like factor 5 (ets domain transcriptionfactor)


[0101] To gain further insight into the observation that different histological grades may be associated with distinct gene expression signatures (FIG. 2b, cluster II), two sets of genes were identified. Each comprised 100 genes correlating with grade I and grade III samples respectively using discriminant analysis. Again, to cancel out potential differences in the absolute levels of expression among individuals, gene expression values were expressed as ratios of ADH, DCIS or IDC to the corresponding normal. Unsupervised two-dimensional clustering revealed three major gene clusters (FIG. 3). One cluster of genes demonstrated decreased expression in all samples with subtle quantitative differences between grade I and grade III (green bar). A second cluster of genes (denoted as the grade III signature) shows markedly increased expression in grade III samples (red bar), whereas a third cluster (grade I signature) demonstrates increased expression primarily in grade I samples (blue bar). The genes of “green bar” genes along with their I.M.A.G.E. Consortium CloneID number, chromosomal location and descriptive identifiers (if known) are listed in Table 5.
5TABLE 5Clone IDlocationDescription471196 2q37ITM3 | integral membrane protein 3796904 6q24-q25PLAGL1 | pleiomorphic adenoma gene-like 132493 2q31.1ITGA6 | “integrin, alpha 6”153470011q21KIAA0830 | KIAA0830 protein712139 2q37.2ARL7 | ADP-ribosylation factor-like 7291478 1p36RUNX3 | runt-related transcription factor 315089719p13.1B3GNT3 | “UDP-GlcNAc: betaGal beta-1,3-N-acetylglucosaminyltransferase 3”1653105 3p14-p12TSP50 | testes-specific protease 5066538416KIAA1609 | KIAA1609 protein84281816q23-q24KARS | lysyl-tRNA synthetase3767118q11.2FLJ21610 | hypothetical protein FLJ2161077330116q22.1CDH3 | “cadherin 3, type 1, P-cadherin (placental)”503671 6Homo sapiens cDNA FLJ14368 fis, clone HEMBA1001122317288311ESTs, Weakly similar to S24195 dopamine receptor D4 [H. sapiens]68489016p12.1FLJ20274 | hypothetical protein FLJ2027459384017q11.2DKFZP564K1964 | DKFZP564K1964 protein12145417p13.1ALOX12 | arachidonate 12-lipoxygenase197913 1p34.2SFPQ | splicing factor proline/glutamine rich (polypyrimidine tract-binding protein-associated)4309020q13.12H-L(3)MBT | lethal (3) malignant brain tumor l(3)mbt protein (Drosophila) homolog814826 2ESTs163506212q13.13DKFZP586A011 | DKFZP586A011 protein8148151601845 7q22-q31.1CAPRI | Ca2+-promoted Ras inactivator19005919p13.3GNG7 | “guanine nucleotide binding protein (G protein), gamma 7”27704419q13.32KIAA1183 | KIAA1183 protein1592530 3p21.31IP6K2 | mammalian inositol hexakisphosphate kinase 243123111q13EFEMP2 | EGF-containing fibulin-like extracellular matrix protein 226725417ESTs, Highly similar to LOX2_HUMAN ARACHIDONATE 12-LIPOXYGENASE[H. sapiens]4367910ESTs29557212q24.21KIAA0682 | KIAA0682 gene product4612912q13.1HDAC7A | histone deacetylase 7A1569077 6EST138242 1ESTs, Moderately similar to MAS2_human mannan-binding lectin serine protease 2precursor [H. sapiens]417637 4p16KIAA1276 | KIAA1276 protein248631 3p21.2-p21.1AMT | aminomethyltransferase (glycine cleavage system protein T)1553530 2KIAA0788 | KIAA0788 protein3070291883169 5p15.32FLJ20303 | hypothetical protein FLJ20303345764 3p23SATB1 | special AT-rich sequence binding protein 1 (binds to nuclear matrix/scaffold-associating DNA's)70396411q23INPPL1 | inositol polyphosphate phosphatase-like 170349Xq13.1MLLT7 | “myeloid/lymphoid or mixed-lineage leukemia (trithorax (Drosophila) homolog);translocated to, 7”186834915q11.2-q21.3PLA2G4B | “phospholipase A2, group IVB (cytosolic)”126466 1p34.1KIAA0467 | KIAA0467 protein1631682 1p32PPIE | peptidylprolyl isomerase E (cyclophilin E)17278319ZNF358 | zinc finger protein 358156687711q13C11orf2 | chromosome 11 open reading frame21630990 3p21.3-p21.2RPL29 | ribosomal protein L2928312419Homo sapiens, clone IMAGE: 3917549, mRNA, partial cds12641510Homo sapiens mRNA; cDNA DKFZp566H0124 (from clone DKFZp566H0124)34416810q23POLL | “polymerase (DNA directed), lambda”82363410ESTs325583EST81074117p13.2GABARAP | GABA(A) receptor-associated protein511831 3MGC12936 | hypothetical protein MGC12936180561 1p13.3GSTM1 | glutathione S-transferase M120621711p11.2NR1H3 | “nuclear receptor subfamily 1, group H, member 3”10866722q12.2SF3A1 | “splicing factor 3a, subunit 1, 120 kD”83979612p13.31LOC51147 | candidate tumor suppressor p33 ING1 homolog502518 3p21LAMB2 | “laminin, beta 2 (laminin S)”81098122q13FLJ20699 | hypothetical protein FLJ206991635059 9Homo sapiens, clone MGC: 16638 IMAGE: 4121964, mRNA, complete cds76717617p13.1TNFSF13 | “tumor necrosis factor (ligand) superfamily, member 13”81035817p13-p11ACADVL | “acyl-Coenzyme A dehydrogenase, very long chain”275771010p11.2ZNF37A | zinc finger protein 37a (KOX 21)1652259 7q31.3LKR/SDH | lysine-ketoglutarate reductase/saccharopine dehydrogenase


[0102] The genes of “red bar” genes along with their I.M.A.G.E. Consortium CloneID number, chromosomal location and descriptive identifiers (if known) are listed in Table 6.
6TABLE 6IMAGEChromosomalClone IDlocationDescription29372722q13.2MGC861 | hypothetical protein MGC861843121 6p22.1-p21.2CLIC1 | chloride intracellular channel 183968212q22UBE2N | ubiquitin-conjugating enzyme E2N (homologous to yeast UBC13)81550119p13.3MGC2721 | hypothetical protein MGC27211587847 2q21MCM6 | “minichromosome maintenance deficient (mis5, S. pombe) 6”1416055 8KIAA0165 | “extra spindle poles, S. cerevisiae, homolog of”201813112p13.2-p13.1RACGAP1 | Rac GTPase activating protein 1147605315q15.1RAD51 | RAD51 (S. cerevisiae) homolog (E coli RecA homolog)86937515q26.1IDH2 | “isocitrate dehydrogenase 2 (NADP+), mitochondrial”95124115q13.3ANKT | nucleolar protein ANKT74381012p13MGC2577 | hypothetical protein MGC2577292936 1p34.3FLJ10468 | hypothetical protein FLJ1046866406 2ESTs, Highly similar to T47163 hypothetical protein DKFZp762E1312.1 [H. sapiens]1517595 9p11.2KIAA0175 | likely ortholog of maternal embryonic leucine zipper kinase2017415 2p24-p21CENPA | centromere protein A (17 kD)34578718p11.31HEC | “highly expressed in cancer, rich in leucine heptad repeats”50430810cen-q26.11FLJ10540 | hypothetical protein FLJ1054076992120q13.12UBE2C | ubiquitin-conjugating enzyme E2C128711 7p15-p14ANLN | “anillin (Drosophila Scraps homolog), actin binding protein”74404716p12.3PLK | polo (Drosophia)-like kinase56498118Homo sapiens, Similar to RIKEN cDNA 2810433K01 gene, clone MGC: 10200 IMAGE: 3909951,mRNA, complete cds259950 8q23CML66 | chronic myelogenous leukemia tumor antigen 6682560610q24.1KNSL1 | kinesin-like 1814270 4q27PMSCL1 | polymyositis/scleroderma autoantigen 1 (75 kD)785368 8p21-p12TOPK | PDZ-binding kinase; T-cell originated protein kinase20906620q13.2-q13.3STK15 | serine/threonine kinase 15739450 1q21.2LASS2 | “longevity assurance(LAG1, S. cerevisiae) homolog 2”170274216q24.3SLC7A5 | “solute carrier family 7 (cationic amino acid transporter, y+ system), member 5”1631634 9q34.11MGC3038 | “hypothetical protein similar to actin related protein 2/3 complex, subunit 5”725454 9q22CKS2 | CDC28 protein kinase 282547017q21-q22TOP2A | topoisomerase (DNA) II alpha (170 kD)796469 1q32.1HSPC150 | HSPC150 protein similar to ubiquitin-conjugating enzyme705064 4p16.3TACC3 | “transforming, acidic coiled-coil containing protein 3”47156817q25HN1 | hematological and neurological expressed 1742707 7ESTs, Weakly similar to MUC2_HUMAN MUCIN 2 PRECURSOR [H. sapiens]624667 9q34.13LOC51117 | CGI-92 protein1422338 2p25-p24RRM2 | ribonucleotide reductase M2 polypeptide70079214q22CDKN3 | cyclin-dependent kinase inhibitor 3 (CDK2-associated dual specificity phosphatase)280375 8p22PRO2000 | PRO2000 protein122241 1p34.2PSMB2 | “proteasome (prosome, macropain) subunit, beta type, 2”2309073 2q33-q34FZD5 | frizzled (Drosophila) homolog 52322367 2p14-p13RTN4 | reticulon 479669417q25BIRC5 | baculoviral IAP repeat-containing 5 (survivin)74677Homo sapiens, Similar to RIKEN cDNA A430107J06 gene, clone MGC: 21416 IMAGE: 4452699,mRNA, complete cds82452417q21.32UGTREL1 | UDP-galactose transporter related825282DKFZP586L0724 | DKFZP586L0724 protein82496217q23.1-q23.3KPNA2 | “karyopherin alpha 2 (RAG cohort 1, importin alpha 1)”4283111q11-q12NTKL | N-terminal kinase-like814054 1q24-25KIAA0040 | KIAA0040 gene product205463520q13.33PSMA7 | “proteasome (prosome, macropain) subunit, alpha type, 7”21086217q24-17q25ACOX1 | “acyl-Coenzyme A oxidase 1, palmitoyl”897997Xp11.22-p11.21SMC1L1 | “SMC1 (structural maintenance of chromosomes 1, yeast)-like 1”76989014q13.1NP | nucleoside phosphorylase756595 1q21S100A10 | “S100 calcium-binding protein A10 (annexin II ligand, calpactin I, light polypeptide(p11))”951233 2q35PSMB3 | “proteasome (prosome, macropain) subunit, beta type, 3”529827Xp22.31SYAP1 | reserved1660666Xp21.1CA5B | “carbonic anhydrase VB, mitochondrial”169675713q22.2KIAA1165 | hypothetical protein KIAA1165361922 1p34ZMPSTE24 | “zinc metalloproteinase, STE24 (yeast, homolog)”823598PSMD12 | “proteasome (prosome, macropain) 26S subunit, non-ATPase, 12”772220 3q21.2PDIR | for protein disulfide isomerase-related703707 8q12.1ASPH | aspartate beta-hydroxylase7886920q13.33GP110 | “cell membrane glycoprotein, 110000M(r) (surface antigen)”147442417Homo sapiens cDNA FLJ31911 fis, clone NT2RP7004751194764717q23.3LOC51651 | CGI-147 protein89760912q23.2FLJ10074 | hypothetical protein FLJ10074753378 4q34.1FLJ22649 | hypothetical protein FLJ22649 similar to signal peptidase SPC22/2312433116CPSF5 | “cleavage and polyadenylation specific factor 5, 25 kD subunit”32750615Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 327506345538 9q21-q22CTSL | cathepsin L753320 8q13.3FLJ20533 | hypothetical protein FLJ20533823907 8q12.2FLJ10511 | hypothetical protein FLJ10511149355 8q13.1TRAM | translocating chain-associating membrane protein347373 8q13.3TCEB1 | “transcription elongation factor B (SIII), polypeptide 1 (15 kD, elongin C)”202894917q21.31PRO1855 | hypothetical protein PRO1855624627 2p25-p24RRM2 | ribonucleotide reductase M2 polypeptide731023 9q34WDR5 | WD repeat domain 578606720p13CDC25B | cell division cycle 25B878330 3Homo sapiens cDNA: FLJ22044 fis, clone HEP09141163113211q12.1PHT2 | peptide transporter 3756442 7q11.2POR | P450 (cytochrome) oxidoreductase823930 7q22.1ARPC1A | “actin related protein 2/3 complex, subunit 1A (41 kD)”268946 2Homo sapiens cDNA FLJ31861 fis, clone NT2RP70013191914863 2p13.3-p13.1DYSF | “dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive)”789012 3p25-p24FBLN2 | fibulin 2781047 2q14BUB1 | budding uninhibited by benzimidazoles 1 (yeast homolog)753428 8Homo sapiens, Similar to RIKEN cDNA 1110014B07 gene, clone MGC: 20766 IMAGE: 4586039,mRNA, complete cds


[0103] The genes of “blue bar” genes along with their I.M.A.G.E. Consortium CloneID number, chromosomal location and descriptive identifiers (if known) are listed in Table 7.
7TABLE 7IMAGEChromosomalClone IDlocationDescription28637819q13.4ZNF135 | zinc finger protein 135 (clone pHZ-17)854763 2q31.1MGC20702 | hypothetical protein MGC20702344959 4p16.2HSA250839 | gene for serine/threonine protein kinase27822218Homo sapiens, clone MGC: 10083 IMAGE: 3897118, mRNA, complete cds167997718Homo sapiens, clone MGC: 10083 IMAGE: 3897118, mRNA, complete cds50495911Homo sapiens mRNA; cDNA DKFZp586G0321 (from clone DKFZp586G0321)34218118q21.3BCL2 | B-cell CLL/lymphoma 250298819p13.3-p13.2ZNF20 | zinc finger protein 20 (KOX 13)590310 2Homo sapiens, clone MGC: 17393 IMAGE: 3914851, mRNA, complete cds18630111Homo sapiens cDNA FLJ12924 fis, clone NT2RP200470935712016Homo sapiens, clone IMAGE: 3538007, mRNA, partial cds20300316p13.3NME4 | “non-metastatic cells 4, protein expressed in”72564914q11.2NFATC4 | “nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 4”2014373 2q11.2HNK-1ST | HNK-1 sulfotransferase18344022q13.33ARSA | arylsulfatase A2014856 1q25.3HLALS | “major histocompatibility complex, class I-like sequence”25661910p11.2HSD17B7 | hydroxysteroid (17-beta) dehydrogenase 7768570 1q21.2FLJ11280 | hypothetical protein FLJ11280297566811p13RAG2 | recombination activating gene 2278430 2q23.3KIF5C | kinesin family member 5C1558233 3ESTs627248 5q23.2SBBI31 | SBBI31 protein151717110p15-p14IL2RA | “interleukin 2 receptor, alpha”1492468 1p32.3KIAA0452 | DEME-6 protein292770 1Homo sapiens, clone IMAGE: 3627860, mRNA, partial cds1456701 1q21BCL9 | B-cell CLL/lymphoma 974314618p11.21FLJ23403 | hypothetical protein FLJ234031557637 5ESTs1583198 5ESTs, Weakly similar to S65824 reverse transcriptase homolog [H. sapiens]741891 6p21.3RAB2L | “RAB2, member RAS oncogene family-like”179572 1Homo sapiens cDNA FLJ14227 fis, clone NT2RP3004095156990216p11.2KIAA0556 | KIAA0556 protein12764618ESTs, Weakly similar to T00365 hypothetical protein KIAA0670 [H. sapiens]782688 1p35.1P28 | “dynein, axonemal, light intermediate polypeptide”188363015KIAA1547 | KIAA1547 protein725340 4p16.3TETRAN | tetracycline transporter-like protein72689010q24.2MGC4643 | hypothetical protein MGC464382322 2p23.3RBSK | ribokinase839382 9Homo sapiens, Similar to RIKEN cDNA 1700017I11 gene, clone MGC: 26847 IMAGE: 4821517,mRNA, complete cds49630 3p14.3CACNA1D | “calcium channel, voltage-dependent, L type, alpha 1D subunit”32050 2Homo sapiens mRNA; cDNA DKFZp586P1124 (from clone DKFZp586P1124)110226TNFRSF10C | “tumor necrosis factor receptor superfamily, member 10c, decoy without anintracellular domain”1932725 1q32.1ZNF281 | zinc finger protein 28127972011Homo sapiens, Similar to RIKEN cDNA 1700008D07 gene, clone MGC: 9830 IMAGE: 3863323,mRNA, complete cds1733262 3p21.3BLu | BLu protein197903 1ESTs, Moderately similar to unnamed protein product [H. sapiens]155685917ESTs, Weakly similar to I38022 hypothetical protein [H. sapiens]72669916Homo sapiens, clone MGC: 9889 IMAGE: 3868330, mRNA, complete cds


[0104] Most striking is the existence of reciprocal gradients in the intensities of these two signatures from grade I to grade III with most grade II lesions exhibiting both signatures to varying degrees (e.g., cases 130, 169, 198). Interestingly, some grade II lesions show an expression pattern that is most similar to either grade I or grade III lesions (case 41 and 43, respectively), and some grade III samples also express the grade I signature (e.g., cases 65, 88 and 112). Histological grade is an important characteristic of breast cancer with proven utility in patient prognostication and treatment (Fitzgibbons, P. L. et al.). For example, tumors of grade III are more likely to recur and are more likely to respond to chemotherapy than those of grade I (Page, D. L. et al. (2001)). However, the current tumor grading system relies mainly on histomorphological criteria, which, although highly successful in differentiating grade I from grade III tumors, are inadequate to score grade II tumors consistently (Dalton, L. W. et al.). This difficulty may be explained by the existence of a transcriptional continuum from grade I to grade III as we observed here. Therefore, a gene expression-based molecular grading system may allow greater precision in classifying breast cancer and provide greater insight into the state of progression of a particular tumor.


[0105] An expanded set of 250 genes that display increased expression in Grade I samples in comparison to Grade III samples are identified in Table 8 by use of their I.M.A.G.E. Consortium CloneID numbers along with their chromosomal location and descriptive identifiers (if known) and relative weights.
8TABLE 8IMAGEChromosomeClone IDWeightLocationDescription3449591.451333 4p16.2HSA250839 | gene for serine/threonine protein kinase5049591.2868711Homo sapiens mRNA; cDNA DKFZp586G0321 (from clone DKFZp586G0321)8148151.24147431461.22181818p11.21FLJ23403 | hypothetical protein FLJ234034176371.208243 4p16KIAA1276 | KIAA1276 protein5029881.13396419p13.3-p13.2ZNF20 | zinc finger protein 20 (KOX 13)16799771.13133718Homo sapiens, clone MGC: 10083 IMAGE: 3897118, mRNA, complete cds3421811.1209818q21.3BCL2 | B-cell CLL/lymphoma 219327251.11409 1q32.1ZNF281 | zinc finger protein 281703491.110469Xq13.1MLLT7 | myeloid/lymphoid or mixed-lineage leukemia (trithorax (Drosophila)homolog); translocated to, 71805611.077508 1p13.3GSTM1 | glutathione S-transferase M11863011.06836911Homo sapiens cDNA FLJ12924 fis, clone NT2RP20047092782221.06564618Homo sapiens, clone MGC: 10083 IMAGE: 3897118, mRNA, complete cds3571201.06290216Homo sapiens, clone IMAGE: 3538007, mRNA, partial cds2486311.04971 3p21.2-p21.1AMT | aminomethyltransferase (glycine cleavage system protein T)430901.02185720q13.12H-L(3)MBT | lethal (3) malignant brain tumor I(3)mbt protein (Drosophila) homolog16316821.021091 1p32PPIE | peptidylprolyl isomerase E (cyclophilin E)7671761.00349517p13.1TNFSF13 | tumor necrosis factor (ligand) superfamily, member 133255831.00279EST18836300.97979515KIAA1547 | KIAA1547 protein320500.979642 2Homo sapiens mRNA; cDNA DKFZp586P1124 (from clone DKFZp586P1124)5025180.962484 3p21LAMB2 | laminin, beta 2 (laminin S)1264150.95706910Homo sapiens mRNA; cDNA DKFZp566H0124 (from clone DKFZp566H0124)823220.946458 2p23.3RBSK | ribokinase29756680.93673711p13RAG2 | recombination activating gene 215582330.931636 3ESTs2566190.92800210p11.2HSD17B7 | hydroxysteroid (17-beta) dehydrogenase 72062170.9279411p11.2NR1H3 | nuclear receptor subfamily 1, group H, member 37268900.92652610q24.2MGC4643 | hypothetical protein MGC464320143730.906969 2q11.2HNK-1ST | HNK-1 sulfotransferase2831240.8969519Homo sapiens, clone IMAGE: 3917549, mRNA, partial cds7418910.887613 6p21.3RAB2L | RAB2, member RAS oncogene family-like496300.885374 3p14.3CACNA1D | calcium channel, voltage-dependent, L type, alpha 1D subunit15925300.871817 3p21.31IP6K2 | mammalian inositol hexakisphosphate kinase 22770440.86833819q13.32KIAA1183 | KIAA1183 protein15668770.86733611q13C11orf2 | chromosome 11 open reading frame28397960.86722112p13.31LOC51147 | candidate tumor suppressor p33 ING1 homolog2797200.86486511Homo sapiens, Similar to RIKEN cDNA 1700008D07 gene, clone MGC:9830IMAGE: 3863323, mRNA, complete cds5118310.854961 3MGC12936 | hypothetical protein MGC1293620148560.849103 1q25.3HLALS | major histocompatibility complex, class I-like sequence16522590.845966 7q31.3LKR/SDH | lysine-ketoglutarate reductase/saccharopine dehydrogenase1727830.84404619ZNF358 | zinc finger protein 3582672540.83882317ESTs, Highly similar to LOX2_HUMAN ARACHIDONATE 12-LIPOXYGENASE[H. sapiens]7253400.826253 4p16.3TETRAN | tetracycline transporter-like protein5938400.8232717q11.2DKFZP564K1964 | DKFZP564K1964 protein1795720.819502 1Homo sapiens cDNA FLJ14227 fis, clone NT2RP30040958547630.818371 2q31.1MGC20702 | hypothetical protein MGC207022863780.81828819q13.4ZNF135 | zinc finger protein 135 (clone pHZ-17)17332620.815457 3p21.3BLu | BLu protein15171710.81248110p15-p14IL2RA | interleukin 2 receptor, alpha8148260.807648 2ESTs1264660.797965 1p34.1KIAA0467 | KIAA0467 protein1102260.796159TNFRSF10C | tumor necrosis factor receptor superfamily, member 10c, decoywithout an intracellular domain3441680.79575510q23POLL | polymerase (DNA directed), lambda1086670.7940222q12.2SF3A1 | splicing factor 3a, subunit 1, 120kD2955720.79203112q24.21KIAA0682 | KIAA0682 gene product8236340.78916410ESTs1382420.787686 1ESTs, Moderately similar to MAS2_HUMAN MANNAN-BINDING LECTIN SERINEPROTEASE 2 PRECURSOR [H. sapiens]1979030.785879 1ESTs, Moderately similar to unnamed protein product [H. sapiens]2927700.784314 1Homo sapiens, clone IMAGE: 3627860, mRNA, partial cds8109810.78411822q13FLJ20699 | hypothetical protein FLJ206991979130.777546 1p34.2SFPQ | splicing factor proline/glutamine rich (polypyrimidine tract-binding protein-associated)1900590.7747419p13.3GNG7 | guanine nucleotide binding protein (G protein), gamma 77826880.77051 1p35.1P28 | dynein, axonemal, light intermediate polypeptide1214540.7696717p13.1ALOX12 | arachidonate 12-lipoxygenase15699020.76421716p11.2KIAA0556 | KIAA0556 protein7266990.76073616Homo sapiens, clone MGC: 9889 IMAGE: 3868330, mRNA, complete cds16018450.759847 7q22-q31.1CAPRI | Ca2+-promoted Ras inactivator7039640.75962511q23INPPL1 | inositol polyphosphate phosphatase-like 11834400.75714822q13.33ARSA | arylsulfatase A4312310.75628111q13EFEMP2 | EGF-containing fibulin-like extracellular matrix protein 28103580.75031217p13-p11 ACADVL| acyl-Coenzyme A dehydrogenase, very long chain15831980.749857 5ESTs, Weakly similar to S65824 reverse transcriptase homolog [H. sapiens]16309900.748442 3p21.3-p21.2RPL29 | ribosomal protein L2918683490.74625715q11.2-q21.3PLA2G4B | phospholipase A2, group IVB (cytosolic)6272480.744679 5q23.2SBBI31 | SBBI31 protein1276460.74367218ESTs, Weakly similar to T00365 hypothetical protein KIAA0670 [H. sapiens]16350590.739062 9Homo sapiens, clone MGC: 16638 IMAGE: 4121964, mRNA, complete cds14567010.732349 1q21BCL9 | B-cell CLL/lymphoma 93457640.72889 3p23SATB1 | special AT-rich sequence binding protein 1 (binds to nuclear matrix/scaffold-associating DNA's)2784300.728595 2q23.3KIF5C | kinesin family member 5C14924680.72665 1p32.3KIAA0452 | DEME-6 protein5903100.725531 2Homo sapiens, clone MGC: 17393 IMAGE: 3914851, mRNA, complete cds7685700.720983 1q21.2FLJ11280 | hypothetical protein FLJ1128018831690.716948 5p15.32FLJ20303 | hypothetical protein FLJ2030316350620.71614212q13.13DKFZP586A011 | DKFZP586A011 protein27577100.71529410p11.2ZNF37A | zinc finger protein 37a (KOX 21)8107410.70903217p13.2GABARAP | GABA(A) receptor-associated protein15690770.708429 6EST16531050.708359 3p14-p12TSP50 | testes-specific protease 5015535300.707954 2KIAA0788 | KIAA0788 protein436790.70723510ESTs7256490.70682614q11.2NFATC4 | nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 46848900.70593416p12.1FLJ20274 | hypothetical protein FLJ2027415568590.70274617ESTs, Weakly similar to I38022 hypothetical protein [H. sapiens]15576370.698307 5ESTs2030030.69757316p13.3NME4 | non-metastatic cells 4, protein expressed in461290.69432112q13.1HDAC7A | histone deacetylase 7A8393820.693177 9Homo sapiens, Similar to RIKEN cDNA 1700017I11 gene, clone MGC:26847IMAGE: 4821517, mRNA, complete cds3070290.6902071840220.68976711p15APBB1 | amyloid beta (A4) precursor protein-binding, family B, member 1 (Fe65)7450770.68115319Homo sapiens mRNA; cDNA DKFZp566J2324 (from clone DKFZp566J2324); partialcds7696000.68017 5p15.2-p13.1UNG2 | uracil-DNA glycosylase 22807760.67782115MGC5139 | hypothetical protein MGC51398109470.67486116p13.11NUDE1 | LIS1-interacting protein NUDE1, rat homolog8248790.67470216p13.3MGC11275 | hypothetical protein MGC112754545030.66950212Homo sapiens, clone IMAGE: 3346451, mRNA, partial cds8119200.658971 9p13IL11RA | interleukin 11 receptor, alpha16363600.65896315q21.1-q21.2FLJ14957 | hypothetical protein FLJ1495725027220.65814611q23LOH11CR2A | loss of heterozygosity, 11, chromosomal region 2, gene A16093720.65729414q11.2RIPK3 | receptor-interacting serine-threonine kinase 33469770.655725 3p24.3KIAA0210 | KIAA0210 gene product2935690.653314 1q25C1orf21 | chromosome 1 open reading frame 2116353070.65174612Homo sapiens, clone IMAGE: 3833472, mRNA2405050.6517214q11.2KIAA0323 | KIAA0323 protein527240.648958FLJ20241 | hypothetical protein FLJ202411201380.64857910q21.1JDP1 | domain containing protein 1740700.648244 1q21.2ENSA | endosulfine alpha1866260.644915 6ESTs, Weakly similar to CYP4_HUMAN 40 KDA PEPTIDYL-PROLYL CIS-TRANSISOMERASE [H. sapiens]2966790.644155 5Homo sapiens clone TCCCTA00151 mRNA sequence21198380.6436811q25ADAMTS8 | a disintegrin-like and metalloprotease (reprolysin type) withthrombospondin type 1 motif, 88134880.643211 1q32.1LOC51235 | hypothetical protein7420940.63985714q32.12FLJ20950 | hypothetical protein FLJ209507052740.638802 2q37.2DGKD | diacylglycerol kinase, delta (130 kD)8262850.633833Homo sapiens cDNA FLJ32001 fis, clone NT2RP70093733582170.631361Xq26.1GPC4 | glypican 47967230.629143Homo sapiens clone CDABP0014 mRNA sequence5298430.62843519ESTs2622510.62773616p13CLCN7 | chloride channel 74904490.623346 5q31RAD50 | RAD50 (S. cerevisiae) homolog7883340.62290911p15.5-p15.4MRPL23 | mitochondrial ribosomal protein L2319099350.62043 8ESTs2508830.61921 3p21UBE1L | ubiquitin-activating enzyme E1-like17076670.61802317Homo sapiens cDNA FLJ31065 fis, clone HSYRA2001142681030.61786912MLC1SA | myosin light chain 1 slow a7733810.61726319q13.33NAPA | N-ethylmaleimide-sensitive factor attachment protein, alpha15595960.61677611ESTs, Highly similar to AF175283 1 zinc metalloendopeptidase [H. sapiens]8252960.616769 1q42.11-q42.3LDLC | low density lipoprotein receptor defect C complementing8668660.616211 3p21.3RASSF1 | Ras association (RalGDS/AF-6) domain family 14906680.613699 3Homo sapiens, clone IMAGE: 4182947, mRNA8240520.613652 6p21.3C6orf1 | chromosome 6 open reading frame 15052430.61267112p11ITPR2 | inositol 1,4,5-triphosphate receptor, type 219113430.61238716p13.3RAB26 | RAB26, member RAS oncogene family16372960.6061210q22-q23RPS24 | ribosomal protein S247532520.60429217q21.31MGC4251 | hypothetical protein MGC425115188900.60261211q13.2-q13.3MTL5 | metallothionein-like 5, testis-specific (tesmin)2345220.601183 1q21.3KIAA1535 | KIAA1535 protein524190.598962 9q13-q21X123 | Friedreich ataxia region gene X1232784830.59847518p11.32TYMS | thymidylate synthetase8776640.59824320FLJ14987 | hypothetical protein FLJ149878266220.59493816p13.12KIAA0430 | KIAA0430 gene product7011120.591773 3p25XPC | xeroderma pigmentosum, complementation group C18596250.591377 8q24BAI1 | brain-specific angiogenesis inhibitor 18129750.586956 9p23KIAA0172 | KIAA0172 protein2140680.58591810p15GATA3 | GATA-binding protein 315878630.581689 3p23-p22ACAA1 | acetyl-Coenzyme A acyltransferase 1 (peroxisomal 3-oxoacyl-Coenzyme Athiolase)15184020.57627517q11.1KIAA1361 | KIAA1361 protein7969960.57565Xq13.1-q13.3IGBP1 | immunoglobulin (CD79A) binding protein 113234480.575218 7q11.23CRIP1 | cysteine-rich protein 1 (intestinal)23885710.57410919p13.1-q12AKAP8 | A kinase (PRKA) anchor protein 8750780.57327612ESTs16046420.572299 6Homo sapiens cDNA FLJ32724 fis, clone TESTI2000951665320.57217920q13.2-q13.3EDN3 | endothelin 322734450.57191720q11.2GHRH | growth hormone releasing hormone3466430.56762610ESTs5952970.563887 1q21.3SNAPAP | SNARE associated protein snapin9713990.56144812cen-q21SYT1 | synaptotagmin I8975500.56106517q21.2MGC2744 | hypothetical protein MGC27442150000.560663 3p22VIPR1 | vasoactive intestinal peptide receptor 11558960.56056411cen-q12.1LOC51035 | ORF17004290.5605310q26GFRA1 | GDNF family receptor alpha 12774630.56006818p11.2C18orf1 | chromosome 18 open reading frame 115877100.55685417p13.1-17p12PER1 | period (Drosophila) homolog 15658490.55621 1q32.1C3IP1 | kelch-like protein C3IP11268510.55574810q22.1FLJ11160 | hypothetical protein FLJ1116024133370.55435911q23.2-q24.2SORL1 | sortilin-related receptor, L(DLR class) A repeats-containing8247530.55402713FLJ22624 | hypothetical protein FLJ22624504710.55305811Homo sapiens cDNA FLJ14242 fis, clone OVARC1000678335000.552872Homo sapiens clone 23556 mRNA sequence7525470.55191615Homo sapiens mRNA; cDNA DKFZp586G1520 (from clone DKFZp586G1520)833580.550166ESTs20963060.55013 8q24.3ARC | activity-regulated cytoskeleton-associated protein1961890.54857418q23CYB5 | cytochrome b-520188080.54627611q14PRCP | prolylcarboxypeptidase (angiotensinase C)15005420.54451716p13.3RGS11 | regulator of G-protein signalling 114700610.544311 3q25SIAH2 | seven in absentia (Drosophila) homolog 217621110.543871 5p14-p13NPR3 | natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptidereceptor C)21161880.54347217q21HDAC5 | histone deacetylase 58266680.542351 6q21KIAA0274 | KIAA0274 gene product267360.54063820Homo sapiens cDNA FLJ30872 fis, clone FEBRA20042936693790.540325 7Homo sapiens, clone IMAGE: 3463399, mRNA, partial cds2217760.53659414ESTs, Weakly similar to T20410 hypothetical protein E02A10.2 —Caenorhabditiselegans [C.elegans]2646320.53573719ESTs7417900.53497 2p13.3FLJ20080 | hypothetical protein FLJ2008016260870.53252 3p21.31DKFZP434A236 | DKFZP434A236 protein8120330.532407 2q35-q37GPC1 | glypican 19505740.53109217q25H3F3B | H3 histone, family 3B (H3.3B)2840220.531011 8p23ARHGEF10 | Rho guanine nucleotide exchange factor (GEF) 10358280.528716 5q23DTR | diphtheria toxin receptor (heparin-binding epidermal growth factor-like growthfactor)22846190.52852219q13.4ZNF132 | zinc finger protein 132 (clone pHZ-12)6819920.528384 7Homo sapiens cDNA FLJ13384 fis, clone PLACE1001062, highly similar to Homosapiens mRNA for lysine-ketoglutarate reductase/saccharopine dehydrogenase439330.52806Xp11.4-p11.3MAOA | monoamine oxidase A7855380.527955Homo sapiens cDNA FLJ32293 fis, clone PROST20017393437600.526569 6q13-15SH3BGRL2 | SH3 domain binding glutamic acid-rich protein like 27855710.52567910DNAJL1 | hypothetical protein similar to mouse Dnajl18095070.52540616p13.3FLJ20568 | hypothetical protein FLJ2056818956640.52422715q26.1PRO2198 | hypothetical protein PRO21988236610.52121814Homo sapiens cDNA FLJ31768 fis, clone NT2RI2007891, moderately similar toDMR-N9 PROTEIN8429800.51990922q12.2DRG1 | developmentally regulated GTP-binding protein 11264190.517789 1q21-q22NIT1 | nitrilase 119260230.516851 7ESTs, Weakly similar to T42727 proliferation potential-related protein-mouse[M. musculus]1328570.51638217Homo sapiens mRNA; cDNA DKFZp586N1323 (from clone DKFZp586N1323)8555860.515352 5q31NR3C1 | nuclear receptor subfamily 3, group C, member 18103310.515056 1q24QSCN6 | quiescin Q62651030.512718 1p36MMEL2 | membrane metallo-endopeptidase-like 215213610.511233 8p21.2KIAA0717 | KIAA0717 protein4320720.50877418q23NFATC1 | nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 120696020.50611516q24.3MC1R | melanocortin 1 receptor (alpha melanocyte stimulating hormone receptor)2831730.505655 4Homo sapiens PAC clone RP1-130H16 from 22q12.1-qter14048410.50104919q13.4ZNF175 | zinc finger protein 17518711160.500004 2Homo sapiens mRNA; cDNA DKFZp434C1714 (from clone DKFZp434C1714); partialcds7583650.498812q13-q15OS4 | conserved gene amplified in osteosarcoma16418940.49854210ESTs14921470.498131Xq13.1RPS4X | ribosomal protein S4, X-linked15586420.497736 2q37.3MLPH | melanophilin16412450.49772318q21.1LOC51320 | hypothetical protein16356490.49764720p13CDS2 | CDP-diacylglycerol synthase (phosphatidate cytidylyltransferase) 24149990.49685517q21ETV4 | ets variant gene 4 (E1A enhancer-binding protein, E1AF)15359570.496325 5p15.3SEC6 | similar to S. cerevisiae Sec6p and R. norvegicus rsec67740820.49588312q22-q23ASCL1 | achaete-scute complex (Drosophila) homolog-like 18110130.494705 1p13.3AMPD2 | adenosine monophosphate deaminase 2 (isoform L)8099980.493372 1p21AMY2A | amylase, alpha 2A; pancreatic20180840.48899 2q24.3SPAK | Ste-20 related kinase1613730.485425 7q11-q22PMS2L4 | postmeiotic segregation increased 2-like 41781370.485162 4q25RPL34 | ribosomal protein L34758860.484926 4ESTs, Weakly similar to E54024 protein kinase [H. sapiens]4293870.484053 7p15.3CHN2 | chimerin (chimaerin) 27429770.481369 7p13DKFZP761I2123 | KIAA1886 protein2406370.480946 1p33-p32.1MGC8974 | hypothetical protein MGC89748383660.480888 1p36.1-p35HMGCL | 3-hydroxymethyl-3-methylglutaryl-Coenzyme A lyase(hydroxymethylglutaricaciduria)7961810.48010213q34GAS6 | growth arrest-specific 6237760.479727 4p15.31QDPR | quinoid dihydropteridine reductase19094330.47806417Homo sapiens cDNA FLJ30754 fis, clone FEBRA200043821609200.477446 1p13PHTF1 | putative homeodomain transcription factor 115005360.47593312pter-p13.31MDS028 | uncharacterized hematopoietic stem/progenitor cells protein MDS0282945370.474189 2q37.3RAB17 | RAB17, member RAS oncogene family7840850.469813 6q25-q26TUSP | tubby super-family protein2398770.469171 5q31HDAC3 | histone deacetylase 36268610.46884411p15EIF4G2 | eukaryotic translation initiation factor 4 gamma, 27419770.466816 6p21.3BF | B-factor, properdin


[0106] Some of the genes within the tumor grade I/III signatures have been previously reported to be associated with breast cancer. Within the grade I signature, two genes, BCL2 and TNFRSF10C, are inhibitors of apoptosis. Various reports in the literature link BCL2 expression to ER-positive, low-grade tumors (van Slooten, H. J. et al.). TNFRSF10C is a decoy receptor (DcR1) for TRAIL, an apoptosis-inducing cytokine of the tumor necrosis factor (TNF) family (Sheridan, J. P. et al.). Without being bound by theory, presence of DcR1 on the surface of breast cancer cells would be expected to block signaling through the cell death receptors activated by TRAIL, thus inhibiting apoptosis.


[0107] Similarly, an expanded set of 250 genes that display increased expression in Grade III samples in comparison to Grade I samples are identified in Table 9 by use of their I.M.A.G.E. Consortium CloneID numbers along with their chromosomal location and descriptive identifiers (if known) and relative weights (which are expressed with a negative sign solely due to the relative comparison).
9TABLE 9IMAGEChromosomeClone IDWeightLocationDescription769921−1.5356820q13.12UBE2C | ubiquitin-conjugating enzyme E2C951241−1.3381515q13.3ANKT | nucleolar protein ANKT1517595−1.3332 9p11.2KIAA0175 | likely ortholog of maternal embryonic leucine zipper kinase1474424−1.3207217Homo sapiens cDNA FLJ31911 fis, clone NT2RP70047512309073−1.29533 2q33-q34FZD5 | frizzled (Drosophila) homolog 5796469−1.27516 1q32.1HSPC150 | HSPC150 protein similar to ubiquitin-conjugating enzyme823598−1.26568PSMD12 | proteasome (prosome, macropain) 26S subunit, non-ATPase, 12700792−1.2523214q22CDKN3 | cyclin-dependent kinase inhibitor 3 (CDK2-associated dual specificityphosphatase)2018131−1.2321712p13.2-p13.1RACGAP1 | Rac GTPase activating protein 1292936−1.20973 1p34.3FLJ10468 | hypothetical protein FLJ104681422338−1.20922 2p25-p24RRM2 | ribonucleotide reductase M2 polypeptide504308−1.1874310cen-q26.11FLJ10540 | hypothetical protein FLJ10540796694−1.1644417q25BIRC5 | baculoviral IAP repeat-containing 5 (survivin)869375−1.1536315q26.1IDH2 | isocitrate dehydrogenase 2 (NADP+), mitochondrial814270−1.14538 4q27PMSCL1 | polymyositis/scleroderma autoantigen 1 (75 kD)42831−1.1287811q11-q12NTKL | N-terminal kinase-like1476053−1.1046215q15.1RAD51 | RAD51 (S. cerevisiae) homolog (E coli RecA homolog)32493−1.10275 2q31.1ITGA6 | integrin, alpha 6149355−1.10225 8q13.1TRAM | translocating chain-associating membrane protein824962−1.0991817q23.1-q23.3KPNA2 | karyopherin alpha 2 (RAG cohort 1, importin alpha 1)1702742−1.0964416q24.3SLC7A5 | solute carrier family 7 (cationic amino acid transporter, y+ system),member 5824524−1.0785417q21.32UGTREL1 | UDP-galactose transporter related128711−1.07401 7p15-p14ANLN | anillin (Drosophila Scraps homolog), actin binding protein843121−1.06508 6p22.1-p21.2CLIC1 | chloride intracellular channel 12017415−1.06388 2p24-p21CENPA | centromere protein A (17 kD)753378−1.0364 4q34.1FLJ22649 | hypothetical protein FLJ22649 similar to signal peptidaseSPC22/23825470−1.0350717q21-q22TOP2A | topoisomerase (DNA) II alpha (170 kD)705064−1.02376 4p16.3TACC3 | transforming, acidic coiled-coil containing protein 32054635−1.0204220q13.33PSMA7 | proteasome (prosome, macropain) subunit, alpha type, 7781047−1.0153 2q14BUB1 | budding uninhibited by benzimidazoles 1 (yeast homolog)1534700−1.0134311q21KIAA0830 | KIAA0830 protein1587847−1.01171 2q21MCM6 | minichromosome maintenance deficient (mis5, S. pombe) 6743810−1.009912p13MGC2577 | hypothetical protein MGC2577897609−0.9937912q23.2FLJ10074 | hypothetical protein FLJ1007466406−0.98421 2ESTs, Highly similar to T47163 hypothetical protein DKFZp762E1312.1[H. sapiens]1631634−0.98233 9q34.11MGC3038 | hypothetical protein similar to actin related protein 2/3 complex,subunit 5624627−0.96436 2p25-p24RRM2 | ribonucleotide reductase M2 polypeptide814054−0.95575 1q24-25KIAA0040 | KIAA0040 gene product773301−0.9129416q22.1CDH3 | cadherin 3, type 1, P-cadherin (placental)1416055−0.91005 8KIAA0165 | extra spindle poles, S. cerevisiae, homolog of345787−0.8955418p11.31HEC | highly expressed in cancer, rich in leucine heptad repeats624667−0.88376 9q34.13LOC51117 | CGI-92 protein786067−0.8771420p13CDC25B | cell division cycle 25B785368−0.87699 8p21-p12TOPK | PDZ-binding kinase; T-cell originated protein kinase564981−0.8551318Homo sapiens, Similar to RIKEN cDNA 2810433K01 gene, clone MGC: 10200IMAGE: 3909951, mRNA, complete cds753320−0.85505 8q13.3FLJ20533 | hypothetical protein FLJ20533529827−0.85016Xp22.31SYAP1 | reserved122241−0.84842 1p34.2PSMB2 | proteasome (prosome, macropain) subunit, beta type, 2712139−0.84823 2q37.2ARL7 | ADP-ribosylation factor-like 7259950−0.83947 8q23CML66 | chronic myelogenous leukemia tumor antigen 66772220−0.83895 3q21.2PDIR | for protein disulfide isomerase-related124331−0.8366416CPSF5 | cleavage and polyadenylation specific factor 5, 25 kD subunit842818−0.8333816q23-q24KARS | lysyl-tRNA synthetase150897−0.8292219p13.1B3GNT3 | UDP-GlcNAc:betaGal beta-1,3-N-acetylglucosaminyltransferase 3823930−0.82876 7q22.1ARPC1A | actin related protein 2/3 complex, subunit 1A (41 kD)210862−0.8231217q24-17q25ACOX1 | acyl-Coenzyme A oxidase 1, palmitoyl731023−0.82276 9q34WDR5 | WD repeat domain 5665384−0.8223216KIAA1609 | KIAA1609 protein815501−0.8210819p13.3MGC2721 | hypothetical protein MGC2721769890−0.8186414q13.1NP | nucleoside phosphorylase209066−0.8112120q13.2-q13.3STK15 | serine/threonine kinase 15471568−0.8102617q25HN1 | hematological and neurological expressed 1725454−0.80701 9q22CKS2 | CDC28 protein kinase 2951233−0.80178 2q35PSMB3 | proteasome (prosome, macropain) subunit, beta type, 3268946−0.79976 2Homo sapiens cDNA FLJ31861 fis, clone NT2RP70013192028949−0.7865117q21.31PRO1855 | hypothetical protein PRO18551914863−0.78621 2p13.3-p13.1DYSF | dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive)744047−0.7773716p12.3PLK | polo (Drosophia)-like kinase703707−0.77579 8q12.1ASPH | aspartate beta-hydroxylase78869−0.7694820q13.33GP110 | cell membrane glycoprotein, 110000M(r) (surface antigen)742707−0.7686 7ESTs, Weakly similar to MUC2_HUMAN MUCIN 2 PRECURSOR [H. sapiens]825606−0.7581710q24.1KNSL1 | kinesin-like 1361922−0.7559 1p34ZMPSTE24 | zinc metalloproteinase, STE24 (yeast, homolog)756595−0.75094 1q21S100A10 | S100 calcium-binding protein A10 (annexin II ligand, calpactin I,light polypeptide (p11))756442−0.7508 7q11.2POR | P450 (cytochrome) oxidoreductase823907−0.74968 8q12.2FLJ10511 | hypothetical protein FLJ10511471196−0.74806 2q37ITM3 | integral membrane protein 3753428−0.74668 8Homo sapiens, Similar to RIKEN cDNA 1110014B07 gene, clone MGC: 20766IMAGE: 4586039, mRNA, complete cds739450−0.74247 1q21.2LASS2 | longevity assurance (LAG1, S. cerevisiae) homolog 21696757−0.7384913q22.2KIAA1165 | hypothetical protein KIAA1165293727−0.7321322q13.2MGC861 | hypothetical protein MGC861839682−0.73112q22UBE2N | ubiquitin-conjugating enzyme E2N (homologous to yeast UBC13)1631132−0.7305311q12.1PHT2 | peptide transporter3327506−0.7296615Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 3275061660666−0.72774Xp21.1CA5B | carbonic anhydrase VB, mitochondrial280375−0.72588 8p22PRO2000 | PRO2000 protein796904−0.71939 6q24-q25PLAGL1 | pleiomorphic adenoma gene-like 1503671−0.71201 6Homo sapiens cDNA FLJ14368 fis, clone HEMBA100112274677−0.71194Homo sapiens, Similar to RIKEN cDNA A430107 J06 gene, clone MGC: 21416IMAGE: 4452699, mRNA, complete cds291478−0.71127 1p36RUNX3 | runt-related transcription factor 3825282 −0.7096DKFZP586L0724 | DKFZP586L0724 protein878330−0.70859 3Homo sapiens cDNA: FLJ22044 fis, clone HEP0914137671−0.7037418q11.2FLJ21610 | hypothetical protein FLJ21610789012−0.7019 3p25-p24FBLN2 | fibulin 2347373−0.70161 8q13.3TCEB1 | transcription elongation factor B (SIII), polypeptide 1 (15 kD, elonginC)2322367−0.69997 2p14-p13RTN4 | reticulon 4897997−0.69961Xp11.22-p11.21SMC1L1 | SMC1 (structural maintenance of chromosomes 1, yeast)-like 1345538−0.69527 9q21-q22CTSL | cathepsin L1947647−0.6937117q23.3LOC51651 | CGI-147 protein3172883−0.6916411ESTs, Weakly similar to S24195 dopamine receptor D4 [H. sapiens]1035796−0.68832 1ESTs, Weakly similar to T33068 hypothetical protein C35E7.9 - Caenorhabditiselegans [C.elegans]746163−0.67918 8ESTs, Weakly similar to ALU1HUMAN ALU SUBFAMILY J SEQUENCECONTAMINATION WARNING ENTRY [H. sapiens]810711−0.6774310q23-q24SCD | stearoyl-CoA desaturase (delta-9-desaturase)462926−0.67573 1q32.2-q41NEK2 | NIMA (never in mitosis gene a)-related kinase 21614140−0.6736815q11.2-q22.33LOC51285 | Ris124781−0.66984 8q24.1SQLE | squalene epoxidase1642496−0.66639 2p24.1MGC11266 | hypothetical protein MGC11266113300−0.66053 9q22.32TRIM14 | tripartite motif-containing 142014034−0.65845 2p12MTHFD2 | methylene tetrahydrofolate dehydrogenase (NAD+ dependent),methenyltetrahydrofolate cyclohydrolase1946448−0.65464 7q31.1CAV2 | caveolin 21635352−0.65164 4q12TPARL | TPA regulated locus753400−0.64918 3q27.1BAF53A | BAF531605426−0.64391 4q12FLJ13352 | hypothetical protein FLJ13352565319−0.64374 8MAL2 | mal, T-cell differentiation protein 2489755−0.6409510q26.3ADAM12 | a disintegrin and metalloproteinase domain 12 (meltrin alpha)1916461−0.6399322Homo sapiens, clone IMAGE: 3605655, mRNA359887−0.63379 1q32.1TIM17 | translocase of inner mitochondrial membrane 17 homolog A (yeast)629944−0.6301818q12MYO5B | myosin VB150314−0.62891 6q13LYPLA1 | lysophospholipase I770355−0.6282921q22.3LSS | lanosterol synthase (2,3-oxidosqualene-lanosterol cyclase)489594−0.6245Xq22.2FLJ11565 | hypothetical protein FLJ11565212640−0.6219Xq28ARHGAP4 | Rho GTPase activating protein 430170−0.62007 4q34CASP3 | caspase 3, apoptosis-related cysteine protease51773−0.61957 7p15-p14MGC3077 | hypothetical protein MGC3077490777−0.619061858892−0.61695Xp22.13MGC4825 | hypothetical protein MGC4825358456−0.61552 7p11.2SEC61G | Sec61 gamma840894−0.6141412q24.2COX6A1 | cytochrome c oxidase subunit VIa polypeptide 1241348−0.61157PCL1 | prenylcysteine lyase1505038−0.61123 8q22.2FLJ20171 | hypothetical protein FLJ20171144880−0.6097619p13.3LOC56932 | hypothetical protein from EUROIMAGE 1759349454896 −0.6057516q11.1-q11.2DNAJA2 | DnaJ (Hsp40) homolog, subfamily A, member 2753236−0.604616 ESTs, Weakly similar to S71512 hypothetical protein T2 - mouse [M. musculus]266218−0.60106418159−0.6002522q13.1SYNGR1 | synaptogyrin 1208718−0.59862 9q12-q21.2ANXA1 | annexin A1781097−0.5971811q13RTN3 | reticulon 3469383−0.59434 8q21C8orf1 | chromosome 8 open reading frame 1725152−0.5937511q11DKFZp762A227 | hypothetical protein DKFZp762A227845363−0.5931317q21.3NME1 | non-metastatic cells 1, protein (NM23A) expressed in1460110−0.5920614q11.2PSMB5 | proteasome (prosome, macropain) subunit, beta type, 5769959−0.591313q34COL4A2 | collagen, type IV, alpha 2796527−0.59108 7q34DKFZp761N0624 | hypothetical protein DKFZp761N0624108425−0.59009 1ESTs, Weakly similar to JC5314 CDC28/cdc2-like kinase associating arginine-serine cyclophilin [H. sapiens]32231−0.58516FLJ12442 | hypothetical protein FLJ12442502690−0.58445 3q21.3-q25.2RPN1 | ribophorin I135221−0.58203 4p16S100P | S100 calcium-binding protein P897813−0.5816717p11.1PAIP1 | polyadenylate binding protein-interacting protein 1824352−0.58119 9q31.2RAD23B | RAD23 (S. cerevisiae) homolog B897751−0.5805717q23TLK2 | tousled-like kinase 2343607−0.5772715q14-q24.3LOC55829 | AD-015 protein51899−0.577216q24.1KIAA0513 | KIAA0513 gene product726645−0.5771616q23CLECSF1 | C-type (calcium dependent, carbohydrate-recognition domain)lectin, superfamily member 1 (cartilage-derived)1591264−0.5755811p15.5-p15.4TALDO1 | transaldolase 1290841−0.57171 6p21.3H2BFA | H2B histone family, member A486626−0.57063 8Homo sapiens, clone IMAGE: 4332938, mRNA221846−0.5676914q24.3-q31CHES1 | checkpoint suppressor 1772913−0.56751 5Homo sapiens cDNA FLJ31951 fis, clone NT2RP7007177, weakly similar toHomo sapiens multiple membrane spanning receptor TRC8 mRNA1686766−0.56178 6q15-q16RAGD | Rag D protein37708−0.5605316q24.3MGC3101 | hypothetical protein MGC3101825740−0.56021 2q32.1DKFZP434J1813 | DKFZp434J1813 protein741139−0.5600920q13.1EYA2 | eyes absent (Drosophila) homolog 2754293−0.55369 2p12C2orf6 | chromosome 2 open reading frame 683363−0.55322 6q24-q25PCMT1 | protein-L-isoaspartate (D-aspartate) O-methyltransferase686552−0.55207 1q42.13GOLPH1 | golgi phosphoprotein 1950429−0.5496212qKIAA1708 | KIAA1708 protein813419−0.54843Xp11.2HADH2 | hydroxyacyl-Coenzyme A dehydrogenase, type II2043167−0.5469410q25.2-q26.2BAG3 | BCL2-associated athanogene 3701115−0.54546 6PRO2013 | hypothetical protein PRO2013795498−0.5439115q26.1HS1-2 | putative transmembrane protein965223−0.5433317q23.2-q25.3TK1 | thymidine kinase 1, soluble377191−0.53874 8p22-q22.1LOC51123 | HSPC038 protein233679 −0.53609 2p23.3FLJ22362 | hypothetical protein FLJ22362590759−0.53571 4q32-q34SC4MOL | sterol-C4-methyl oxidase-like358083−0.53534 3q29KIAA0226 | KIAA0226 gene product810612−0.53335 1q21S100A11 | S100 calcium-binding protein A11 (calgizzarin)220395−0.52987 2p22.3FLJ23293 | likely ortholog of mouse ADP-ribosylation-like factor 6 interactingprotein 2280699−0.52812 7UCC1 | upregulated in colorectal cancer gene 12016775−0.5268716p12GPRC5B | G protein-coupled receptor, family C, group 5, member B470124−0.52331 5p13.2RAD1 | RAD1 (S. pombe) homolog154707−0.51926 2p23-p21MPV17 | MpV17 transgene, murine homolog, glomerulosclerosis785933−0.51884Xp21.1SRPX | sushi-repeat-containing protein, X chromosome2062825−0.5181920q11.23KIAA0964 | KIAA0964 protein2009491−0.51791 4q22.1-q23LOC51191 | cyclin-E binding protein 11534493−0.51765 8ESTs150003 −0.5167 8q22.2FLJ13187 | phafin 2950600−0.51409 1Homo sapiens mRNA; cDNA DKFZp586C1019 (from clone DKFZp586C1019)1455394−0.51333 7p15.2HCS | cytochrome c811918−0.5131820p12.1KIAA0952 | KIAA0952 protein415191−0.51 2p25.3KIAA0161 | KIAA0161 gene product32927−0.50974 8q24.3FBXL6 | f-box and leucine-rich repeat protein 61845744−0.50818325160−0.50752 3q13.13NP25 | neuronal protein812048−0.5054220pter-p12PRNP | prion protein (p27-30) (Creutzfeld-Jakob disease, Gerstmann-Strausler-Scheinker syndrome, fatal familial insomnia)84161−0.50451DKFZP434F195 | DKFZP434F195 protein897806−0.5023614q21-q24HIF1A | hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helixtranscription factor)814378−0.5002619q13.1SPINT2 | serine protease inhibitor, Kunitz type, 2188335−0.49789EMR2 | egf-like module containing, mucin-like, hormone receptor-likesequence 21585492−0.49501 9ESTs133213−0.4933211q21FUT4 | fucosyltransferase 4 (alpha (1,3) fucosyltransferase, myeloid-specific)73009−0.49156 5Homo sapiens, clone MGC: 9628 IMAGE: 3913311, mRNA, complete cds785707−0.4903115q26.1PRC1 | protein regulator of cytokinesis 184464−0.49025 1q42.12FLJ12806 | hypothetical protein FLJ12806770066−0.4896511q25KIAA0056 | KIAA0056 protein41208−0.48944 8p21BMP1 | bone morphogenetic protein 11698036−0.4890420q13.2UBE2V1 | ubiquitin-conjugating enzyme E2 variant 11435862−0.4889Xp22.32MIC2 | antigen identified by monoclonal antibodies 12E7, F21 and O13768452−0.4844921Homo sapiens EST from clone 491476, full insert824426−0.48229 7q22.1PDAP1 | PDGFA associated protein 1768561−0.4812917q11.2-q21.1SCYA2 | small inducible cytokine A2 (monocyte chemotactic protein 1,homologous to mouse Sig-je)377275−0.4809211q22-q23TRIM29 | tripartite motif-containing 29470128−0.4815q21-q22MYO1E | myosin IE809901−0.47984 9q21-q22COL15A1 | collagen, type XV, alpha 150772−0.47983 7p14-p13MGC3251 | hypothetical protein MGC32511843843−0.4790212q14.1SRGAP1 | KIAA1304 protein823940−0.4789717q21TOB1 ═ transducer of ERBB2, 1564492−0.4774911p11.12MTCH2 | mitochondrial carrier homolog 2290101−0.47734XESTs263894−0.4770416p12.1QPRT | quinolinate phosphoribosyltransferase (nicotinate-nucleotidepyrophosphorylase (carboxylating))202901−0.47699 9q34.1VAV2 | vav 2 oncogene1607229−0.47609 6q22-q23TPD52L1 | tumor protein D52-like 1812050−0.47584 8q24TRC8 | patched related protein translocated in renal cancer1637756−0.47545 1p36.3-p36.2ENO1 | enolase 1, (alpha)813410−0.47231 8q22.3POLR2K | polymerase (RNA) II (DNA directed) polypeptide K (7.0 kD)358162−0.4719711q13.1HSU79266 | protein predicted by clone 236272062238−0.47184 2q37.1PSMD1 | proteasome (prosome, macropain) 26S subunit, non-ATPase, 1753215−0.47119 7q21GNAI1 | guanine nucleotide binding protein (G protein), alpha inhibiting activitypolypeptide 1739126−0.46952 8q24.3TSTA3 | tissue specific transplantation antigen P35B1917941−0.46918 7p13H2AV | histone H2A.F/Z variant111362−0.4689320q11.2OSBPL2 | oxysterol-binding protein-like 21456348−0.46742 9p24.1-p23SAS | N-acetylneuraminic acid phosphate synthase; sialic acid synthase263716−0.4663621q22.3COL6A1 | collagen, type VI, alpha 1810156−0.46594 2DTYMK | deoxythymidylate kinase (thymidylate kinase)115443−0.46519HSPC216 | hypothetical protein32299−0.4642718p11.2IMPA2 | inositol(myo)-1(or 4)-monophosphatase 21434897−0.46024 2q14-q32COL5A2 | collagen, type V, alpha 22028916−0.4590510Homo sapiens mRNA for Hmob33 protein, 3′untranslated region2020898−0.45878 7q22PLOD3 | procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3487797−0.45837 1p22.1DR1 | down-regulator of transcription 1, TBP-binding (negative cofactor 2)284734−0.45795 6q21-q22WASF1 | WAS protein family, member 179520−0.45632 8q12.1RAB2 | RAB2, member RAS oncogene family812977−0.4536812Homo sapiens mesenchymal stem cell protein DSC96 mRNA, partial cds810899−0.45368ESTs428163−0.45286 3ESTs, Weakly similar to NAH6_HUMAN SODIUM/HYDROGEN EXCHANGER6 [H. sapiens]613056−0.4526111p13RCN1 | reticulocalbin 1, EF-hand calcium binding domain741474−0.4524919q13.1GPI | glucose phosphate isomerase768989−0.4514714Homo sapiens cDNA FLJ12874 fis, clone NT2RP2003769754702−0.45087 2p25.1-p24.1KIAA0846 | KIAA0846 protein246800−0.45055 7p13FLJ10803 | hypothetical protein FLJ10803246304−0.450321q21.1BTG3 | BTG family, member 3


[0108] The grade III signature contains genes known to be involved in cell cycle control (CKS2, CDC25B, MCM6), chromosomal segregation (STK15, CENPA and TACC3), and DNA recombination and repair (RAD51, UBE2N, TOP2A, RRM2). In particular, CDC25B, a potential oncogene, transforms murine diploid fibroblasts into high-grade tumors (Galaktionov, K. et al.). STK15, a centrosomal protein kinase, is frequently amplified in breast cancer, and its quantitative expression levels positively correlate with tumor grade (Zhou, H. et al. (1998b)). RAD51 has recently been shown to interact with the tumor suppressor BRCA1 (Chen, J. J., et al. (1999)), and its expression also positively correlates with tumor grade in breast cancer (Maacke, H. et al.). It has not been previously known or suspected, however, whether the expression of these genes would be capable of differentiating grade III breast cancer cells from grade I breast cancer cells. Without being bound by theory, abnormal expression of the genes associated with DNA recombination and repair and those associated with centrosomal function may result in greater genome instability, thus driving the evolution of aggressively growing and high-grade cancer cells. The data thus verified the association of several known genes with breast tumorigenesis and uncovered additional novel associations, which together may underlie the molecular basis of current tumor grading systems in breast cancer.


[0109] The question of whether unique gene expression changes are associated with stage progression, specifically, the transition from noninvasive (DCIS) to invasive (IDC) growth, is also addressed by the present invention. The inventors have noticed that these two pathological stages are highly similar to each other with no striking differences at the level of gene expression (FIGS. 2-3). To increase our sensitivity in detecting differential gene expression between DCIS and IDC, each IDC sample was compared directly to its corresponding patient-matched DCIS sample where available. 1,688 genes showing at least a 2-fold difference between IDC and DCIS in at least 3 different sample pairs were selected and subjected to unsupervised two-dimensional hierarchical clustering. One prominent cluster of genes demonstrated elevated expression in IDC as compared with DCIS, predominately amongst the grade III IDC samples (FIG. 4). These genes, along with their I.M.A.G.E. Consortium CloneID number, along with their chromosomal location and descriptive identifiers (if known) are listed in Table 10.
10TABLE 10IMAGEChromosomeClone IDLocation Description79549815q26.1 HS1-2 | putative transmembrane protein43150515q26.1 HS1-2 | putative transmembrane protein74113920q13.1 EYA2 | eyes absent (Drosophila) homolog 215345922p12 C2orf6 | chromosome 2 open reading frame 62904229q13-q21 ZNF216 | zinc finger protein 21616098361q31 GLUL | glutamate-ammonia ligase (glutamine synthase)5055752q35 FLJ10116 | hypothetical protein FLJ1011614185211q13.5-q14.1 P2RY2 | purinergic receptor P2Y, G-protein coupled, 212125112q13.1 MGC5576 | hypothetical protein MGC5576610326-1012q12-12q14.3 K-ALPHA-1 | tubulin, alpha, ubiquitous7254549q22 CKS2 | CDC28 protein kinase 27565027p22 NUDT1 | nudix (nucleoside diphosphate linked moiety X)-type motif 150430810cen-q26.11 FLJ1054O | hypothetical protein FLJ1054020623296q13-q21 TTK | TTK protein kinase56498118 Homo sapiens, Similar to RIKEN cDNA 2810433K01 gene, clone MGC:10200IMAGE:3909951, mRNA, complete cds9510808q24.3 RECQL4 | RecQ protein-like 42803758p22 PRO2000 | PRO2000 protein5302198 Homo sapiens cDNA FLJ32554 fis, clone SPLEN10001065944381q12-1q21.2 DJ328E19.C1.1 | hypothetical protein4702327 ESTs, Weakly similar to |37356 epithelial microtubule-associated protein, 115K [H.sapiens]2910571p32 CDKN2C | cyclin-dependent kinase inhibitor 2C (p18, inhibits CDK4)147605315q15.1 RAD51 | RAD51 (S. cerevisiae) homolog (E coli RecA homolog)1214362q11.2 MGC4677 | hypothetical protein MGC467770079214q22 CDKN3 | cyclin-dependent kinase inhibitor 3 (CDK2-associated dual specificity phosphatase)30863310q23-q24 HELLS | helicase, lymphoid-specific8095888q12.1 GGH | gamma-glutamyl hydrolase (conjugase, folylpolygammaglutamyl hydrolase)14553947p15.2 HCS | cytochrome c79669417q25 BIRC5 | baculoviral IAP repeat-containing 5 (survivin)201813112p13.2-p13.1 RACGAP1 | Rac GTPase activating protein 115878472q21 MCM6 | minichromosome maintenance deficient (mis5, S. pombe) 674381012p13 MGC2577 | hypothetical protein MGC257774404716p12.3 PLK | polo (Drosophia)-like kinase7050644p16.3 TACC3 | transforming, acidic coiled-coil containing protein 31518591810899ESTs20189765q35.1 PTTG1 | pituitary tumor-transforming 120174152p24-p21 CENPA | centromere protein A (17kD)81550119p13.3 MGC2721 | hypothetical protein MGC27216246272p25-p24 RRM2 | ribonucleotide reductase M2 polypeptide14223382p25-p24 RRM2 | ribonucleotide reductase M2 polypeptide610326-812q12-12q14.3 K-ALPHA-1 | tubulin, alpha, ubiquitous7976112q22 TMPO | thymopoietin610326-212q12-12q14.3 K-ALPHA-1 | tubulin, alpha, ubiquitous610326-412q12-12q14.3 K-ALPHA-1 | tubulin, alpha, ubiquitous610326-312q12-12q14.3 K-ALPHA-1 | tubulin, alpha, ubiquitous14760651p36.1-p35 STMN1 | stathmin 1/oncoprotein 1829378511 ESTs, Weakly similar to A46010 X-linked retinopathy protein [H.sapiens]4778117 TEM7 | tumor endothelial marker 7 precursor4151025q31 CDC25C | cell division cycle 25C86937515q26.1 IDH2 | isocitrate dehydrogenase 2 (NADP+), mitochondrial95124115q13.3 ANKT | nucleolar protein ANKT8142704q27 PMSCL1 | polymyositis/scleroderma autoantigen 1 (75kD)7853688p21-p12 TOPK | PDZ-binding kinase; T-cell originated protein kinase664062 ESTs, Highly similar to T47163 hypothetical protein DKFZp762E1312.1 [H.sapiens]2929361p34.3 FLJ10468 | hypothetical protein FLJ1046815175959p11.2 KIAA0175 | likely ortholog of maternal embryonic leucine zipper kinase1287117p15-p14 ANLN | anillin (Drosophila Scraps homolog), actin binding protein20040220q11.22-q12 DJ616B8.3 | hypothetical protein dJ616B8.382547017q21-q22 TOP2A | topoisomerase (DNA) II alpha (170kD)76989014q13.1 NP | nucleoside phosphorylase7964691q32.1 HSPC15O | HSPC150 protein similar to ubiquitin-conjugating enzyme53131917p13.1 STK12 | serine/threonine kinase 1214160558 KIAA0165 | extra spindle poles, S. cerevisiae, homolog of76992120q13.12 UBE2C | ubiquitin-conjugating enzyme E2C77099283968212q22 UBE2N | ubiquitin-conjugating enzyme E2N (homologous to yeast UBC13)84036420cen-q13.1 AHCY | S-adenosylhomocysteine hydrolase27691520q11.2 DNMT3B | DNA (cytosine-5-)-methyltransferase 3 beta


[0110] Interestingly, many of the genes in this cluster have been identified already within the grade III signature cluster (FIG. 3). These include genes involved in the cell cycle (e.g., MCM6, TOP2A, CKS2, CDC25C), centrosomal function (TACC3, CENPA), and DNA repair (RAD51, RRM2). Thus, a subset of genes that are expressed at high levels in grade III DCIS are further elevated in IDC, suggesting an intriguing link between the two lines of cancer progression, i.e., tumor grade and invasion. Indeed, and without being bound by theory, RRM2, the M2 subunit of ribonucleotide reductase (RR), which catalyzes a rate-limiting step in DNA synthesis and repair, may play a dual role in both proliferative growth and invasion; overexpression of RRM2 in human cancer cells enhances their invasive potential (Zhou, B. S. et al. (1998c)), whereas its decreased expression inhibits cancer cell proliferation (Chen, S. et al. (2000)). In addition, centrosome amplification (e.g., induced by overexpression of STK15, Zhou et al. 1998b) may result in both high tumor grade and increased invasion potential due to altered cytoskeletal architecture (Lingle, W. L. et al.). However, these genes are not associated with the transition of grade I DCIS to grade I IDC, suggesting that the latter may employ a different mechanism(s) to gain invasion potential.


[0111] Without being bound by theory, and offered for the purposes of improving the understanding of the present invention and its possible applications, the above LCM-derived gene expression profiles of the various phenotypic stages of breast cancer are consistent with a modified model of breast cancer progression (FIG. 5). In this model, breast cancer develops along two dimensions, one of which consists of stage transitions from normal to ADH to DCIS to IDC and another consists of tumor grade progression from grade I to II to III. This model is supported by existing histopathological and clinical data (see Dupont, W. D. et al.; Marshall, L. M. et al.; Betsill, W. L. et al.; and Page, D. L. et al. (1982)) and the following lines of evidence presented above. First, extensive changes in gene expression occur in ADH and persist in DCIS/IDC, suggesting a molecular linkage between ADH and DCIS/IDC. Second, the identified 200 genes whose expression levels quantitatively correlate with tumor grade progression in both DCIS and IDC indicate a transcriptional continuum from low to high-grade tumors. Finally, grade III DCIS and IDC differ quantitatively in the expression of the same genes associated with tumor grade progression. It is thus proposed that the various subtypes (e.g., ER+ and ER− subtypes) of breast cancer represent snapshots of this two-dimensional progression scheme; for example (and without limiting the invention), during the progression from grade I through grade III, ER-positive lesions evolve into ER-negative ones. The present invention thus provides the identity, and thus sequences, of various genes associated with the initiation and progression of breast cancer, and so provides for novel diagnostic, preventative and therapeutic strategies for women with breast cancer. This includes the ability to utilize the grade of DCIS/IDC breast cancer, irrespective of which stage of breast cancer is actually present, as a criterion for decisions concerning breast cancer diagnosis and treatment.


[0112] The following Table 11 summarizes the contents of Tables 2-10
11TABLE 11TableDescription2Genes with elevated expression in ADH and persisting throughDCIS and IDC cells compared to normal cells3Genes with highest expression in grade III DCIS or IDC cells4Genes with decreased expression in ADH, DCIS and IDC cellscompared to normal cells5Genes correlated with grade I and III samples and decreasedexpression in all samples6Genes with increased expression in grade III(DCIS and/or IDC) samples7Genes with increased expression in grade I(DCIS and/or IDC) samples8250 genes with increased expression in grade I(DCIS and/or IDC) samples9250 genes with increased expression in grade III(DCIS and/or IDC) samples10Genes with quantitative differences in expression betweenDCIS and IDC samples


[0113] Having now generally described the invention, the same will be more readily understood through reference to the following examples which are provided by way of illustration, and are not intended to be limiting of the present invention, unless specified.



EXAMPLES


Example I

[0114] Materials and Methods


[0115] Clinical specimen collection and clinicopathological parameters. All breast specimens were obtained from the Massachusetts General Hospital between 1998 and 2001. Thirty-six breast cancer patients were selected, 31 of which were diagnosed with two or more pathological stages of breast cancer progression, and 5 of which were diagnosed with pre-invasive disease only. Three healthy women who underwent elective mammoplasty reduction were selected as disease-free normal controls. Tissue specimens that demonstrated one or more pathological lesions (ADH, DCIS and IDC) were selected for the study. Cases of ADH were selected as proliferative epithelial lesions that possessed some, but not all, of the features of carcinoma in situ (Page, D. L. et al. (1992)) and most closely resemble those lesions described as CAPSS (Oyama, T. et al. and Fraser, J. L. et al.). DCIS and IDC were classified (histological grade) according to the European classification (Holland, R. et al.) and by the Nottingham combined histological grade (Elston, C. W. et al.), respectively. ER and PR expression were determined by immunohistochemical staining (negative when none of the tumor cell nuclei showed staining), and Her-2 expression determined by immunohistochemistry or FISH. This study was approved the Massachusetts General Hospital human research committee in accordance with NIH human research study guidelines.


[0116] LCM and RNA isolation and amplification. Each component (Normal, ADH, DCIS or IDC) was laser capture microdissected in triplicate (from consecutive tissue sections) as described (Sgroi et al.) using a PixCell II LCM system (Arcturus Engineering Inc., Mountain View, Calif.). Total RNA was extracted from the captured cells using the Picopure™ RNA Isolation Kit (Arcturus). T7-based RNA amplification was carried out using the RiboAmp™ kit (Arcturus). Briefly, the RNA from each sample was primed with an oligo-dT primer containing a T7 promoter sequence, reverse transcribed and then converted to double stranded cDNA. The cDNA templates were then used in an in vitro transcription reaction using T7 RNA polymerase to generate amplified RNA (aRNA). To obtain enough aRNA for a microarray experiment, a second round of RNA amplification was performed on all samples. To serve as reference in microarray hybridizations, a human universal reference RNA from Stratagene (La Jolla, Calif.) was amplified identically.


[0117] Fabrication of microarrays. Sequence-verified human cDNA clones were obtained from Research Genetics (Huntsville, Ala.). cDNA clones (from the I.M.A.G.E. Consortium via Research Genetics) inserts were amplified by PCR, gel-purified, and spotted onto a 1×3-inch SuperAmine™ (TeleChem International, Sunnyvale, Calif.) glass microscope slide using an OmniGrid™ robotic arrayer (GeneMachines, San Carlos, Calif.). As used herein, the I.M.A.G.E. Consortium CloneID, or the IMAGE CloneID, lists the identifiers of the cDNA clones on the microarrays according to the I.M.A.G.E. Consortium and Research Genetics (www.resgen.com/). This provides a unique single identifier for each clone. Descriptive names of clones (or genes) use the UniGene symbols and titles (www.ncbi.nlm.nih.gov/UniGene/).


[0118] Probe labeling and hybridization. cDNA was transcribed from aRNA in the presence of 5-(3-aminoallyl)-2′-deoxyuridine 5′-triphosphate (aminoallyl dUTP) using Stratagene's FairPlay kit™ (La Jolla, Calif.). Cy3 or Cy5 mono- reactive dye (Amersham, Piscataway, N.J.) was conjugated onto purified cDNA and the residual dye was removed using QiaQuick PCR Purification columns (Qiagen, Valencia, Calif.). Each Cy5-labeled cDNA was hybridized together with the Cy3-labeled reference probe to a microarray in 40 μL hybridizationr solution (5×SSC, 0.1 μg/pL COT I, 0.2% SDS, 50% formamide) at a concentration of 25 ng/μL per channel for 17 hrs at 42° C. in >60% relative humidity.


[0119] Washing, scanning and image analysis. After hybridization, slides were washed as follows: 1×SSC, 0.2% SDS at 42° for 5 min (two times), 1×SSC, 0.2% SDS at 55° C. for 5 min, 0.1×SSC, 0.2% SDS at 55° C. for 5 min and 0.1×SSC at RT for 2 min. Washed slides were scanned using ScanArray 5000 (PerkinElmer, Billerica, Mass.), and Cy5/Cy3-signals were quantitated using ImaGene 4.2 (BioDiscovery, Los Angeles, Calif.).


[0120] Data processing. Fluorescent intensities of Cy5 and Cy3 channels on each slide were subjected to spot filtering and normalization. Spots flagged by ImaGene were excluded from further analysis. Normalization was performed using a robust nonlinear local regression method (Yang, Y. H. et al.). The normalized ratios of Cy5/Cy3 were used to represent the relative gene expression levels in the experimental samples. Measurements from replicate samples were averaged after normalization.


[0121] Cluster and discriminant analysis. Hierarchical cluster analysis was performed in GeneMaths (v1.5, Applied-Maths, Austin, Tex.) using the cosine correlation coefficient as a measure of similarity between two genes or samples and complete linkage. Linear discriminant analysis with variance was performed within GeneMaths.



Example II

[0122] Genes Showing Significant Differences in the Pair-Wise Comparisons of Normal vs. ADH, Normal vs. DCIS and Normal vs. IDC by Linear Discriminant Analysis


[0123] 2-3 independent LCM captures were made from the same breast biopsy for each disease state (normal, ADH, DCIS or IDC), and RNA from each capture was amplified, labeled, and hybridized to 2 identical 12,000-element microarrays, resulting in from 4 to 6 data points per gene per disease state. The replicate data points were averaged to represent the expression level of each gene at each cellular state, which was further transformed as data points which are the log2 value of the ratio of data from patient matched disease/normal samples or the log2 value of the ratio of data from patient matched IDC/DCIS samples.



REFERENCES

[0124] DeRisi, J., et al., Use of a cDNA microarray to analyse gene expression patterns in human cancer, Nature Genetics, (1996) 14:457-460.


[0125] Hedenfalk, I., et al., Gene-Expression Profiles In Heredity Breast Cancer, The New England Journal of Medicine, (Feb. 22, 2001) 344:8:539-548.


[0126] Golub, T. R., et al., Molecular Classification of Cancer: Class Discovery and Class Prediction by Gene Expression Monitoring, Science, (Oct. 15, 1999) 286:531-537.


[0127] Perou, Charles M., et al., Molecular portraits of human breast tumours, Nature, (Aug. 17, 2000) 406:747-752.


[0128] Garber, Mitchell E., et al., Diversity of gene expression in adenocarcinoma of the lung, Proc. Natl. Acad. Sci. USA, (Nov. 20, 2001) 98:24:13784-13789.


[0129] Perou, Charles M., et al., Distinctive gene expression patterns in human mammary epithelial cells and breast cancers, Proc. Natl. Acad. Sci. USA, (August 1999) 96:9212-9217.


[0130] Sgrio, Dennis C., et al., In Vivo Gene Expression Profile Analysis of Human Breast Cancer Progression, Cancer Research, (Nov. 15, 1999) 59:5656-5661.


[0131] Sorlie, Therese, et al., Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications, Proc. Natl. Acad. Sci., (Sep. 11, 2001) 98:19:10869-10874.


[0132] Alizadeh, Ash A., et al., Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling, Nature, (Feb. 3, 2000) 403:503-511.


[0133] Bittner, M., et al., Molecular classification of cutaneous malignant melanoma by gene expression profiling, Nature (Aug. 3, 2000) 406:536-540.


[0134] West, Mike, et al., Predicting the clinical status of human breast cancer by using gene expression profiles, Proc. Natl. Acad. Sci., (Sep. 25, 2001) 98:20:11462-11467.


[0135] Zhou, J. et al. A novel transcription factor, ELF5, belongs to the ELF subfamily of ETS genes and maps to human chromosome 11 p13-15, a region subject to LOH and rearrangement in human carcinoma cell lines. Oncogene 17, 2719-32. (1998a).


[0136] Fitzgibbons, P. L. et al. Prognostic factors in breast cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 124, 966-78. (2000).


[0137] Page, D. L. et al. Prediction of node-negative breast cancer outcome by histologic grading and S-phase analysis by flow cytometry: an Eastern Cooperative Oncology Group Study (2192). Am J Clin Oncol 24, 10-8. (2001).


[0138] Dalton, L. W. et al. Histologic grading of breast cancer: linkage of patient outcome with level of pathologist agreement. Mod Pathol 13, 730-5. (2000).


[0139] van Slooten, H. J. et al. Expression of Bcl-2 in node-negative breast cancer is associated with various prognostic factors, but does not predict response to one course of perioperative chemotherapy. Br J Cancer 74, 78-85. (1996).


[0140] Sheridan, J. P. et al. Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors. Science 277, 818-21. (1997).


[0141] Galaktionov, K. et al. CDC25 phosphatases as potential human oncogenes. Science 269, 1575-7. (1995).


[0142] Zhou, H. et al. Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation. Nat Genet 20, 189-93. (1998b).


[0143] Chen, J. J., Silver, D., Cantor, S., Livingston, D. M. & Scully, R. BRCA1, BRCA2, and Rad51 operate in a common DNA damage response pathway. Cancer Res 59, 1752s-1756s. (1 999).


[0144] Maacke, H. et al. Over-expression of wild-type Rad51 correlates with histological grading of invasive ductal breast cancer. Int J Cancer 88, 907-13. (2000).


[0145] Zhou, B. S. et al. Overexpression of transfected human ribonucleotide reductase M2 subunit in human cancer cells enhances their invasive potential. Clin Exp Metastasis 16, 43-9. (1998c).


[0146] Chen, S., Zhou, B., He, F. & Yen, Y. Inhibition of human cancer cell growth by inducible expression of human ribonucleotide reductase antisense cDNA. Antisense Nucleic Acid Drug Dev 10, 111-6. (2000).


[0147] Lingle, W. L. et al. Centrosome amplification drives chromosomal instability in breast tumor development. Proc Natl Acad Sci USA 99, 1978-83. (2002).


[0148] Dupont, W. D. & Page, D. L. Risk factors for breast cancer in women with proliferative breast disease. N Engl J Med 312, 146-51. (1985).


[0149] Marshall, L. M. et al. Risk of breast cancer associated with atypical hyperplasia of lobular and ductal types. Cancer Epidemiol Biomarkers Prev 6, 297-301. (1997).


[0150] Betsill, W. L., Jr., Rosen, P. P., Lieberman, P. H. & Robbins, G. F. Intraductal carcinoma. Long-term follow-up after treatment by biopsy alone. Jama 239, 1863-7. (1978).


[0151] Page, D. L., Dupont, W. D., Rogers, L. W. & Landenberger, M. Intraductal carcinoma of the breast: follow-up after biopsy only. Cancer 49, 751-8. (1982).


[0152] Page, D. L. & Rogers, L. W. Combined histologic and cytologic criteria for the diagnosis of mammary atypical ductal hyperplasia. Hum Pathol 23, 1095-7. (1992).


[0153] Oyama, T., Maluf, H. & Koerner, F. Atypical cystic lobules: an early stage in the formation of low-grade ductal carcinoma in situ. Virchows Arch 435, 413-21. (1999).


[0154] Fraser, J. L., Raza, S., Chorny, K., Connolly, J. L. & Schnitt, S. J. Columnar alteration with prominent apical snouts and secretions: a spectrum of changes frequently present in breast biopsies performed for microcalcifications. Am J Surg Pathol 22, 1521-7. (1998).


[0155] Holland, R. et al. Ductal carcinoma in situ: a proposal for a new classification. Semin Diagn Pathol 11, 167-80. (1994).


[0156] Elston, C. W. & Ellis, I. O. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19, 403-10. (1991).


[0157] Yang, Y. H. et al. Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res 30, e15. (2002).


[0158] Unger, M. A. et al. Characterization of adjacent breast tumors using oligonucleotide microarrays. Breast Cancer Res 3, 336-41 (2001).


[0159] van't Veer, L. J. et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 415, 530-6. (2002).


[0160] Gruvberger, S. et al. Estrogen receptor status in breast cancer is associated with remarkably distinct gene expression patterns. Cancer Res 61, 5979-84. (2001).


[0161] Luo, L. et al. Gene expression profiles of laser-captured adjacent neuronal subtypes. Nat Med 5, 117-22. (1999).


[0162] Lennon et al. The I.M.A.G.E. Consortium: An Integrated Molecular Analysis of Genomes and their Expression. Genomics 33:151-152 (1996).


[0163] All references cited herein, including patents, patent applications, and publications, are hereby incorporated by reference in their entireties, whether previously specifically incorporated or not.


[0164] Having now fully described this invention, it will be appreciated by those skilled in the art that the same can be performed within a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention and without undue experimentation.


[0165] While this invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications. This application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth.


Claims
  • 1. An array comprising polynucleotide probes, capable of hybridizing to nucleic acid molecules of more than one of the genes listed in Table 2, hybridized to nucleic acids derived from a cell suspected of being non-normal.
  • 2. The array of claim 1 comprising 3-11 of the genes.
  • 3. The array of claim 2 comprising 5-11 of the genes.
  • 4. The array of claim 3 comprising all 11 genes.
  • 5. The array of claim 1 wherein said cell is from a subject afflicted with, or suspected of having, breast cancer.
  • 6. The array of claim 1 wherein said cell is a human cell.
  • 7. The array of claim 1 wherein said nucleic acids derived from a cell are prepared by quantitative PCR.
  • 8. The array of claim 1 wherein at least one polynucleotide probe hybridizes to CRIP1 nucleic acid molecules.
  • 9. The array of claim 1 wherein said non-normal cells are selected from ADH, DCIS, and IDC.
  • 10. An array comprising polynucleotide probes, capable of hybridizing to nucleic acid molecules of more than one of the genes in Table 3, hybridized to nucleic acids derived from a cell suspected of being DCIS or IDC.
  • 11. The array of claim 10 comprising 3-9 of the genes.
  • 12. The array of claim 11 comprising 5-9 of the genes.
  • 13. The array of claim 12 comprising all 9 genes.
  • 14. The array of claim 10 wherein said cell is from a subject afflicted with, or suspected of having, breast cancer.
  • 15. The array of claim 10 wherein said cell is a human cell.
  • 16. The array of claim 10 wherein said nucleic acids derived from a cell are prepared by quantitative PCR.
  • 17. An array comprising polynucleotide probes, capable of hybridizing to nucleic acid molecules of more than one of the genes in Table 4, hybridized to nucleic acids derived from a cell suspected of being non-normal.
  • 18. The array of claim 17 comprising 3-9 of the genes.
  • 19. The array of claim 18 comprising 5-9 of the genes.
  • 20. The array of claim 19 comprising all 9 genes.
  • 21. The array of claim 17 wherein said cell is from a subject afflicted with, or suspected of having, breast cancer.
  • 22. The array of claim 17 wherein said cell is a human cell.
  • 23. The array of claim 17 wherein said nucleic acids derived from a cell are prepared by quantitative PCR.
  • 24. The array of claim 17 wherein at least one polynucleotide probe hybridizes to ELF5 nucleic acid molecules.
  • 25. The array of claim 17 wherein said non-normal cells are selected from ADH, DCIS, and IDC
  • 26. A method to determine the presence, grade or stage of breast cancer cells in a sample from a subject comprising assaying said sample for expression of one or more genes in Table 2 and/or one or more genes in Table 4.
  • 27. The method of claim 26 wherein said assaying comprises preparing RNA from said sample.
  • 28. The method of claim 27 wherein said RNA is used for quantitative PCR.
  • 29. The method of claim 28 wherein said assaying comprises using an array.
  • 30. The method of claim 26 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 31. The method of claim 30 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 32. The method of claim 26 wherein said non-normal cells are selected from ADH, DCIS, and IDC.
  • 33. A method to determine the presence, grade or stage of breast cancer cells in a sample from a subject comprising assaying said sample for expression of one or more genes selected from Table 2 and/or Table 4 and identifying said sample as containing non-normal breast cancer cells, optionally without grading by histomorphological criteria.
  • 34. The method of claim 33 wherein said assaying comprises preparing RNA from said sample.
  • 35. The method of claim 34 wherein said RNA is used for quantitative PCR.
  • 36. The method of claim 33 wherein said assaying comprises using an array.
  • 37. The method of claim 33 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 38. The method of claim 37 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 39. The method of claim 33 wherein said non-normal cells are selected from ADH, DCIS, and IDC.
  • 40. A method to determine the presence of DCIS and/or IDC breast cancer cells of grade III in a sample from a subject comprising assaying said sample for expression of one or more genes selected from Table 3, Table 6, and/or Table 9 and identifying said sample as containing breast cancer cells of grade III, optionally without grading by histomorphological criteria.
  • 41. The method of claim 40 wherein said assaying comprises preparing RNA from said sample.
  • 42. The method of claim 41 wherein said RNA is used for quantitative PCR.
  • 43. The method of claim 40 wherein said assaying comprises using an array.
  • 44. The method of claim 40 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 45. The method of claim 44 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 46. A method to determine the grade of either DCIS or IDC cancer progression in a sample from a subject comprising assaying said sample for expression of one or more genes listed in Table 10.
  • 47. The method of claim 46 wherein said assaying comprises preparing RNA from said sample.
  • 48. The method of claim 47 wherein said RNA is used for quantitative PCR.
  • 49. The method of claim 46 wherein said assaying comprises using an array.
  • 50. The method of claim 49 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 51. The method of claim 50 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 52. A method to determine the presence of DCIS and/or IDC breast cancer cells of grade I in a sample from a subject comprising assaying said sample for expression of one or more genes selected from Table 7 and/or Table 8 and identifying said sample as containing breast cancer cells of grade I, optionally without grading by histomorphological criteria.
  • 53. The method of claim 52 wherein said assaying comprises preparing RNA from said sample.
  • 54. The method of claim 53 wherein said RNA is used for quantitative PCR.
  • 55. The method of claim 52 wherein said assaying comprises using an array.
  • 56. The method of claim 55 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 57. The method of claim 56 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 58. A method to determine the presence of DCIS and/or IDC breast cancer cells of grade I and/or grade III in a sample from a subject comprising assaying said sample for decreased expression of one or more genes selected from Table 5.
  • 59. The method of claim 58 wherein said assaying comprises preparing RNA from said sample.
  • 60. The method of claim 59 wherein said RNA is used for quantitative PCR.
  • 61. The method of claim 58 wherein said assaying comprises using an array.
  • 62. The method of claim 58 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 63. The method of claim 62 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 64. An array comprising polynucleotide probes, capable of hybridizing to nucleic acid molecules of more than one of the genes listed in Table 5, hybridized to nucleic acids derived from a cell suspected of being DCIS or IDC.
  • 65. The array of claim 41 comprising 3-11 of the genes.
  • 66. The array of claim 64 wherein said cell is from a subject afflicted with, or suspected of having, breast cancer.
  • 67. The array of claim 64 wherein said cell is a human cell.
  • 68. The array of claim 64 wherein said nucleic acids derived from a cell are prepared by quantitative PCR.
  • 69. An array comprising polynucleotide probes, capable of hybridizing to nucleic acid molecules of more than one of the genes listed in Table 6 and/or Table 9, hybridized to nucleic acids derived from a cell suspected of being grade III DCIS and/or IDC.
  • 70. The array of claim 69 comprising 3-11 of the genes.
  • 71. The array of claim 69 wherein said cell is from a subject afflicted with, or suspected of having, breast cancer.
  • 72. The array of claim 69 wherein said cell is a human cell.
  • 73. The array of claim 69 wherein said nucleic acids derived from a cell are prepared by quantitative PCR.
  • 74. An array comprising polynucleotide probes, capable of hybridizing to nucleic acid molecules of more than one of the genes listed in Table 7 and/or Table 8, hybridized to nucleic acids derived from a cell suspected of being grade I DCIS and/or IDC.
  • 75. The array of claim 74 comprising 3-11 of the genes.
  • 76. The array of claim 74 wherein said cell is from a subject afflicted with, or suspected of having, breast cancer.
  • 77. The array of claim 74 wherein said cell is a human cell.
  • 78. The array of claim 74 wherein said nucleic acids derived from a cell are prepared by quantitative PCR.
  • 79. An array comprising polynucleotide probes, capable of hybridizing to nucleic acid molecules of more than one of the genes listed in Table 10, hybridized to nucleic acids derived from a cell suspected of being DCIS or IDC.
  • 80. The array of claim 79 comprising 3-11 of the genes.
  • 81. The array of claim 79 wherein said cell is from a subject afflicted with, or suspected of having, breast cancer.
  • 82. The array of claim 79 wherein said cell is a human cell.
  • 83. The array of claim 79 wherein said nucleic acids derived from a cell are prepared by quantitative PCR.
  • 84. A method to determine therapeutic treatment for a patient having cells suspected of being non-normal in a sample therefrom comprising identifying said patient as having grade III DCIS and/or IDC after assaying said cells for expression of more than one gene listed in Table 3 and selecting the appropriate treatment for a patient having cells of such a grade.
  • 85. The method of claim 84 wherein said assaying comprises preparing RNA from said cells.
  • 86. The method of claim 85 wherein said RNA is used for quantitative PCR.
  • 87. The method of claim 84 wherein said assaying comprises using an array.
  • 88. The method of claim 87 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 89. The method of claim 88 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 90. The method of claim 84 wherein said non-normal cells are selected from ADH, DCIS, and IDC.
  • 91. A method to determine therapeutic treatment for a patient having non-normal cells in a sample therefrom comprising identifying said patient as having grade III DCIS and/or IDC after assaying said cells for expression of more than one gene listed in Table 6 and/or Table 9, and selecting the appropriate treatment for a patient having cells of such a grade.
  • 92. The method of claim 91 wherein said assaying comprises preparing RNA from said cells.
  • 93. The method of claim 92 wherein said RNA is used for quantitative PCR.
  • 94. The method of claim 91 wherein said assaying comprises using an array.
  • 95. The method of claim 91 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 96. The method of claim 95 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 97. The method of claim 91 wherein said non-normal cells are selected from ADH, DCIS, and IDC.
  • 98. A method to determine therapeutic treatment for a patient having non-normal cells in a sample therefrom comprising identifying said patient as having grade I DCIS and/or IDC after assaying said cells for expression of more than one gene listed in Table 7 and/or Table 8, and selecting the appropriate treatment for a patient having cells of such a grade.
  • 99. The method of claim 98 wherein said assaying comprises preparing RNA from said cells.
  • 100. The method of claim 99 wherein said RNA is used for quantitative PCR.
  • 101. The method of claim 98 wherein said assaying comprises using an array.
  • 102. The method of claim 98 wherein said sample is a ductal lavage or fine needle aspiration sample.
  • 103. The method of claim 102 wherein said sample is microdissected to isolate one or more cells suspected of being breast cancer cells.
  • 104. The method of claim 98 wherein said non-normal cells are selected from ADH, DCIS, and IDC.
RELATED APPLICATIONS

[0001] This application is a continuation-in-part of U.S. patent application Ser. No. 10/028,018 filed Dec. 21, 2001, which is hereby incorporated in its entirety as if fully set forth.

Continuation in Parts (1)
Number Date Country
Parent 10028018 Dec 2001 US
Child 10211015 Aug 2002 US