WO2007061876A2 - Methods and compositions involving intrinsic genes - Google Patents

Methods and compositions involving intrinsic genes Download PDF

Info

Publication number
WO2007061876A2
WO2007061876A2 PCT/US2006/044737 US2006044737W WO2007061876A2 WO 2007061876 A2 WO2007061876 A2 WO 2007061876A2 US 2006044737 W US2006044737 W US 2006044737W WO 2007061876 A2 WO2007061876 A2 WO 2007061876A2
Authority
WO
WIPO (PCT)
Prior art keywords
genes
intrinsic
gene
cancer
expression
Prior art date
Application number
PCT/US2006/044737
Other languages
French (fr)
Other versions
WO2007061876A3 (en
WO2007061876A8 (en
Inventor
Matthew Ellis
Charles M. Perou
Philip Bernard
Robert A. PALAIS
Original Assignee
University Of Utah Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Utah Research Foundation filed Critical University Of Utah Research Foundation
Priority to CA002630974A priority Critical patent/CA2630974A1/en
Priority to US12/094,898 priority patent/US20090299640A1/en
Priority to EP06844413A priority patent/EP1954708A4/en
Publication of WO2007061876A2 publication Critical patent/WO2007061876A2/en
Publication of WO2007061876A3 publication Critical patent/WO2007061876A3/en
Publication of WO2007061876A8 publication Critical patent/WO2007061876A8/en
Priority to US13/959,575 priority patent/US20140087959A1/en
Priority to US15/043,022 priority patent/US20170044618A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • Described herein is a method of diagnosing cancer, the method comprising comparing expression levels of a combination of genes from Table 21 to test nucleic acids wherein specific expression patterns of the test nucleic acids indicates a cancerous state.
  • a method of quantitating level of expression of a test nucleic acid comprising: a) comparing gene expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes; and b) quantitating level of expression of the test nucleic acid.
  • a method for determining prognosis based on the expression patterns in a subject diagnosed with cancer comprising: a) comparing expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes, b) identifying a subtype of cancer ot the subject, and c) prognosis (ie, outcome) and treatment decisions based on the subtype of cancer in the subject.
  • a method of classifying cancer in a subject comprising: a) identifying intrinsic genes of the subject to be used to classify the cancer; b) obtaining a sample from the subject; c) amplifying and detecting levels of intrinsic genes in the subject; and d)classifying cancer or subject based upon results of step c.
  • Also disclosed is a method of diagnosing cancer in a subject the method comprising: a) amplifying and detecting intrinsic genes; and b) diagnosing cancer based on expression levels of the gene within the subject.
  • a method of deriving a minimal intrinsic gene set for making biological classifications of cancer comprising: a) collecting data from multiple samples from the same individual to identify potential intrinsic classifier genes; b) weighting intrinsic classifier genes of multiple individuals identified using the method of step a relative to each other and forming classification clusters; c) estimating the number of clusters formed in step b) and assigning individual samples to classification clusters; d) identifying genes that optimally distinguish the samples in the assigned groups of step c); e) performing iterative cross- validation with a nearest centroid classifier and overlapping gene sets of various sizes using the genes identified in step d); and f) choosing a gene set which provides the highest class prediction accuracy when compared to the classifications made in step b).
  • a method of assigning a sample to an intrinsic subtype comprising a) creating an intrinsic subtype average profile (centroid) for each subtype; b) individually comparing a new sample to each centroid; and c) assigning the new sample to the centroid that is most similar to the expression profile of new sample.
  • Figure 1 shows the expression levels for the five genes shown by tissue sample. Top: raw data. Bottom: log-scale.
  • Figure 2 shows the expression levels of the 10 genes shown by sample and tissue type. Vandesompele data set in log-scale. 13.
  • Figure 3 shows the mean squared error (MSE) of each gene by tissue-type. The sign is determined by the direction of the bias. The MSE is broken down into the contributing components of the squared bias (Bias ⁇ 2) and the variance (Sigma ⁇ 2). Vandesompele data set.
  • MSE mean squared error
  • Figure 4 shows two-way hierarchical clustering of microarray data for the same samples assayed by qRT-PCR. Samples were classified based on the expression of 402 "intrinsic" genes defined in Sorlie et al. 2003. The expression level for each gene is shown relative to the median expression of that gene across all the samples with high expression represented by red and low expression represented by green. Genes with median expression are black and missing values are gray.
  • the sample-associated dendrogram shows the same classes seen by qRT-PCR ( Figure 5). Samples are grouped into Luminal, HER2+/ER-, Normal-like, and Basal-like subtypes. Overall, 114/123 (93%) primary breast samples classified the same between microarray and qRT-PCR.
  • Figure 5 shows two-way hierarchical clustering of real-time qRT-PCR data from 126 unique samples.
  • the sample-associated dendrogram (5A) shows the same classes seen by microarray. Samples are grouped into Luminal (blue), HER2+/ER- (pink), Normal-like (green), and Basal-like (red) subtypes. The expression level for each gene is shown relative to the median expression of that gene across all the samples with high expression represented by red and low expression represented by green. Genes with median expression are black and missing values are gray.
  • a minimal set of 37 "intrinsic" genes (5B) was used to classify tumors into their primary "intrinsic" subtypes.
  • the “intrinsic” gene set was supplemented using PgR and EGFR (5C), and proliferation genes (5D).
  • the genes in 1C and ID were clustered separately in order to determine agreement between the minimal 37 qRT-PCR "intrinsic” set (5A) and the larger 402 microarray "intrinsic” set.
  • Figure 6 shows Receiver Operator Curves.
  • the agreement between immunohistochemistry (IHC) and gene expression is shown for ER (6A), PR (6B), and HER2 (6C) using ROC.
  • a cut-off for relative gene copy number was selected by minimizing the sum of the observed false positive and false negative errors.
  • the sensitivity and specificity of the resulting classification rule were estimated via bootstrap adjustment for optimism. Since many biomarkers having concordant expression and can serve as surrogates for one another, we tested the accuracy of using GATA3 and GRB7 as surrogates (dotted lines) for calling ER and HER2 protein status, respectively. There was overall good agreement between gene expression and IHC status for ER and PR, but poor agreement between gene expression and IHC status for HER2.
  • the surrogate markers had similar accuracy to the actual markers for predicting HiC status.
  • Figure 7 shows outcome for "intrinsic" subtypes.
  • Classifications were made from real-time qRT-PCR data using the minimal 37 "intrinsic" gene list. Pairwise log-rank tests were used to test for equality of the hazard functions among the intrinsic classes. Tumors in the Normal Breast-like subtype were excluded from the analyses since this class maybe artificially created from having a sample comprised primarily of normal cells.
  • Figure 8 shows grade and proliferation as predictors of relapse free survival.
  • Kaplan-Meier plots are shown for grade (8A) and the proliferation genes (8B) using Cox regression analysis.
  • the analysis for the proliferation genes was performed on continuous expression data, although the plots are shown in tertiles.
  • the proliferation index (log average of the 14 proliferation genes) has significant predictive value for outcome, even after correcting for other clinical parameters important for survival.
  • Figure 9 shows co-clustering of real-time qRT-PCR and microarray data using 50 genes and 252 samples.
  • the relative copy number (qRT-PCR) and R/G ratio (microarray) for each gene was Iog2 transformed and combined into a single dataset using distance weighted discrimination. Two-way hierarchical clustering was performed on the combined dataset using Spearman correlation and average linkage.
  • the sample associated dendrogram (5A) shows the same classes as seen in Figure 1. Samples are classified as Basal-like (red), HER2+/ER-, Luminal, and Normal-like. The expression level for each gene is shown relative to the median expression of that gene across all the samples with overexpressed genes and underexpressed genes, as well as average expression.
  • the gene associated dendrogram (5B) shows that the Luminal tumors and Basal-like tumors differentially express estrogen associated genes (cluster 1); as well as basal keratins (KRT 5 and 17), inflammatory response genes (CX3CL1 and SLPI), and genes in the Wnt pathway (FZD7) (cluster 3).
  • the main distinguishers of the HER2+/ER- group are low expression of genes in cluster 1 and high expression of genes on the 1/q12 amplicon (ERBB2 and GRB7) (cluster 4).
  • the proliferation genes (cluster 2) have high expression in the ER negative tumors (Basal-like and HER2+/ER-) and low expression in ER positive (Luminal) and Normal-like samples.
  • Figure 10 shows a flow chart of the steps of deriving minimal intrinsic gene sets for making biological classifications of breast cancer.
  • Figure 11 shows an overview and flow of the data sets used and analyses performed.
  • Figure 12 shows a hierarchical cluster analysis of the training set using the Intrinsic/UNC gene set.
  • 146 microarrays representing 105 tumors and 9 normal breast samples were analyzed using the 1300 gene Intrinsic/UNC gene set.
  • A) Overview of the complete cluster diagram (the full cluster diagram can be found as Supplemental Figure 1).
  • F Proliferation associated expression cluster. The genes in red are mentioned in the text.
  • Figure 13 shows Androgen Receptor (AR) immunohistochemistry on human breast tumors.
  • Figure 14 shows hierarchical cluster analysis the combined test set of 311 tumors and 4 normal breast samples analyzed using the Intrinsic/UNC gene set reduced to 306 genes.
  • Figure 15 shows univariate Kaplan-Meier survival plots using RFS as the endpoint, for the common clinical parameters present within the combined test set of 311 tumors. Survival plots for A) ER status, B) node status, C) grade, and D) tumor size. Kaplan-Meier survival plots for intrinsic subtype analyses. A) Relapse-free survival for the 105 patients/tumors training set classified using hierarchical clustering and complete 1300 gene the Litrinsic/UNC list. B) Relapse-free survival for the 315 sample combined test set analyzed using the Intrinsic/UNC list reduced to 306 genes. C) Survival analysis of the 60 adjuvant tamoxifen-treated patients from the Ma et al.
  • Figure 17 shows grade and proliferation as predictors of relapse free survival.
  • a Cox regression model was used to determine probability of relapse over time.
  • Kaplan-Meier curves show time to event given different grades and levels of proliferation.
  • Grade was scored as low (green), medium (red) or high (blue).
  • the proliferation score was based on continuous expression data and is shown as textiles that correspond to low (green), medium (red), and high (blue) levels of expression.
  • the proliferation meta-gene (Iog2 average of the 14 proliferation genes) showed significant value in predicting relapse, even after correcting for other clinical parameters important for survival (Table 1).
  • Figure 18 shows an agreement plot between fresh frozen (FF) and formalin-fixed paraffin-embedded (FFPE) for the estrogen receptor gene (ESRl) after normalization to the 5 housekeepers.
  • Figure 19 shows line graphs which show the effects at each step of data processing.
  • the raw (pre-normalized) data shows a negative bias for all genes likely due to lower RNA quality in the FFPE tissue (Fig 2A).
  • Much of the bias was corrected by normalization to the 'housekeeper' genes and using DWD adjustment. As expected, DWD had a significant effect on bias (m) but did not effect other measurements of agreement (Fig 2B-D).
  • Figure 20 shows a large dynamic range of ESRl expression provides clear separation of the tumors from both FF and FFPE.
  • Figure 21 shows a scatter plot of ER status by immunohistochemistry versus expression or ESRl.
  • Figure 22 shows a scatter plot of average copy number in an formalin-fixed paraffin embedded (FFPE) sample versus a fresh frozen (FF) sample. The results are normalized to all five housekeepers.
  • FFPE formalin-fixed paraffin embedded
  • Figure 23 shows a scatter plot of average copy number in an formalin-fixed paraffin embedded (FFPE) sample versus a fresh frozen (FF) sample. The results are after DWD.
  • FFPE formalin-fixed paraffin embedded
  • Figure 24 shows hierarchical clustering of the 124 sample training set using the "intrinsic" gene set identified in Hu et al, which shows 4 distinct classes representing Luminal, HER2+/ER-, Basal-like, and Normal-like.
  • Figure 25 shows MA P3m test (4Og X 35s) gene scores, as well as FF PCR test (4Og X 35s) gene scores for for basal, HER-2, luminal and normal like.
  • Ranges can be expressed herein as from “about” one particular value, and/or to "about” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10" is also disclosed.
  • the "subject” can include, for example, domesticated animals, such as cats, dogs, etc., livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), laboratory animals (e.g., mouse, rabbit, rat, guinea pig, etc.) mammals, non-human mammals, primates, non-human primates, rodents, birds, reptiles, amphibians, fish, and any other animal.
  • livestock e.g., cattle, horses, pigs, sheep, goats, etc.
  • laboratory animals e.g., mouse, rabbit, rat, guinea pig, etc.
  • mammals non-human mammals, primates, non-human primates, rodents, birds, reptiles, amphibians, fish, and any other animal.
  • the subject can be a mammal such as a primate or a human.
  • Treating does not mean a complete cure. It means that the symptoms of the underlying disease are reduced, and/or that one or more of the underlying cellular, physiological, or biochemical causes or mechanisms causing the symptoms are reduced. It is understood that reduced, as used in this context, means relative to the state of the disease, including the molecular state of the disease, not just the physiological state of the disease.
  • reduce or other forms of reduce means lowering of an event or characteristic. It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative value to be referred to.
  • reduceds phosphorylation means lowering the amount of phosphorylation that takes place relative to a standard or a control.
  • inhibit or other forms of inhibit means to hinder or restrain a particular characteristic. It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative Valueto ⁇ e 11 referred to':' 1
  • inhibits phosphorylation means ndering or restraining the amount of phosphorylation that takes place relative to a standard or a control.
  • prevent or other forms of prevent means to stop a particular characteristic or condition. Prevent does not require comparison to a control as it is typically more absolute than, for example, reduce or inhibit. As used herein, something could be reduced but not inhibited or prevented, but something that is reduced could also be inhibited or prevented. It is understood that where reduce, inhibit or prevent are used, unless specifically indicated otherwise, the use of the other two words is also expressly disclosed. Thus, if inhibits phosphorylation is disclosed, then reduces and prevents phosphorylation are also disclosed.
  • the term "therapeutically effective" means that the amount of the composition used is of sufficient quantity to ameliorate one or more causes or symptoms of a disease or disorder.
  • carrier means a compound, composition, substance, or structure that, when in combination with a compound or composition, aids or facilitates preparation, storage, administration, delivery, effectiveness, selectivity, or any other feature of the compound or composition for its intended use or purpose.
  • a carrier can be selected to minimize any degradation of the active ingredient and to minimize any adverse side effects in the subject.
  • cell as used herein also refers to individual cells, cell lines, or cultures derived from such cells.
  • a “culture” refers to a composition comprising isolated cells of the same or a different type.
  • an Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound.
  • a weight percent of a component is based on the total weight of the formulation or composition in which the component is included.
  • Primers are a subset of probes which are capable of supporting some type of enzymatic manipulation and which can hybridize with a target nucleic acid such that the enzymatic manipulation can occur.
  • a primer can be made from any combination of nucleotides or nucleotide derivatives or analogs available in the art which do not interfere with the enzymatic manipulation.
  • Probes are molecules capable of interacting with a target nucleic acid, typically in a sequence specific manner, for example through hybridization. The hybridization of nucleic acids is well understood in the art and discussed herein. Typically a probe can be made from any combination of nucleotides or nucleotide derivatives or analogs available in the art.
  • the methodology described herein can be used to make a classification that distinguishes 2 or more intrinsic subtypes of breast cancer.
  • the intrinsic subtypes can be classes therein), HER2 ER- an c asses t ere n , Basa an classes therein), Normal-like (and classes therein).
  • the steps for finding the minimal intrinsic gene set for making subtype (and class) distinctions are as follows.
  • the first step is to use microarray data from biological replicates from the same patient to find intrinsic classifier genes.
  • a data set of tumors and normal breast samples can be used, hi one embodiment, these data sets can comprise paired biological replicates to identify the intrinsic gene set. This is described, for example, in Perou et al. (2000), which is herein incorporated by reference in its entirety for its teaching regarding finding intrinsic classifier genes.
  • Perou et al. the molecular portraits revealed in the patterns of gene expression not only uncovered similarities and differences among the tumors, but also point to a biological interpretation. Variation in growth rate, in the activity of specific signalling pathways, and in the cellular composition of the tumors were all reflected in the corresponding variation in the expression of specific subsets of genes.
  • hierarchical cluster microarray data was obtained using an intrinsic gene set.
  • data can be combined from different microarray platforms for clustering using methods described in Example 2.
  • the "intrinsic gene set" from the first step (above) is tested on new tumors and normal breast samples after combining different datasets (such as cross platform analyses) and common genes/elements are hierarchically clustered.
  • a two-way average linkage hierarchical cluster analysis can be performed using a centered Pearson correlation metric and the program "Cluster" (Eisen et al. 1998), with the data being displayed relative to the median expression for each gene (i.e. median centering of the rows/genes).
  • the number of clusters formed in the microarray dataset is estimated, and samples/tumors are assigned to clusters based on the sample-associated dendrogram groupings.
  • the "test set” is used as a training set to create subtype centroids based upon the expression of the common intrinsic genes. New samples are assigned to the subtype corresponding to the nearest centroid when using Spearman correlation values.
  • genes are found that optimally distinguish the samples in the assigned groups using the ratio of between-group to within-group sums of squares (the entire microarray dataset is used in this analysis).
  • An example of this can be found in Chung et al, Cancer Cell 2004, herein incorporated by reference in its entirety for its teaching concerning identification of genes that optimally distinguish samples.
  • '3 1 S: 1 in the!' f ⁇ ftlfstep iterative cycles of 10-fo cross-va at on are per orme w t a nearest centroid classifier and overlapping gene sets of varying sizes. In other words, each gene and gene set are ranked based upon the metric from step four above, and various overlapping and every increasing sized genes lists are used in a 10-fold cross validation.
  • the smallest gene set which provides the highest class prediction accuracy when compared to the classifications made by the complete microarray- based intrinsic gene set is chosen. Subtypes are assigned for each gene set and the minimal gene set with the highest agreement in sample assignment to the full intrinsic gene set is chosen, hi one example, using a 1410 intrinsic gene set as disclosed in Example 2, 100 genes were identified (see Table 12 (7p 100), after the "Examples" section) that are important for identifying 7 different biological classes of breast cancer. Specific steps and sample sets used to develop the 7-class predictor as shown in Figure 11. Also disclosed in Table 13 is an extended list of genes for classification resulting from the 7p analyses. This list is ranked in terms of significance for separating the different classes of intrinsic classifier genes.
  • Another set of intrinsic genes that can be used for classification is found in Table 21, along with the primers that can be used to amplify those genes. It should be noted that the primers are optional and exemplary only, as any primer that can amplify a given gene can be used.
  • the minimal intrinsic gene set (identified using the methods described above, and found in Tables 12 and 13) has prognostic and predictive significance in breast cancer.
  • the complete assay for making these biological "intrinsic” classifications includes 3 "housekeeper” genes (MRPLl 9, PUMl, and PSMC4) for normalizing the quantitative data.
  • proliferation genes can also be used in combination with the housekeeper genes for providing a quantitative measurement of grade and for assessing prognosis in breast cancer.
  • SSP Single Sample Predictor
  • the Single Sample Predictor/SSP is based upon the Nearest Centroid method presented in (Hastie et al. 2001).
  • the subtype centroids can be used to make subtype predictions on additional test sets (e.g., homogenously treated subjects from clinical trial groups).
  • the resulting classifications are then analyzed using Kaplan-Meier survival plots to determine prognostic and therapeutic significance.
  • An example of SSP can be found in Example 2. 1. Intrinsic genes and cancer
  • An intrinsic gene is a gene that shows little variance within repeated samplings of the same tumor, but which shows high variance across tumors. Disclosed herein are genes that can be used as intrinsic genes with the methods disclosed herein. The intrinsic genes disclosed herein can be genes that have less than or equal to 0.00001, 0.0001, 0.001, 0.01, 0.1, 0.2.
  • these levels of variation can also be applied across 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 or more tissues, and the level of variation compared. It is also understood that variation can be determined as discussed in the examples using the algorithms as disclosed herein.
  • Intransic gene set is defined herein as comprising one or more intrinsic genes.
  • Minimal intrinsic gene set is defined herein as being derived from an intrinsic gene set, and is considered the fewest number of intrinsic genes that can be used to classify a sample.
  • a set of 212 minimal intrinsic genes as found in Table 21. These genes can be used alone, or in combination, as intrinsic genes for the purposes of classification, prognosis, and diagnosis of cancer, for example. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99
  • Described herein is a method of diagnosing cancer, the method comprising comparing expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes, w ere n spec c express on patterns o t e test nucleic acids indicates a cancerous state.
  • Also disclosed is a method of quantitating level of expression of a test nucleic acid comprising: a) comparing gene expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes; and b) quantitating level of expression of the test nucleic acid.
  • a method of prognosing outcome in a subject diagnosed with cancer comprising: a) comparing expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes, b) identifying a subtype of cancer of the subject, and c) prognosing the outcome based on the subtype of cancer of the subject.
  • the intrinsic genes disclosed herein can be normalized to control housekeeper genes and used in a qRT-PCR diagnostic assay that uses relative copy number to assess risk or therapeutic response in cancer.
  • MRPL19 SEQ ID NO:1
  • PSMC4 SEQ ID NO:2
  • SF3A1 SEQ IDNO:3
  • PUMl SEQ ID NO:4
  • ACTB SEQ ID NO:5
  • GAPD GAPD
  • Other genes include GUSB, RPLPO, and TFRC, whose sequences can be found in Geribank. These are part of the 212 gene list.
  • Other genes as disclosed herein can also be considered intrinsic genes.
  • the intrinsic genes can be used in any combination or singularly in any method described herein. It is also understood that any nucleic acid related to the expression control genes, such as the RNA, mRNA, exons, introns, or 5' or 3' upstream or downstream sequence, or DNA or gene can be used or identified in any of the methods or with any of the compositions disclosed herein.
  • the disclosed methods involve using specific intrinsic genes or gene sets or expression control genes or gene sets such that they are detected in some way or their expression product is detected in some way.
  • the expression of a gene or its expression product will be detected by a primer or probe as disclosed herein.
  • the expression of the genes of interest can be detected after or during an amplification process, such as RT-PCR, including quantitative PCR. 3.
  • Microarrays have shown that gene expression patterns can be used to molecularly classify various types of cancers into distinct and clinically significant groups.
  • a microarray breast cancer classification system has been recapitulated using real-time quantitative (q)RT-PCR (Example 2).
  • q real-time quantitative
  • Statistical analyses were performed on multiple independent microarray datasets to select an "intrinsic" gene set that can classify breast tumors into four different subtypes designated as Luminal, Normal-like, HER2+/ER-, and Basal-like. Intrinsic genes, as described in Perou et al.
  • intrinsic genes are the classifier genes for breast cancer classification and each classifier gene can be normalized to the housekeeper (or control) genes in order to make the classification.
  • the expression data and classifications from microarray and real-time qRT-PCR were respectively compared using 123 unique breast samples (117 invasive carcinomas, 1 fibroadenoma and 5 normal tissues) and 3 cells lines.
  • the overall correlation for the 50 genes in common between microarray and qRT-PCR was 0.76.
  • Example 2 illustrates how intrinsic gene sets can be minimized from microarray data and used on fresh tissue in a qRT-PCR assay to recapitulate the microarray classifications. It also shows the importance of the 'proliferation' genes in risk stratifying Luminal (ER+) breast tumors.
  • Example 3 discusses a version of the intrinsic gene set from Hu et a an s ows again ow t can be minimized to provide intrinsic classifications on both fresh and FFPE tissue and using microarray or qRT-PCR data. Validated primer sequences from FFPE tissues for 212 genes important for breast cancer diagnostics are presented in Table 21.
  • TNM staging system provides information about the extent of disease and has been the "gold standard" for prognosis (Henson, et al. (1991) Cancer 68:2142-2149; Fitzgibbons, et al (2000) Arch Pathol Lab Med 124:966-978).
  • the grade of the tumor is also prognostic for relapse free survival (RFS) and overall survival (OS) (Elston et al. (1991) Histopathology 19:403-410). Tumor grade is determined from histological assessment of tubule formation, nuclear pleomorphism, and mitotic count. Due to the subjective nature of grading and difficulties standardizing methods, there has been less than optimal agreement between pathologists (Dalton et al. (1994) Cancer 73:2765-2770).
  • proliferation assays such as S-phase fraction and mitotic index
  • S-phase fraction and mitotic index have shown to be independent prognostic indicators and could be used in conjunction with, or instead of grade.
  • proliferation genes can be used in a qRT-PCR assay and the genes can be averaged to produce a proliferation meta-gene that correlates with grade but is more prognostic (Figure 17).
  • ER expression is a predictive marker, it also serves as a surrogate marker for describing a tumor biology that is characteristically less aggressive (e.g. lower grade) than ER-negative tumors (Fisher et al. (1981) Breast Cancer Res Treat 1:37-41).
  • Microarrays have elucidated the richness and diversity in the biology of breast cancer and have identified many genes that associate with ER-positive and ER-negative tumors (Perou et al. (2000) Nature 406:747-752; West et al. (2001) Proc Natl Acad Sci U S A 98:11462-11467; Gruvberger et al. (2001) Cancer Res 61:5979-5984).
  • samples are separated primarily based on ER status (Sotiriou et al. (2003) Proc Natl Acad Sci U S A 100:10393-10398).
  • Breast tumors of the "Luminal" subtype are ER positive and have a similar keratin expression profile as the epithelial cells lining the lumen of the breast ducts (Taylor- Papadimitriou et al. (1989) J Cell Sci 94:403-413; Perou et al. (2000) New Technologies for life sciences: A Trends Guide:67-76).
  • ER-negative tumors can be broken into two main subtypes, namely those that overexpress (and are DNA amplified for) HER2 and GRB7 (HER2+/ER-), and "Basal-like" tumors that have an expression profile similar to basal epithelium and express Keratin 5, 6B and 17.
  • Luminal tumors are aggressive and typically more deadly than Luminal tumors; however, there are subtypes of Luminal tumors that lead to poor outcome despite being ER-positive. For instance, Sorlie et al. identified a Luminal B subtype with similar outcomes to the HER2+/ER- and Basal-like subtypes, and Sotiriou et al. showed that there are 3 different types of Luminal tumors with different outcomes. The Luminal tumors with poor outcomes consistently share the histopathological ieattr ⁇ Wbeing Higher" grade and the molecular feature of highly expressing proliferation genes.
  • proliferation genes show periodicity in expression through the cell cycle and have a variety of functions necessary for cell growth, DNA replication, and mitosis (Whitfield et al. (2002) MoI Biol Cell 13:1977-2000; Ishida et al. MoI Cell Biol 21:4684- 4699). Despite their diverse functions, proliferation genes have similar gene expression profiles when analyzed by hierarchical clustering. As might be expected, proliferation genes correlate with grade, the mitotic index ( Perou et al. (1999) Proc Natl Acad Sci U S A 96:9212- 9217), and outcome ( S ⁇ rlie et al. (2001) Proc Natl Acad Sci U S A 98:10869-10874).
  • Proliferation genes are often selected when supervised analysis is used to find genes that correlate with patient outcome. For example, the SAM264 "survival" list presented in Sorlie et al., the 231 “prognosis classifier” list in van't Veer et al., and the “485 prognostic gene” list in Sotiriou et al., identified common proliferation genes (PCNA, TOP2A, CENPF). This suggests that all these studies are likely tracking a similar phenotype.
  • Microarray used in conjunction with RT-PCR provides a powerful system for discovering and translating genomic markers into the clinical laboratory for molecular diagnostics. Although these platforms are fundamentally very different, the quantitative data across the methods have a high correlation. In fact, the data across the methods is no more disparate then across different microarray platforms.
  • hierarchical clustering it has been shown that a biological classification of breast cancer derived from microarray data can be recapitulated using real-time qRT-PCR.
  • Biological classification by real-time qRT-PCR makes the important clinical distinction between ER positive and ER negative tumors and identifies additional subtypes that have prognostic (ie, correlate to outcome) and predictive value (ie, correlate to treatment response).
  • the benefit of using real-time qRT-PCR for cancer diagnostics is that new informative markers can be readily validated and implemented, making tests expandable and/or tailored to the individual. For instance, it has been shown that including proliferation genes serves a similar purpose to grade but is more prognostic. Since grade has been shown to be universal as a prognostic factor in cancer, it is likely that the same markers correlate to grade and are important for survival in other tumor types.
  • Real-time qRT-PCR is attractive for clinical use because it is fast, reproducible, tissue sparing, and able to be automated.
  • genomic profiling should currently be used for ancillary testing, the fact that normal tissues can be distinguished from tumor tissue shows that these molecular assays may eventually be used for cancer diagnostics without histological corroboration.
  • a method of classifying cancer in a subject comprising: a) identifying intrinsic genes of the subject to be used to classify the cancer; b) obtaining a sample from the subject; c) amplifying and detecting levels of intrinsic genes in the subject; and d)classifying cancer based upon results of step c.
  • the sample can be fresh, or can be an FFPE sample.
  • the cancer can be breast cancer, for example.
  • the breast cancer can be classified into one of four or more groups: luminal, normal-like, HER2+/ER- and basal-like, for example.
  • the sample can be fresh, or can be an FFPE sample.
  • any method of assaying any gene discussed herein can be performed.
  • methods of assaying gene copy number or mRNA expression copy number can be performed.
  • RT-PCR, PCR, quantitative PCR, and any other forms of nucleic acid amplification can be performed.
  • methods of hybridization such as blotting, such as Northern or Southern techniques, such as chip and microarray techniques and any other techniques involving hybridizing of nucleic acids.
  • compositions can be used to diagnose or prognose any disease where uncontrolled cellular proliferation occurs such as cancers.
  • a non-limiting list of different types of cancers is as follows: lymphomas (Hodgkins and non-Hodgkins), leukemias, carcinomas, carcinomas of solid tissues, squamous cell carcinomas, adenocarcinomas, sarcomas, gliomas, high grade gliomas, blastomas, neuroblastomas, plasmacytomas, histiocytomas, melanomas, adenomas, hypoxic tumours, myelomas, AIDS-related lymphomas or sarcomas, metastatic cancers, or cancers in general.
  • a representative but non-limiting list of cancers that the disclosed compositions can be used to diagnose or prognose is the following: lymphoma, B cell lymphoma, T cell lymphoma, mycosis fungoides, Hodgkin's Disease, myeloid leukemia, bladder cancer, brain cancer, nervous system cancer, head and neck cancer, squamous cell carcinoma of head and neck, kidney cancer, lung cancers such as small cell lung cancer and non-small cell lung cancer, neuroblastoma/glioblastoma, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, liver cancer, melanoma, squamous cell carcinomas of the mouth, throat, larynx, and lung, colon cancer, cervical cancer, cervical carcinoma, breast cancer, and epithelial cancer, renal cancer, genitourinary cancer, pulmonary cancer, esophageal carcinoma, head and neck carcinoma, large bowel cancer, hematopoietic cancers; testicular cancer; colon and rectal cancers, pro
  • Compounds disclosed herein may also be used for the diagnosis or prognosis of precancer conditions such as cervical and anal dysplasias, other dysplasias, severe dysplasias, hyperplasias, atypical hyperplasias, and neoplasias.
  • precancer conditions such as cervical and anal dysplasias, other dysplasias, severe dysplasias, hyperplasias, atypical hyperplasias, and neoplasias.
  • ' f ⁇ " " ⁇ li ⁇ of identifying a minimal intrinsic gene set
  • a minimal intrinsic gene set for making biological classifications of cancer comprising: a) collecting data from multiple samples from the same or different individuals to identify potential intrinsic classifier genes (microarray data can be used in this step, for example); b) weighting intrinsic classifier genes of multiple individuals identified using the method of step a relative to each other and forming classification clusters (weighting can be done, for example, by forming hierarchical clusters); c) estimating the number of clusters formed in step b) and assigning individual samples to clusters; d) identifying genes that optimally distinguish the samples in the assigned groups of step c); e) performing iterative cross-validation with a nearest centroid classifier and overlapping gene sets of various sizes using the genes identified in step d); and f) choosing a gene set which provides the highest class prediction accuracy when compared to the classifications made in step b).
  • a method of assigning a sample to an intrinsic subtype comprising a) creating an intrinsic subtype average profile (centroid) for each subtype; b) individually comparing a new sample to each centroid; and c) assigning the new sample to the centroid that is most similar to the new sample.
  • SSP Single Sample Predictor
  • homology and identity mean the same thing as similarity.
  • the use of the word homology is used between two non-natural sequences it is understood that this is not necessarily indicating an evolutionary relationship between these two sequences, but rather is looking at the similarity or relatedness between their nucleic acid sequences.
  • Many of the methods for determining homology between two evolutionarily related molecules are routinely applied to any two or more nucleic acids or proteins for the purpose of measuring sequence similarity regardless of whether they are evolutionarily related or not.
  • variants of genes and proteins herein disclosed typically have at least, about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent homology to the stated sequence or the native sequence.
  • the homology can be calculated after aligning the two sequences so that the homology is at its highest level.
  • a sequence recited as having a particular percent homology to another sequence refers to sequences that have the recited homology as calculated by any one or more of the calculation methods described above.
  • a first sequence has 80 percent homology, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent homology to the second sequence using the Zuker calculation method even if the first sequence does not have 80 percent homology to the second sequence as calculated by any of the other calculation methods.
  • a first sequence has 80 percent homology, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent homology to the second sequence using both the Zuker calculation method and the Pearson and Lipman calculation method even if the first sequence does not have 80 percent homology to the second sequence as calculated by the Smith and Waterman calculation method, the Needleman and Wunsch calculation method, the Jaeger calculation methods, or any of the other calculation methods.
  • a first sequence has 80 percent homology, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent homology to the second sequence using each of calculation methods (although, in practice, the different calculation methods will often result in different calculated homology percentages). . yb i ization/selective hybridization
  • hybridization typically means a sequence driven interaction between at least two nucleic acid molecules, such as a primer or a probe and a gene.
  • Sequence driven interaction means an interaction that occurs between two nucleotides or nucleotide analogs or nucleotide derivatives in a nucleotide specific manner. For example, G interacting with C or A interacting with T are sequence driven interactions. Typically sequence driven interactions occur on the Watson-Crick face or Hoogsteen face of the nucleotide.
  • the hybridization of two nucleic acids is affected by a number of conditions and parameters known to those of skill in the art. For example, the salt concentrations, pH, and temperature of the reaction all affect whether two nucleic acid molecules will hybridize.
  • selective hybridization conditions can be defined as stringent hybridization conditions.
  • stringency of hybridization is controlled by both temperature and salt concentration of either or both of the hybridization and washing steps.
  • the conditions of hybridization to achieve selective hybridization may involve hybridization in high ionic strength solution (6X SSC or 6X SSPE) at a temperature that is about 12-25°C below the Tm (the melting temperature at which half of the molecules dissociate from their hybridization partners) followed by washing at a combination of temperature and salt concentration chosen so that the washing temperature is about 5 0 C to 2O 0 C below the Tm.
  • the temperature and salt conditions are readily determined empirically in preliminary experiments in which samples of reference DNA immobilized on filters are hybridized to a labeled nucleic acid of interest and then washed under conditions of different stringencies. Hybridization temperatures are typically higher for DNA-RNA and RNA-RNA hybridizations. The conditions can be used as described above to achieve stringency, or as is known in the art. (Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989; Kunkel et al. Methods Enzymol. 1987:154:367, 1987 which is herein incorporated by reference for material at least related to hybridization of nucleic acids).
  • a preferable stringent hybridization condition for a DNA:DNA hybridization can be at about 68 0 C (in aqueous solution) in 6X SSC or 6X SSPE followed by washing at 68°C.
  • Stringency of hybridization and washing if desired, can be reduced accordingly as the degree of complementarity desired is decreased, and further, depending upon the G-C or A-T richness of any area wherein variability is searched for.
  • stringency of hybridization and washing if desired, can be increased accordingly as homology desired is increased, and further, depending upon the G- C or A-T richness of any area wherein high homology is desired, all as known in the art.
  • selective hybridization conditions would be when at least about, 60, 65, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 percent of the limiting nucleic acid is bound to the non-limiting nucleic acid.
  • the non-limiting primer is in for example, 10 or 100 or 1000 fold excess.
  • This type of assay can be performed at under conditions where both the limiting and non-limiting primer are for example, 10 fold or 100 fold or 1000 fold below their k d , or where only one of the nucleic acid molecules is 10 fold or 100 fold or 1000 fold or where one or both nucleic acid molecules are above their k d .
  • selective hybridization conditions are when at least about, 60, 65, 70, 71, 72, 73, 74, 75, 76, 77,
  • Preferred conditions also include those suggested by the manufacturer or indicated in the art as being appropriate for the enzyme performing the manipulation.
  • nucleic acid based there are a variety of molecules disclosed herein that are nucleic acid based, including for example the nucleic acids that encode, for example, the intrinsic genes disclosed herein (Table 12), as well as various functional nucleic acids.
  • the disclosed nucleic acids are made up of for example, nucleotides, nucleotide analogs, or nucleotide substitutes. Non- limiting examples of these and other molecules are discussed herein. It is understood that for example, when a vector is expressed in a cell, that the expressed mRNA will typically be made up of A, C, G, and U.
  • an antisense molecule is introduced into a cell or cell environment through for example exogenous delivery, it is advantagous that the antisense molecule be made up of nucleotide analogs that reduce the degradation of the antisense molecule in the cellular environment.
  • a nucleotide is a molecule that contains a base moiety, a sugar moiety and a phosphate moiety. Nucleotides can be linked together through their phosphate moieties and sugar moieties creating an internucleoside linkage.
  • the base moiety of a nucleotide can be adenin-9-yl (A), cytosin-1-yl (C), guanin-9-yl (G), uracil-1-yl (U), and thymin-1-yl (T).
  • the sugar moiety of a nucleotide is a ribose or a deoxyribose.
  • the phosphate moiety of a nucleotide is pentavalent phosphate.
  • An non-limiting example of a nucleotide would be 3'- AMP (3'-adenosine monophosphate) or 5'-GMP (5'-guanosine monophosphate).
  • primers and probes can be produced for the actual gene (DNA) or expression product (mRNA) or intermediate expression products which are not fully processed into mRNA. Discussion of a particular gene is also a disclosure of the DNA, mRNA, and intermediate RNA products associated with that particular gene.
  • compositions including primers and probes which are capable of interacting with the intrinsic genes disclosed herein, as well as the any other genes or nucleic acids discussed herein, hi certain embodiments the primers are used to support DNA amplification reactions.
  • the primers will be capable of being extended in a sequence specific manner.
  • Extension of a primer in a sequence specific manner includes any methods wherein the sequence and/or composition of the nucleic acid molecule to which the primer is hybridized or otherwise associated directs or influences the composition or sequence of the product produced by the extension of the primer.
  • Extension of the primer in a sequence specific manner therefore includes, but is not limited to, PCR, DNA sequencing, DNA extension, DNA polymerization, RNA transcription, or reverse transcription.
  • the primers are used for the DNA amplification reactions, such as PCR or direct sequencing. It is understood that in certain embodiments the primers can also be extended using non-enzymatic techniques, where for example, the nucleotides or oligonucleotides used to extend the primer are modified such that they will chemically react to extend the primer in a sequence specific manner.
  • the disclosed primers hybridize with the disclosed genes or regions of the disclosed genes or they hybridize with the complement of the disclosed genes or complement of a region of the disclosed genes.
  • the size of the primers or probes for interaction with the disclosed genes in certain embodiments can be any size that supports the desired enzymatic manipulation of the primer, such as DNA amplification or the simple hybridization of the probe or primer.
  • a typical disclosed primer or probe would be at least 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 61, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97,
  • the disclosed primers or probes can be less than or equal to 6, 7, 8, 9, 10, 11, 12 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475,
  • the primers for the disclosed genes in certain embodiments can be used to produce an amplified DNA product that contains the desired region of the disclosed genes.
  • typically the size of the product will be such that the size can be accurately determined to within 10, 5, 4, 3, or 2 or 1 nucleotides. 1 10.
  • this product is at least 20, 21, 22, 23, 24, 25, 27, 2 8 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 61, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950,
  • the product is less than or equal to 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 61, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850,
  • primers and probes are designed such that they are targeting as specific region in one of the genes disclosed herein. It is understood that primers and probes having an interaction with any region of any gene disclosed herein are contemplated: In other words, primers and probes of any size disclosed herein can be used to target any region specifically defined by the genes disclosed herein. Thus, primers and probes of any size can begin hybridizing with nucleotide 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or any specific nucleotide of the genes or gene expression products disclosed herein. Furthermore, it is understood that the primers and probes can be of a contiguous nature meaning that they have continuous base pairing with the target nucleic acid for which they are complementary.
  • primers and probes which are not contiguous with their target complementary sequence.
  • primers and probes which have at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 500, or more bases which are not contiguous across the length of the primer or probe.
  • primers and probes which have less than or equal to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 500, or more bases which are not contiguous across the length of the primer or probe.
  • the primers or probes are designed such that they are able to hybridize specifically with a target nucleic acid.
  • Specific hybridization refers to the ability to bind a particular nucleic acid or set of nucleic acids preferentially over other nucleic acids.
  • the level of specific hybridization of a particular probe or primer with a target nucleic acid can be affected by salt conditions, buffer conditions, temperature, length of time of hybridization, wash conditions, and visualization conditions.
  • By increasing the specificity of hybridization means decreasing the number of nucleic acids that a given primer or probe hybridizes to typically under a given set of conditions. For example, at 20 degrees Celsius under a given set of conditions a given probe may hybridize with 10 nucleic acids in a sample.
  • a decrease in specificity of hybridization means an increase in the number of nucleic acids that a given primer or probe hybridizes to typically under a given set of conditions. For example, at 700 mM NaCl under a given set of conditions a particular probe or primer may hybridize with 2 nucleic acids in a sample, however when the salt concentration is increased to 1 Molar NaCl the primer or probe may hybridize with 6 nucleic acids in the same sample.
  • the salt can be any salt such as those made from the alkali metals: Lithium, Sodium, Potassium, Rubidium, Cesium, or Francium or the alkaline earth metals: Beryllium, Magnesium, Calcium, Strontium, Barium, or Radiumsodium, or the transition metals: Scandium, Titanium, Vanadium, Chromium, Manganese, Iron, Cobalt, Nickel, Copper, Zinc, Yttrium, Zirconium, Niobium, Molybdenum, Technetium, Ruthenium, Rhodium, Palladium, Silver, Cadmium, Hafnium, Tantalum, Tungsten, Rhenium, Osmium, Iridium, Platinum, Gold, Mercury, Rutherfordium, Dubniuni, Seaborgium, Bohrium, Hassium, Meitnerium, Ununnilium, Unununium or Unuribium at any molar strength to promoter the desired condition, such as 1, 0.7, .5,
  • the buffer conditions can be any buffer such as TRIS at any pH, such as 5.0, 5.5, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.1, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.5, or 9.0.
  • pHs above or below 7.0 increase the specificity of hybridization.
  • the temperature of hybridization can be any temperature.
  • the temperature of hybridization can occur at 20°, 21°, 22°, 23°, 24°, 25°, 26°, 27°, 28°, 29°, 31°, 32°, 33°, 34°, 35°, 36°, 37°, 38°, 39°, 40°, 41°, 42°, 43°, 44°, 45°, 46°, 47°, 48°, 49°, 50°, 51°, 52°, 53°, 54°, 55°, 56°, 57°, 58°, 59°, 60°, 61°, 62°, 63°, 64°, 65°, 66°, 67°, 68°, 69°, 70°, 81°, 82°, 83°, 84°, 85°, 86°,87°, 88°, 89°, 90°, 91°, 92°, 93°, 94°, 95°, 96°, 97°, 98°, or 99°,
  • the length of time of hybridization can be for any time.
  • the length of time can be for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 120, 150, 180, 210, 240, 270, 300, 360, minutes or 7, 8, 9, 10, 11, 12, 13,
  • wash conditions can be used including no wash step.
  • wash conditions occur by a change in one or more of the other conditions designed to require more specific binding, by for example increasing temperature or decreasing the salt or changing the length of time of hybridization.
  • kits comprising nucleic acids which can be used in the methods disclosed herein and, for example, buffers, salts, and other components to be used in the methods disclosed herein.
  • kits for identifying minimal intrinsic gene sets comprising nucleic acids, such as in a microarray.
  • specific minimal intrinsic genes used for classifying cancer such as those found in Table 21. As described above, these intrinsic genes can be used in any combination or permutation, and any combination of permutation of these genes can be used in a kit.
  • kits comprising instructions. 5. Chips and micro arrays
  • chips where at least one address is the sequences or part of the sequences set forth in any of the nucleic acid sequences disclosed herein.
  • chips where at least one address is a variant of the sequences or part of the sequences set forth in any of the nucleic acid sequences disclosed herein.
  • compositions disclosed herein and the compositions necessary to perform the disclosed methods can be made using any method known to those of skill in the art for that particular reagent or compound unless otherwise specifically noted.
  • the nucleic acids such as, the oligonucleotides to be used as primers can be made using standard chemical synthesis methods or can be produced using enzymatic methods or any other known method. Such methods can range from standard enzymatic digestion followed by nucleotide fragment isolation (see for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.
  • compositions can be used in a variety of ways as research tools.
  • the compositions can be used for example as targets in combinatorial chemistry protocols or other screening protocols to isolate molecules that possess desired functional properties related to the disclosed genes.
  • compositions can also be used diagnostic tools related to diseases, such as cancers, such as those listed herein.
  • the disclosed compositions can be used as discussed herein as either reagents in micro arrays or as reagents to probe or analyze existing microarrays.
  • the disclosed compositions can be used in any known method for isolating or identifying single nucleotide polymorphisms.
  • the compositions can also be used in any method for determining allelic analysis of for example, the genes disclosed herein.
  • the compositions can also be used in any known method of screening assays, related to chip/micro arrays.
  • the compositions can also be used in any known way of using the computer readable embodiments of the disclosed compositions, for example, to study relatedness or to perform molecular modeling analysis related to the disclosed compositions.
  • RNA 6000 Nano LabChip Kit (Agilent Technologies, Palo Alto, CA). All samples used had discernable 18S and 28S ribosomal peaks.
  • First strand cDNA was synthesized from approximately 1.5 mg total RNA using 500 ng Oligo(dT)12-18 and Superscript T ⁇ . reverse transcriptase (1st Strand Kit, Invitrogen, Carlsbad, CA). The reaction was held at 42°C for 50 min followed by a 15-min step at 7O 0 C.
  • the cDNA was washed on a QIAquick PCR purification column and stored at - 80°C in TE' (25 mM Tris, 1 mM EDTA) at a concentration of 5 ng/ul (concentration estimated from the starting RNA concentration used in the reverse transcription).
  • Primer design Genbank sequences were downloaded from Evidence viewer (NCBI website) into the Lightcycler Probe Design Software (Roche Applied Science, Indianapolis, IN). All primer sets were designed to have a Tm » 6O 0 C, GC content » 50% and to generate a PCR amplicon ⁇ 200 bps. Finally, BLAT and BLAST searches were performed on primer pair sequences using the UCSC Genome Bioinformatics (http://genome.ucsc.edu/) and NCBI (http://www.ncbi.nhn.nih.gov/BLAST/) to check for uniqueness. Primer sets and identifiers are provided in supplementary Table 8.
  • each 20 ⁇ L reaction included IX PCR buffer with 3 mM MgC12 (Idaho Technology Inc., Salt Lake City, UT), 0.2 mM each of dATP, dCTP, and dGTP, 0.1 mM dTTP, 0.3 mM dUTP (Roche, Indianapolis, IN), 10 ng cDNA and IU Platinum Taq (Invitrogen, Carlsbad, CA).
  • the dsDNA dye SYBR Green I (Molecular Probes, Eugene, OR) was used for all quantification (1/50000 final).
  • PCR amplifications were performed on the Lightcycler (Roche, Indianapolis, IN) using an initial denaturation step (94 0 C, 90 sec) owe y 5 cycles: 'denaturation (94 0 C, 3 sec), annealing (58°C, 5 sec with 20°C/s transition), and extension (72°C, 6 sec with 2° C/sec transition). Fluorescence (530 nm) from the dsDNA dye SYBR Green I was acquired each cycle after the extension step. Specificity of PCR was determined by post-amplification melting curve analysis. Reactions were automatically cooled to 60°C at a rate of 3°C/s and slowly heated at 0.1 °C/s to 95°C while continuously monitoring fluorescence.
  • Microarray hybridizations were carried out on Agilent Human oligonucleotide microarrays (1 A-vl, 1 A-v2 and custom designed 1 A-vl based microarrays) using 2 ⁇ g each of Cy3-labeled "reference” and Cy5-labeled “experimental” sample. Hybridizations were done using the Agilent hybridization kit and a Robbins Scientific "22k chamber” hybridization oven. The arrays were incubated overnight and then washed once in 2X SSC and 0.0005% triton X-102 (10 min), twice in 0.1XSSC (5 ⁇ iih) " ,” aiidlnen immersed ' into Agilent Stabilization and Drying solution for 20 seconds.
  • DWD Distance We ghted Discrimination
  • Luminal tumors with IHC data were scored positive for ER. Conversely, 50 out of 56 (89%) tumors classified as HER2+/ER- or Basal-like were negative for ER by IHC.
  • Cluster analysis showed that the Luminal tumors co-express ER and estrogen responsive genes such as LIV1/SLC39A6, X-box binding protein 1 (XBPl), and hepatocyte nuclear factor 3a (HNF3A/FOXA1).
  • the gene with the highest correlation in expression to ESRl was GATA3 (0.79, 95% CI: 0.71 - 0.85).
  • MammaPrintTM is a microarray assay based on the 70 gene prognosis signature originally identified by van't Veer et al. On the test set validation, the 70 gene assay found that individuals with a poor prognostic signature had approximately a 50% chance of remaining free of distant metastasis at 10 years while those with a good-prognostic signature had a 85% chance of remaining free of disease.
  • Oncotype Dx (Genomic Health Lie) - a real-time qRT-PCR assay that uses 16 classifiers to assess if patients with ER positive tumors are at low, intermediate, or high risk for relapse. While recurrence can be predicted with high and low risk tumors, patients in the intermediate risk group still have variable outcomes and need to be diagnosed more accurately. . _ , f " 55. In genef aij'tumors that have a low risk of early recurrence are low grade and have low expression of proliferation genes. Due to the correlation of proliferation genes with grade and their significance in predicting outcome, a group of 14 proliferation genes were assayed.
  • Example 2 A New Breast Tumor Intrinsic Gene List Identifies Novel Characteristics that are conserveed Across Microarray Platforms
  • a training set of 105 tumors were used to derive a new breast tumor "intrinsic" gene list and validated it using a combined test set of 315 tumors compiled from three independent microarray studies. An unchanging Single Sample Predictor was also used, and applied to three additional test sets.
  • the Mrinsic/UNC gene set identified a number of findings not seen in previous analyses including 1) significance in multivariate testing, 2) that the proliferation signature is an intrinsic property of tumors, 3) the high expression of many Kallikrein genes in Basal-like tumors, and 4) the expression of the Androgen Receptor within the HER2+/ER- and Luminal tumor subtypes.
  • the Single Sample Predictor that was based upon subtype average profiles, was able to identity groups of patients within a test set of local therapy only patients, and two independent tamoxifen-treated patient sets, which showed significant differences in outcomes.
  • the analyses demonstrates that the "intrinsic" subtypes add valtL'et6 ' 'the ex ' ⁇ st ⁇ ri'g fep * ertoire of clinical markers used for breast cancer patients.
  • the computation approach also provides a means for quickly validating gene expression profiles using publicly available data.
  • breast cancers represent a spectrum of diseases comprised of different tumor subtypes, each with a distinct biology and clinical behavior. Despite this heterogeneity, global analyses of primary breast tumors using microarrays have identified gene expression signatures that characterize many of the essential qualities important for biological and clinical classification. Using cDNA microarrays, five distinct subtypes of breast tumors arising from at least two distinct cell types (basal-like and luminal epithelial cells) were previously identified (Perou et al. 2000; Sorlie et al. 2001; Sorlie et al. 2003).
  • This molecular taxonomy was based upon an "intrinsic" gene set, which was identified using a supervised analysis to select genes that showed little variance within repeated samplings of the same tumor, but which showed high variance across tumors (Perou et al. 2000).
  • An intrinsic gene set reflects the stable biological properties of tumors and typically identifies distinct tumor subtypes that have prognostic significance, even though no knowledge of outcome was used to derive this gene set.
  • RNA was purified from each sample using the Qiagen RNeasy Kit according to the manufacturer's protocol (Qiagen, Valencia CA) and using 10-50 milligram of tissue per sample. The integrity of the RNA was determined using the RNA 6000 Nano LabChip Kit and an Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, CA). The total RNA labeling and hybridization protocol used is described in the Agilent low RNA input linear amplification kit (http://www.chem.agilent.com/Scripts/PDS. asp?lPage 10003 ) with the following modifications: 1) a Qiagen PCR purification kit was used to clean up the cRNA and 2) all reagent volumes were cut in half.
  • RNA 0.3 ⁇ g each
  • Microarray hybridizations were carried out on Agilent Human oligonucleotide microarrays (lA-vl, 1A-V2 and custom designed lA-vl based microarrays) using 2 ⁇ g of Cy3-labeled Reference and 2 ⁇ g of Cy5-labeled experimental sample. Hybridizations were done using the Agilent hybridization kit and a Robbins Scientific "22k chamber" hybridization oven.
  • the arrays were incubated overnight and then washed once in 2X SSC and 0.0005% triton X-105 (10 min), twice in 0.1XSSC (5 min), and then immersed into Agilent Stabilization and Drying solution for 20 seconds.
  • AU microarrays were scanned using an Axon Scanner GenePix 4000B.
  • the image files were analyzed with GenePix Pro 4.1 and loaded into the UNC Microarray Database at the University of North Carolina at Chapel Hill (https://genome.unc.edu/) where a Lowess normalization procedure was performed to adjust the Cy3 and Cy5 channels(Yang et al. 2002).
  • Intrinsic gene set analysis A new breast tumor intrinsic gene set was derived, called the "Intrinsic/UNC" list using 105 patients (146 total arrays) and 15 repeated tumor samples that were different physical pieces (and RNA preparations) of the same tumor, 9 tumor-metastasis pairs and 2 normal sample pairs (26 paired samples in total, Table 11). This sample size was chosen based upon Basal-like, Luminal A, Luminal B, HER2+/ER-, and Normal-like samples, which occur at a frequency of 15%, 40%, 15%, 20%, and 10%, respectively; and it was estimated that most clinically relevant classes would constitute at least lU% " of the affected population, and it was hoped to acquire at least 10 samples from each class in the new data set. Therefore, a sample size of 100 tumors was deemed adequate to identify most classes that might be present in breast cancer patients.
  • An intrinsic analysis identifies genes that have low variability in expression within paired samples and high variability in expression across different tumors; for an intrinsic analysis, each gene receives a score that is the average "within-pair variance” (the average square before/after difference), as well as the "between- subject variance” (the variance of the pair averages across subjects).
  • the ratio D (within-pair variance)/(between-subject variance) was then computed, and those genes with a small value of D (i.e. cut-off) declared to be "intrinsic".
  • the choice of a value of D was set at one standard deviation below the mean intrinsic score of all genes. This analysis resulted in the selection of 1410 microarray elements representing 1300 genes.
  • the Single Sample Predictor/SSP is based upon the Nearest Centroid method presented in (Hastie et al. 2001). More specifically, the combined test set was utilized, and 306 Litrinsic/UNC gene set hierarchical cluster presented in Figure 14, as the starting point to create five Subtype Mean Centroids.
  • a mean vector (centroid) for each of the five intrinsic subtypes (LumA, LumB, HER2+/ER-, Basal-like and Normal Breast-like) was created by averaging the gene expression profiles for the samples clearly assigned to each group (which limited the analysis to 249 samples total); the hierarchical clustering dendrogram in Figure 14 were used as a guide for deciding those samples to group together.
  • each sample's correlation to each centroid was calculated using a Spearman correlation and a sample was assigned to the centroid it was closest to, and the test set was then split into a local only therapy test set, and a tamoxifen- treated test set. Finally, the SSP was applied to the 105 sample original training set after DWD normalization.
  • HER2+ expression cluster was observed that contained genes from the 17ql 1 amplicon including HER2/ERBB2 and GRB7 ( Figure 12D).
  • the HER2+ expression subtype (pink dendrogram branch in Figure 12B) was predominantly ER-negative (i.e. HER2+/ER-), but showed expression of the Androgen Receptor (AR) gene.
  • AR Androgen Receptor
  • Basal-like expression cluster was also present and contained genes characteristic of basal epithelial cells such as SOX9, CKl 7, c-KIT, FOXCl and P-Cadherin (Figure 13E). These analyses extend the Basal-like expression profile to contain four Kallikrein genes (KLK5-8), which are a family of serine proteases that have diverse functions and proven utility as biomarkers (e.g. KLK3/PSA); however, it should be noted that KLK3/PSA was not part of the basal profile.
  • KLK3/PSA Kallikrein genes
  • a Luminal/ER+ cluster was present and contained ER, XBPl, FOXAl and GAT A3 ( Figure 12C).
  • GATA3 has recently been shown to be somatically mutated in some ER+ breast tumors (Usary et al. 2004), and some of the genes in Figure 12C are GAT A3 -regulated (FOXAl, TFF3 and AGR2).
  • the Luminal/ER+ cluster contained many new biologically relevant genes such as AR ( Figure 12C), FBPl (a key enzyme in gluconeogenesis pathway) and BCMPl 1.
  • a "combined test set" of 315 breast samples was made (311 tumors and 4 normal breast samples) that was a single data set created by combining together the data from Sorlie et al. 2001 and 2003 (cDNA microarrays), van't Veer et al. 2002 (custom Agilent oligo microarrays) and Sotiriou et al. 2003 (cDNA microarrays).
  • the IFN-regulated cluster contained STATl, which is likely the transcription factor that regulates expression of these IFN-regulated genes (Bromberg et al. 1996; Matikainen et al. 1999).
  • the IFN cluster was one of the first expression patterns to be identified in breast tumors (Perou et al. 1999), and since has been linked to positive lymph node metastasis status and a poor prognosis (Huang et al. 2003; Chung et al. 2004).
  • the effectiveness of the DWD normalization is evident upon close examination of the sample associated dendrogram, which shows that every subtype is populated by samples from each data set (i.e. significant inter-data set mixing). ff6 ' .
  • the Intrinsic/UNC list contained many more genes and a biologically relevant pattern of expression not seen in the Intrinsic/Stanford lists (i.e. proliferation signature), therefore, it can be more biologically representative of breast tumors.
  • the Intrinsic/UNC list can also be more valuable because it provides a larger number of genes for performing across data set analyses and thus, classifications made across different platforms are less susceptible to artifactual groupings as a result of gene attrition.
  • an intrinsic subtype average profile was created for each subtype using the combined test set presented in Figure 14, and then a new sample is individually compared to each centroid and assigned to the subtype/centroid that it is the most similar to using Spearman correlation.
  • an intrinsic subtype can be assigned to any sample, from any data set, one at a time.
  • the final additional test set analyzed was a second adjuvant tamoxifen-treated patient set created by combining similarly treated patients from Chang et al.
  • This study identified a number of new biologically relevant "intrinsic" features of breast tumors and methods that are important for the microarray community. These new biological features include the 1) demonstration that proliferation is a stable and intrinsic feature of breast tumors, 2) identification of the high expression of many Kallikrein genes in Basal-like tumors, and 3) demonstration that there are multiple types of "HER2-positive” tumors; the HER2-positive tumors falling into the "HER2+/ER-” intrinsic subtype were also shown to associate with the expression of the Androgen Receptor, while those not falling into this group were present in the LumB or Luml subtypes and usually showed better outcomes . relative to the HER2+/ER- tumors.
  • LumA LumA
  • LumB LumA
  • Recurrence Score predictor of Paik et al.
  • outcome predictions for tamoxifen- treated ER+ tumors were stratified based mostly on the expression of genes in the HER2- amplicon (HER2 and GRB7), genes of proliferation (STKl 5 and MYBL2), and genes associated with positive ER status (ESRl and BCL2).
  • HER2- amplicon and/or proliferation genes gives a high Recurrence Score (and correlates with LumB because most HER2+ and ER+ tumors fall into this subtype), while low expression of these genes and high expression of ER status genes gives a low Recurrence Score (and correlates with LumA).
  • Example 3 Agreement in Breast Cancer Classification between Microarray and qRT-PCR from Fresh-Frozen and Formalin-Fixed Paraffin- Embedded Tissues
  • Microarray analyses of breast cancers have identified different biological groups that are important for prognosis and treatment.
  • a real-time quantitative (q)RT-PCR assay has been developed for profiling breast tumors from formalin-fixed paraffin-embedded (FFPE) tissues and evaluate its performance relative to fresh-frozen (FF) RNA samples.
  • FFPE formalin-fixed paraffin-embedded
  • Micro array ' data from 124 breast samples were used as a training set for classifying tumors into four different previously defined molecular subtypes of Luminal, HER2+/ER-, Basal-like, and Normal-like.
  • Sample class predictors were developed from hierarchical clustering of microarray data using two different centroid-based algorithms: Prediction Analysis of Microarray and a Single Sample Predictor.
  • the training set data was applied to predicting sample class on an independent test set of 35 breast tumors procured as both fresh-frozen and formalin-fixed, paraffin embedded tissues (70 samples).
  • Classification of the test set samples was determined from microarray data using a large 1300 gene set, and using a minimized version of this gene list (40 genes).
  • the minimized gene set was also used in a real-time qRT-PCR assay to predict sample subtype from the fresh-frozen and formalin- fixed, paraffin embedded tissues. Agreement between primer set performance on fresh-frozen and formalin-fixed, paraffin embedded tissues was evaluated using diagonal bias, diagonal correlation, diagonal standard deviation, concordance correlation coefficient, and subtype assignment.
  • centroid-based algorithms (Prediction Analysis of Microarray and Single Sample Predictor) had complete agreement in classification from formalin-fixed, paraffin- embedded tissues using qRT-PCR and the minimized 'intrinsic' gene set (40 classifiers). There was 94% (33/35) concordance between the diagnostic algorithms when comparing subtype classification from fresh-frozen tissue using microarray (large and minimized gene set) and qRT-PCR data. By qRT-PCR alone, there was 97% (34/35) concordance between fresh-frozen and formalin-fixed, paraffin embedded tissues using Prediction Analysis of Microarray and 91% (32/35) concordance using Single Sample Predictor. Finally, we used several analytical techniques to assess primer set performance between fresh-frozen and formalin-fixed, paraffin- embedded tissues and found that the ratio of the diagonal standard deviation to the dynamic range was the best method for assessing agreement on a gene-by-gene basis.
  • RNA from FF samples was isolated using the RNeasy Midi Kit (Qiagen, Valencia, CA) and treated on-column with DNase I to eliminate contaminating DNA. The RNA was stored at -8O 0 C until used for cDNA synthesis.
  • cDNA synthesis for each sample was performed in 40 ⁇ l total volume reaction containing 600ng total RNA.
  • Total RNA was first mixed with 2 ⁇ l gene specific cocktail containing 55 primers (each anti-sense primer at lpmol/ ⁇ l) and 2 ⁇ l 10 niM dNTP Mix (1OmM each dATP, dGTP, dCTP, dTTP at ⁇ H7). Reagents were heated at 65 0 C for 5 minutes in a PTC-100 Thermal Cycler (MJ Research, Inc.) and briefly centrifuged.
  • Primers were designed using Roche LightCycler Probe Design Software 2.0. Reference gene sequences were obtained through NCBI LocusLink and optimal primer sites were found with the aid of Evidence Viewer (http ://www.ncbi.nlm.nih. gov). Primers sets were selected to avoid known insertions/deletions and mismatches while including all isoforms possible. Amplicons were limited to 60-100bp in length due to the degraded condition of the FFPE mRNA. When possible, RNA specific amplicons were localized between exons spanning large introns (>1 kb). Finally, NCBI BLAST was used to verify gene target specificity of each primer set. Primer sequences are presented in Table 1.
  • Primers were synthesized by Operon, Inc. (Huntsville, AL), re-suspended in TE to a final concentration of 6OuM, and stored at -8O 0 C. Each new FFPE primer set was assessed for performance through qRT-PCR runs with three serial 10-fold dilutions of reference cDNA in duplicate and two no template control reactions. Primers were verified for use when they fulfilled the following criteria: 1) target Cp ⁇ 30 in 10 ng reference cDNA; 2) PCR efficiency >1.75; 3) no primer- dimers in presence of template as determined through post amplification melting curve analysis; and 4) no primer-dimers in negative template control before cycle 40.
  • PCR amplification was carried out on the Roche LightCycler 2.0. Each reaction contained 2 ⁇ l cDNA (2.5ng) and 18 ⁇ l of PCR master mix with the following final concentration of reagents: 1 U Platinum Taq, 5OmM Tris-HCl (pH 9.1), 1.6mM (NH 4 ) 2 SO 4 , 0.4mg/ ⁇ l BSA, 4mM MgCl 2 , 0.2mM dATP, 0.2mM dCTP, 0.2mM dGTP, 0.6mM dUTP, 1/40000 dilution of SYBR Green I dye (Molecular Probes, Eugene, OR, USA), and 0.4 ⁇ M of both forward and reverse primers for the selected target.
  • the PCR was done with an initial denaturation step at 94 0 C for 90s and then 50 cycles of denaturation (94 0 C, 3s), annealing (58 0 C, 6s), and extension (72 0 C, 6s). Fluorescence acquisition (530nm) was taken once each cycle at the end of the extension phase.
  • a post-amplification melting curve program was initiated by heating to 94 0 C for 15 s, cooling to 58 0 C for 15 seconds, and slowly increasing the temperature (0.1°C/s) to 95 0 C while continuously measuring fluorescence.
  • Each PCR run contained a no template control, a calibrator reference in triplicate, and each sample in duplicate.
  • the calibrator reference sample was comprised of 3 breast cancer cell lines (MCF7, SKBR3, and ME16C2) and Stratagene Universal Human Reference RNA (Stratagene, La Jolla, CA, USA) represented in equal parts.
  • the crossing point (C p ) for each reaction was automatically calculated by the Roche LightCycler Software 4.0.
  • RNA samples were assayed versus a common reference that was a mixture of Stratagene's Human Universal Reference total RNA (Stratagene, La Jolla, CA, USA) enriched with equal amounts of RNA from the MCF7 and ME16C cell lines.
  • Microarray hybridizations were carried out on Agilent Human oligonucleotide microarrays using 2 ⁇ g Cy3-labeled 'reference' sample and 2 ⁇ g Cy5-labeled 'experimental' sample.
  • the real-time qRT-PCR assay consisted of 5 housekeeper genes (Szabo et al. Genome Biol 2004, 5:R59), 5 proliferation genes for risk stratification of the Luminal (ER- positive) tumors, and 40 'intrinsic' genes important for distinguishing biological subtypes of breast cancer.
  • the minimal 40 'intrinsic' classifiers were statistically selected from a larger 1300 'intrinsic' gene set previously reported in Hu et al (2006). The larger gene set was minimized as described in Perreard et al (2006). Briefly, a semi-supervised classification method was used in which samples are hierarchical clustered and assigned subtypes based on the sample-associated dendrogram.
  • Samples were designated as Luminal, HER2+/ER— , Basal- like, or Normal-like.
  • the best class distinguishers were identified according to the ratio of between-group to within-group sums of squares.
  • a 10-fold cross-validation was performed using a nearest centroid classifier and testing overlapping gene sets of varying sizes. The smallest gene set which provided the highest class prediction accuracy when compared to the classifications made by the complete microarray-based intrinsic gene set was selected.
  • x and y denote the sample means of the x and y measurements, respectively.
  • This method does not provide information about the extent of deviation but allows measurements with different units to be compared. Further, if we let p denote the correlation coefficient and O ⁇ and O ⁇ the respective standard deviations, then
  • a breast cancer subtype predictor was developed in PAM (http://www- stat. Stanford. edu/ ⁇ tibs/P AM ⁇ and SSP using 124 breast samples and the 'intrinsic' gene set identified in Hu et al (2006).
  • the training set contained representative samples of Luminal (64 samples), HER2+/ER- (23 samples), Basal-like (28 samples), and Normal-like (9 samples) subtypes.
  • Classification of an independent test set (35 matched FF and FFPE samples) was done using a large (1300 genes) and minimized (40 genes) version of the 'intrinsic' set. Subtypes were assigned based on Spearman correlation to the centroid.
  • the qRT-PCR data from the test set was merged with the microarray data of the training set prior to classification using distance weighted discrimination (Benito et al. Bioinformatics 2004, 20:105-1.14).
  • the gold standard for classification of the training and test samples was based on FF tissue RNA and using the classifications obtained when performing hierarchical clustering analysis using the 1300 gene intrinsic gene set from microarray data, b) Results
  • genes with the highest diagonal correlation between FF and FFPE also had the largest dynamic range in expression (e.g., ESRl, TFF3, COX6C, and FBPl).
  • Housekeeper genes and other genes with low variability in expression had the lowest diagonal correlation since they form more of a cloud than a line around the diagonal. The housekeeper genes all had high agreement in terms of having low variability in expression across samples in the FF and FFPE tissues.
  • the concordance correlation coefficient considers both bias and scale shift when determining agreement.
  • the median concordance correlation coefficient between FF and FFPE for the raw data of the 45 genes (housekeepers excluded) was 0.28. Normalization to housekeepers raised the CCC median to 0.48, and adjusting with DWD brought the median to 0.61. Only 27% of the genes had a CCC value greater than 0.5, whereas 47% of the genes were above that value in the normalized data, and 76% were above that when using DWD adjusted normalized data.
  • a comparison of the CCC value to the ratio of the diagonal standard deviation over the dynamic range identified many of the same primer sets as good (or poor) performers from the FFPE derived samples.
  • Hierarchical clustering of the 124 sample training set using the "intrinsic" gene set identified in Hu et al shows 4 distinct classes representing Luminal, HER2+/ER-, Basal- like, and Normal-like (Figure 24). Centroid classifiers were developed from the microarray expression data using PAM and SSP (Hu et al. 2006, Tibshirani et al. 2002). Class predictions were made on the test set using microarray (large and minimized 'intrinsic' sets) and qRT-PCR data (15). Each individual microarray (large and minimized) and PCR datasets were DWD merged with the training set prior to subtype class prediction.
  • a previous qRT-PCR assay for identifying biological subtypes was based on an intrinsic gene set derived from first generation microarrays that contained 8,100 genes.
  • the current intrinsic set was derived from a different microarray platform (cDNA versus Agilent), contained a larger number of genes (427 vs. 1300), and used pre-treatment samples only (Hu et al. 2006.
  • the overlap in the minimized gene set developed here versus the list in Perreard et al. was 14 out of 40, which is not surprising since there were only 108 genes in common between the larger intrinsic gene sets. It has been shown that the new intrinsic gene set reproducibly identifies the same breast cancer subtypes within independent datasets (i.e. pure training and test sets), and that the biological classification adds significant clinical information when used in a multivariate Cox analysis.
  • This method of classification is considered semi-supervised since data from hierarchical clustering is initially used to develop a centroid or shrunken centroid from a training set and new samples are then classified based on the distance to the centroid. In this way, the training set is not only necessary for initial discovery and validation but the data continues to be used as a reference base for future classification of new samples. Similarly, the Oncotype Dx assay established cut points for risk of relapse from a training set and this classifier rule is applied to new samples to derive a recurrence score.
  • Cronin et al used Pearson correlation to show that the genes with the highest correlation in microarray maintained their association with qRT-PCR. They used short amplicons and control 'housekeeper' genes in the qRT-PCR assay to correct biases between FF and FFPE tissues. Although correlation provides information about the linearity and slope (positive or negative correlation) of the data, it does not indicate the amount of bias, scale shift, or data spread. These additional measurements are helpful in determining whether the discrepancies in the data can be compensated for experimentally (e.g., housekeeper genes) or by software algorithms.
  • Translating an assay from microarray to qRT-PCR provides a second level of gene validation and allows the test to be used on archived FFPE tissue blocks from clinical trials or on samples submitted for routine diagnostics (Paik et al. 2004; Cronin et al. Am J Pathol 2004, 164:35-42).
  • qRT-PCR on formalin-fixed paraffin-embedded tissue can be effectively used for gene expression based diagnostics for translation into the clinical laboratory.
  • the FFPE procured RNA provided accurate subtype classifications in breast cancer, and in some instances provided more tumor specific information than the FF derived samples.
  • This study also developed methodologies that have wider application for developing qRT-PCR assays for subtype classification in a wide variety of cancer types. These gene expression based tests can provide powerful new prognostic clinical tools and aid in more appropriate individualized treatment decisions.
  • Intrinsic gene list Intrinsic qene list
  • Rasmussen RP "Quantification on the LightCycler. In Rapid Cycle Real-Time PCR: Methods and Applications” Edited by Wittwer CT, Meuer, S., Nakagawara, K. Heidelberg, Springer Verlag, pp 21-34 (2001).

Abstract

Disclosed are compositions and methods related intrinsic gene sets and methods and compositions related to detecting and classifying cancer.

Description

METHODS AND COMPOSITIONS INVOLVING INTRINSIC GENES
This application claims priority to U.S. Provisional Patent Application No. 60/739,155, which was filed on 23 November 2005.
I. ACKNOWLEDGEMENTS
1. This work was supported in part by the National Cancer Institute (P50-CA58223-11 and R33 CA097769-01 and UOl CAl 14722. The United States Government may have certain rights in the inventions disclosed herein.
II. BACKGROUND
2. A major challenge for microarray studies, especially those with clinical implications, is validation (Ioannidis 2005; Jenssen and Hovig 2005; Michiels et al. 2005). Due to the practical considerations of cost and accessing large numbers of fresh samples with associated clinical information, very few microarray studies have analyzed enough samples to allow the findings to be extended to the general population. Furthermore, it has been difficult to combine and/or validate results from independent laboratories due to differences in sample preparation, patient demographics and the microarray platforms used. An accepted method for validation is to derive a prognostic gene set from a "training set" and then apply it to a "test set" that was not used in any way, to derive the prognostic gene set (Simon et al. 2003); the "purest" test sets have also been suggested to be comprised of samples not contained in the training set and not generated by the primary investigators (Ioannidis 2005). What is needed in the art is a new breast tumor intrinsic gene list that identifies new and important biological features of breast tumors and validates this predictor using a true test set.
III. SUMMARY
3. Described herein is a method of diagnosing cancer, the method comprising comparing expression levels of a combination of genes from Table 21 to test nucleic acids wherein specific expression patterns of the test nucleic acids indicates a cancerous state.
4. Also, disclosed is a method of quantitating level of expression of a test nucleic acid comprising: a) comparing gene expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes; and b) quantitating level of expression of the test nucleic acid.
5. Also disclosed is a method for determining prognosis based on the expression patterns in a subject diagnosed with cancer comprising: a) comparing expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes, b) identifying a subtype of cancer ot the subject, and c) prognosis (ie, outcome) and treatment decisions based on the subtype of cancer in the subject.
6. Disclosed is a method of classifying cancer in a subject, comprising: a) identifying intrinsic genes of the subject to be used to classify the cancer; b) obtaining a sample from the subject; c) amplifying and detecting levels of intrinsic genes in the subject; and d)classifying cancer or subject based upon results of step c.
7. Also disclosed is a method of diagnosing cancer in a subject the method comprising: a) amplifying and detecting intrinsic genes; and b) diagnosing cancer based on expression levels of the gene within the subject.
8. Disclosed herein is a method of deriving a minimal intrinsic gene set for making biological classifications of cancer comprising: a) collecting data from multiple samples from the same individual to identify potential intrinsic classifier genes; b) weighting intrinsic classifier genes of multiple individuals identified using the method of step a relative to each other and forming classification clusters; c) estimating the number of clusters formed in step b) and assigning individual samples to classification clusters; d) identifying genes that optimally distinguish the samples in the assigned groups of step c); e) performing iterative cross- validation with a nearest centroid classifier and overlapping gene sets of various sizes using the genes identified in step d); and f) choosing a gene set which provides the highest class prediction accuracy when compared to the classifications made in step b).
9. Also disclosed is a method of assigning a sample to an intrinsic subtype, comprising a) creating an intrinsic subtype average profile (centroid) for each subtype; b) individually comparing a new sample to each centroid; and c) assigning the new sample to the centroid that is most similar to the expression profile of new sample.
IV. BRIEF DESCRIPTION OF THE DRAWINGS
10. The accompanying drawings, which are incorporated in and constitute a part of this specification, illustrate several embodiments and together with the description illustrate the disclosed compositions and methods.
11. Figure 1 shows the expression levels for the five genes shown by tissue sample. Top: raw data. Bottom: log-scale.
12. Figure 2 shows the expression levels of the 10 genes shown by sample and tissue type. Vandesompele data set in log-scale. 13. Figure 3 shows the mean squared error (MSE) of each gene by tissue-type. The sign is determined by the direction of the bias. The MSE is broken down into the contributing components of the squared bias (BiasΛ2) and the variance (SigmaΛ2). Vandesompele data set.
14. Figure 4 shows two-way hierarchical clustering of microarray data for the same samples assayed by qRT-PCR. Samples were classified based on the expression of 402 "intrinsic" genes defined in Sorlie et al. 2003. The expression level for each gene is shown relative to the median expression of that gene across all the samples with high expression represented by red and low expression represented by green. Genes with median expression are black and missing values are gray. The sample-associated dendrogram shows the same classes seen by qRT-PCR (Figure 5). Samples are grouped into Luminal, HER2+/ER-, Normal-like, and Basal-like subtypes. Overall, 114/123 (93%) primary breast samples classified the same between microarray and qRT-PCR.
15. Figure 5 shows two-way hierarchical clustering of real-time qRT-PCR data from 126 unique samples. The sample-associated dendrogram (5A) shows the same classes seen by microarray. Samples are grouped into Luminal (blue), HER2+/ER- (pink), Normal-like (green), and Basal-like (red) subtypes. The expression level for each gene is shown relative to the median expression of that gene across all the samples with high expression represented by red and low expression represented by green. Genes with median expression are black and missing values are gray. A minimal set of 37 "intrinsic" genes (5B) was used to classify tumors into their primary "intrinsic" subtypes. The "intrinsic" gene set was supplemented using PgR and EGFR (5C), and proliferation genes (5D). The genes in 1C and ID were clustered separately in order to determine agreement between the minimal 37 qRT-PCR "intrinsic" set (5A) and the larger 402 microarray "intrinsic" set.
16. Figure 6 shows Receiver Operator Curves. The agreement between immunohistochemistry (IHC) and gene expression is shown for ER (6A), PR (6B), and HER2 (6C) using ROC. A cut-off for relative gene copy number was selected by minimizing the sum of the observed false positive and false negative errors. The sensitivity and specificity of the resulting classification rule were estimated via bootstrap adjustment for optimism. Since many biomarkers having concordant expression and can serve as surrogates for one another, we tested the accuracy of using GATA3 and GRB7 as surrogates (dotted lines) for calling ER and HER2 protein status, respectively. There was overall good agreement between gene expression and IHC status for ER and PR, but poor agreement between gene expression and IHC status for HER2. The surrogate markers had similar accuracy to the actual markers for predicting HiC status.
17. Figure 7 shows outcome for "intrinsic" subtypes. Kaplan-Meier plots showing relapse free survival (RFS) and overall survival (OS) for patients with Luminal tumors compared to those with HER2+/ER- or Basal-like tumors. Patients with Luminal tumors showed significantly better outcomes for RFS (3A) and OS (3B) compared to HER2+/ER- (RFS: ρ=0.023; OS: p=0.003) and Basal-like (RFS: ρ=0.065; OS: p=0.002) tumors. Classifications were made from real-time qRT-PCR data using the minimal 37 "intrinsic" gene list. Pairwise log-rank tests were used to test for equality of the hazard functions among the intrinsic classes. Tumors in the Normal Breast-like subtype were excluded from the analyses since this class maybe artificially created from having a sample comprised primarily of normal cells.
18. Figure 8 shows grade and proliferation as predictors of relapse free survival. Kaplan-Meier plots are shown for grade (8A) and the proliferation genes (8B) using Cox regression analysis. The analysis for the proliferation genes was performed on continuous expression data, although the plots are shown in tertiles. The proliferation index (log average of the 14 proliferation genes) has significant predictive value for outcome, even after correcting for other clinical parameters important for survival. Furthermore, when we include both grade and the proliferation index (and stage) in a model for RFS, we find that the proliferation index is the superior predictor (Grade p=0.51; Proliferation index p=0.047).
19. Figure 9 shows co-clustering of real-time qRT-PCR and microarray data using 50 genes and 252 samples. The relative copy number (qRT-PCR) and R/G ratio (microarray) for each gene was Iog2 transformed and combined into a single dataset using distance weighted discrimination. Two-way hierarchical clustering was performed on the combined dataset using Spearman correlation and average linkage. The sample associated dendrogram (5A) shows the same classes as seen in Figure 1. Samples are classified as Basal-like (red), HER2+/ER-, Luminal, and Normal-like. The expression level for each gene is shown relative to the median expression of that gene across all the samples with overexpressed genes and underexpressed genes, as well as average expression. The gene associated dendrogram (5B) shows that the Luminal tumors and Basal-like tumors differentially express estrogen associated genes (cluster 1); as well as basal keratins (KRT 5 and 17), inflammatory response genes (CX3CL1 and SLPI), and genes in the Wnt pathway (FZD7) (cluster 3). The main distinguishers of the HER2+/ER- group are low expression of genes in cluster 1 and high expression of genes on the 1/q12 amplicon (ERBB2 and GRB7) (cluster 4). The proliferation genes (cluster 2) have high expression in the ER negative tumors (Basal-like and HER2+/ER-) and low expression in ER positive (Luminal) and Normal-like samples.
20. Figure 10 shows a flow chart of the steps of deriving minimal intrinsic gene sets for making biological classifications of breast cancer.
21. Figure 11 shows an overview and flow of the data sets used and analyses performed.
22. Figure 12 shows a hierarchical cluster analysis of the training set using the Intrinsic/UNC gene set. 146 microarrays, representing 105 tumors and 9 normal breast samples were analyzed using the 1300 gene Intrinsic/UNC gene set. A) Overview of the complete cluster diagram (the full cluster diagram can be found as Supplemental Figure 1). B) Experimental sample associated dendrogram. The 26 paired samples used for the intrinsic analysis are identified by the black bars. C) Luminal/ER+ gene expression cluster with GATA3-regulated genes shown in pink. D) HER2 and GRB7 containing expression cluster. E) Basal epithelial enriched expression cluster. F) Proliferation associated expression cluster. The genes in red are mentioned in the text. The Single Sample Predictor/SSP was applied back onto this training data set with the individual sample classifications identified using colored squares (Pink=HER2+/ER-, Red=Basal-like, Dark Blue=Luminal A, Light Blue=Luminal B, and Green=Normal Breast-like).
23. Figure 13 shows Androgen Receptor (AR) immunohistochemistry on human breast tumors. A) AR staining on the HER2+/ER- subtype tumor BR00-0284. B) AR staining on the HER2+/ER- subtype tumor PB455 showing nuclear localization. C)AR staining on the Luminal subtype tumor BR01-0246. D) Lack of AR staining on the Basal-like subtype tumor BR97-0137. The magnification is approximately 200X.
24. Figure 14 shows hierarchical cluster analysis the combined test set of 311 tumors and 4 normal breast samples analyzed using the Intrinsic/UNC gene set reduced to 306 genes. A) Overview of the complete cluster diagram. B) Experimental sample associated dendrogram. C) Luminal/ER+ gene expression cluster with GAT A3 -regulated genes in pink text. D) HER2 and GRB7 containing expression cluster. E) Interferon-regulated cluster containing STATl. F) Basal epithelial enriched cluster. G) proliferation cluster.
25. Figure 15 shows univariate Kaplan-Meier survival plots using RFS as the endpoint, for the common clinical parameters present within the combined test set of 311 tumors. Survival plots for A) ER status, B) node status, C) grade, and D) tumor size.
Figure imgf000007_0001
Kaplan-Meier survival plots for intrinsic subtype analyses. A) Relapse-free survival for the 105 patients/tumors training set classified using hierarchical clustering and complete 1300 gene the Litrinsic/UNC list. B) Relapse-free survival for the 315 sample combined test set analyzed using the Intrinsic/UNC list reduced to 306 genes. C) Survival analysis of the 60 adjuvant tamoxifen-treated patients from the Ma et al. 2004 study who were classified as either LumA, LumB or Normal Breast-like using the Single Sample Predictor. D) Survival analysis of the 96 local treatment only (i.e. surgery alone) test set patients taken from Chang et al. 2005, which were classified using the Single Sample Predictor. E) Survival analysis of a second pure test set of 45 patients treated with adjuvant tamoxifen and classified using the Single Sample Predictor. F) Relapse-free survival for the 105 patients/tumors training set, and classified using the Single Sample Predictor. All p-values were based on a log-rank test.
27. Figure 17 shows grade and proliferation as predictors of relapse free survival. A Cox regression model was used to determine probability of relapse over time. Kaplan-Meier curves show time to event given different grades and levels of proliferation. Grade was scored as low (green), medium (red) or high (blue). The proliferation score was based on continuous expression data and is shown as textiles that correspond to low (green), medium (red), and high (blue) levels of expression. The proliferation meta-gene (Iog2 average of the 14 proliferation genes) showed significant value in predicting relapse, even after correcting for other clinical parameters important for survival (Table 1). Furthermore, when both grade and proliferation were used in a model for RFS, it was found that the proliferation meta-gene is the better predictor (Grade p=0.51; Proliferation index p=0.047).
28. Figure 18 shows an agreement plot between fresh frozen (FF) and formalin-fixed paraffin-embedded (FFPE) for the estrogen receptor gene (ESRl) after normalization to the 5 housekeepers.
29. Figure 19 shows line graphs which show the effects at each step of data processing. The raw (pre-normalized) data shows a negative bias for all genes likely due to lower RNA quality in the FFPE tissue (Fig 2A). Much of the bias was corrected by normalization to the 'housekeeper' genes and using DWD adjustment. As expected, DWD had a significant effect on bias (m) but did not effect other measurements of agreement (Fig 2B-D).
30. Figure 20 shows a large dynamic range of ESRl expression provides clear separation of the tumors from both FF and FFPE. 31. Figure 21 shows a scatter plot of ER status by immunohistochemistry versus expression or ESRl.
32. Figure 22 shows a scatter plot of average copy number in an formalin-fixed paraffin embedded (FFPE) sample versus a fresh frozen (FF) sample. The results are normalized to all five housekeepers.
33. Figure 23 shows a scatter plot of average copy number in an formalin-fixed paraffin embedded (FFPE) sample versus a fresh frozen (FF) sample. The results are after DWD.
34. Figure 24 shows hierarchical clustering of the 124 sample training set using the "intrinsic" gene set identified in Hu et al, which shows 4 distinct classes representing Luminal, HER2+/ER-, Basal-like, and Normal-like.
35. Figure 25 shows MA P3m test (4Og X 35s) gene scores, as well as FF PCR test (4Og X 35s) gene scores for for basal, HER-2, luminal and normal like.
V. DETAILED DESCRIPTION
36. Before the present compounds, compositions, articles, devices, and/or methods are disclosed and described, it is to be understood that they are not limited to specific synthetic methods or specific recombinant biotechnology methods unless otherwise specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
A. Definitions
37. As used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a pharmaceutical carrier" includes mixtures of two or more such carriers, and the like.
38. Ranges can be expressed herein as from "about" one particular value, and/or to "about" another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about," it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as "about" that particular value in addition to the value itself. For example, if the value "10" is disclosed, then "about 10" is also disclosed. It is also understood that when a value is disclosed that "less than or equal to" the value, "greater than or equal to the value" and possible ranges between values are also disclosed, as appropriately understood by the skilled artisan. For example, if the value "10" is disclosed the "less than or equal to 10"as well as "greater than or equal to 10" is also disclosed. It is also understood that the throughout the application, data is provided in a number of different formats, and that this data, represents endpoints and starting points, and ranges for any combination of the data points. For example, if a particular data point "10" and a particular data point 15 are disclosed, it is understood that greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15 are considered disclosed as well as between 10 and 15. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.
39. As used throughout, by a "subject" is meant an individual. Thus, the "subject" can include, for example, domesticated animals, such as cats, dogs, etc., livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), laboratory animals (e.g., mouse, rabbit, rat, guinea pig, etc.) mammals, non-human mammals, primates, non-human primates, rodents, birds, reptiles, amphibians, fish, and any other animal. The subject can be a mammal such as a primate or a human.
40. "Treating" or "treatment" does not mean a complete cure. It means that the symptoms of the underlying disease are reduced, and/or that one or more of the underlying cellular, physiological, or biochemical causes or mechanisms causing the symptoms are reduced. It is understood that reduced, as used in this context, means relative to the state of the disease, including the molecular state of the disease, not just the physiological state of the disease.
41. By "reduce" or other forms of reduce means lowering of an event or characteristic. It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative value to be referred to. For example, "reduces phosphorylation" means lowering the amount of phosphorylation that takes place relative to a standard or a control.
42. By "inhibit" or other forms of inhibit means to hinder or restrain a particular characteristic. It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative Valueto^e11 referred to':'1 For example, "inhibits phosphorylation" means ndering or restraining the amount of phosphorylation that takes place relative to a standard or a control.
5.By "prevent" or other forms of prevent means to stop a particular characteristic or condition. Prevent does not require comparison to a control as it is typically more absolute than, for example, reduce or inhibit. As used herein, something could be reduced but not inhibited or prevented, but something that is reduced could also be inhibited or prevented. It is understood that where reduce, inhibit or prevent are used, unless specifically indicated otherwise, the use of the other two words is also expressly disclosed. Thus, if inhibits phosphorylation is disclosed, then reduces and prevents phosphorylation are also disclosed.
6.By "specific expression pattern" is meant an elevation or reduction of expression of given genes when compared with a control or a standard. One of ordinary skill in the art is capable of identifying and measuring the expression of gene patterns of genes related to the methods disclosed herein.
43. The term "therapeutically effective" means that the amount of the composition used is of sufficient quantity to ameliorate one or more causes or symptoms of a disease or disorder.
Such amelioration only requires a reduction or alteration, not necessarily elimination. The term "carrier" means a compound, composition, substance, or structure that, when in combination with a compound or composition, aids or facilitates preparation, storage, administration, delivery, effectiveness, selectivity, or any other feature of the compound or composition for its intended use or purpose. For example, a carrier can be selected to minimize any degradation of the active ingredient and to minimize any adverse side effects in the subject.
44. Throughout the description and claims of this specification, the word "comprise" and variations of the word, such as "comprising" and "comprises," means "including but not limited to," and is not intended to exclude, for example, other additives, components, integers or steps.
45. The term "cell" as used herein also refers to individual cells, cell lines, or cultures derived from such cells. A "culture" refers to a composition comprising isolated cells of the same or a different type.
46. References in the specification and concluding claims to parts by weight, of a particular element or component in a composition or article, denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed. Thus, in a compound containing 2 parts by weight of component X and 5 parts by weight component Y, an Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound.
47. A weight percent of a component, unless specifically stated to the contrary, is based on the total weight of the formulation or composition in which the component is included.
48. In this specification and in the claims which follow, reference will be made to a number of terms which shall be defined to have the following meanings:
49. "Optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
50. "Primers" are a subset of probes which are capable of supporting some type of enzymatic manipulation and which can hybridize with a target nucleic acid such that the enzymatic manipulation can occur. A primer can be made from any combination of nucleotides or nucleotide derivatives or analogs available in the art which do not interfere with the enzymatic manipulation.
51. "Probes" are molecules capable of interacting with a target nucleic acid, typically in a sequence specific manner, for example through hybridization. The hybridization of nucleic acids is well understood in the art and discussed herein. Typically a probe can be made from any combination of nucleotides or nucleotide derivatives or analogs available in the art.
B. Compositions and methods
52. Disclosed herein are methods and compositions for deriving a minimal intrinsic gene set for making biological classifications of cancer. Also disclosed are methods of using intrinsic genes in a real-time qRT-PCR assay for cancer classification, prognosis and/or treatment. Described herein are several algorithms for use in combination in order to generate a statistically validated minimal gene set that makes biological classifications of cancers. While the methods disclosed herein are generally useful with any type of cancer, breast cancer is specifically used as an example herein. Below follows a list of specific cancers that are useful with the methods disclosed herein, and the example of breast cancer is not intended to be limiting, but rather exemplary. The samples disclosed herein can be obtained from a variety of sources, including fresh tissue, fresh-frozen samples, or formalin-fixed paraffin-embedded samples.
53. The methodology described herein can be used to make a classification that distinguishes 2 or more intrinsic subtypes of breast cancer. The intrinsic subtypes can be
Figure imgf000012_0001
classes therein), HER2 ER- an c asses t ere n , Basa an classes therein), Normal-like (and classes therein). The steps for finding the minimal intrinsic gene set for making subtype (and class) distinctions are as follows.
54. The first step is to use microarray data from biological replicates from the same patient to find intrinsic classifier genes. For example, a data set of tumors and normal breast samples can be used, hi one embodiment, these data sets can comprise paired biological replicates to identify the intrinsic gene set. This is described, for example, in Perou et al. (2000), which is herein incorporated by reference in its entirety for its teaching regarding finding intrinsic classifier genes. In Perou et al., the molecular portraits revealed in the patterns of gene expression not only uncovered similarities and differences among the tumors, but also point to a biological interpretation. Variation in growth rate, in the activity of specific signalling pathways, and in the cellular composition of the tumors were all reflected in the corresponding variation in the expression of specific subsets of genes.
55. In the second step of the method disclosed herein, hierarchical cluster microarray data was obtained using an intrinsic gene set. Here, data can be combined from different microarray platforms for clustering using methods described in Example 2. Specifically, the "intrinsic gene set" from the first step (above) is tested on new tumors and normal breast samples after combining different datasets (such as cross platform analyses) and common genes/elements are hierarchically clustered. For example, a two-way average linkage hierarchical cluster analysis can be performed using a centered Pearson correlation metric and the program "Cluster" (Eisen et al. 1998), with the data being displayed relative to the median expression for each gene (i.e. median centering of the rows/genes).
56. In the third step, the number of clusters formed in the microarray dataset is estimated, and samples/tumors are assigned to clusters based on the sample-associated dendrogram groupings. In other words, the "test set" is used as a training set to create subtype centroids based upon the expression of the common intrinsic genes. New samples are assigned to the subtype corresponding to the nearest centroid when using Spearman correlation values.
57. In the fourth step, genes are found that optimally distinguish the samples in the assigned groups using the ratio of between-group to within-group sums of squares (the entire microarray dataset is used in this analysis). An example of this can be found in Chung et al, Cancer Cell 2004, herein incorporated by reference in its entirety for its teaching concerning identification of genes that optimally distinguish samples. '31S:1 in the!' fϊftlfstep, iterative cycles of 10-fo cross-va at on are per orme w t a nearest centroid classifier and overlapping gene sets of varying sizes. In other words, each gene and gene set are ranked based upon the metric from step four above, and various overlapping and every increasing sized genes lists are used in a 10-fold cross validation.
59. In the sixth, and final step, the smallest gene set which provides the highest class prediction accuracy when compared to the classifications made by the complete microarray- based intrinsic gene set is chosen. Subtypes are assigned for each gene set and the minimal gene set with the highest agreement in sample assignment to the full intrinsic gene set is chosen, hi one example, using a 1410 intrinsic gene set as disclosed in Example 2, 100 genes were identified (see Table 12 (7p 100), after the "Examples" section) that are important for identifying 7 different biological classes of breast cancer. Specific steps and sample sets used to develop the 7-class predictor as shown in Figure 11. Also disclosed in Table 13 is an extended list of genes for classification resulting from the 7p analyses. This list is ranked in terms of significance for separating the different classes of intrinsic classifier genes. Another set of intrinsic genes that can be used for classification is found in Table 21, along with the primers that can be used to amplify those genes. It should be noted that the primers are optional and exemplary only, as any primer that can amplify a given gene can be used.
60. The minimal intrinsic gene set (identified using the methods described above, and found in Tables 12 and 13) has prognostic and predictive significance in breast cancer. The complete assay for making these biological "intrinsic" classifications includes 3 "housekeeper" genes (MRPLl 9, PUMl, and PSMC4) for normalizing the quantitative data. In addition, it has been shown that proliferation genes can also be used in combination with the housekeeper genes for providing a quantitative measurement of grade and for assessing prognosis in breast cancer.
61. Also disclosed herein is the Single Sample Predictor (SSP). The Single Sample Predictor/SSP is based upon the Nearest Centroid method presented in (Hastie et al. 2001). The subtype centroids (either all intrinsic genes or the minimal gene lists) can be used to make subtype predictions on additional test sets (e.g., homogenously treated subjects from clinical trial groups). The resulting classifications are then analyzed using Kaplan-Meier survival plots to determine prognostic and therapeutic significance. An example of SSP can be found in Example 2. 1. Intrinsic genes and cancer
62. An intrinsic gene is a gene that shows little variance within repeated samplings of the same tumor, but which shows high variance across tumors. Disclosed herein are genes that can be used as intrinsic genes with the methods disclosed herein. The intrinsic genes disclosed herein can be genes that have less than or equal to 0.00001, 0.0001, 0.001, 0.01, 0.1, 0.2. 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 1,000, 10,000, or 100,000% variation between two samples from the same tissue. It is also understood that these levels of variation can also be applied across 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 or more tissues, and the level of variation compared. It is also understood that variation can be determined as discussed in the examples using the algorithms as disclosed herein.
63. "Intrinsic gene set" is defined herein as comprising one or more intrinsic genes. "Minimal intrinsic gene set" is defined herein as being derived from an intrinsic gene set, and is considered the fewest number of intrinsic genes that can be used to classify a sample.
64. Disclosed herein is a set of 212 minimal intrinsic genes, as found in Table 21. These genes can be used alone, or in combination, as intrinsic genes for the purposes of classification, prognosis, and diagnosis of cancer, for example. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154. 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199 of the genes can be used with the methods disclosed herein for analyzing samples.
65. Described herein is a method of diagnosing cancer, the method comprising comparing expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes, w ere n spec c express on patterns o t e test nucleic acids indicates a cancerous state.
66. Also disclosed is a method of quantitating level of expression of a test nucleic acid comprising: a) comparing gene expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes; and b) quantitating level of expression of the test nucleic acid.
67. Also disclosed is a method of prognosing outcome in a subject diagnosed with cancer comprising: a) comparing expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes, b) identifying a subtype of cancer of the subject, and c) prognosing the outcome based on the subtype of cancer of the subject.
68. The intrinsic genes disclosed herein can be normalized to control housekeeper genes and used in a qRT-PCR diagnostic assay that uses relative copy number to assess risk or therapeutic response in cancer. For example, MRPL19 (SEQ ID NO:1), PSMC4 (SEQ ID NO:2), SF3A1 (SEQ IDNO:3), PUMl (SEQ ID NO:4), ACTB (SEQ ID NO:5) and GAPD (SEQ ID NO:6). Other genes include GUSB, RPLPO, and TFRC, whose sequences can be found in Geribank. These are part of the 212 gene list. Other genes as disclosed herein can also be considered intrinsic genes.
69. The intrinsic genes can be used in any combination or singularly in any method described herein. It is also understood that any nucleic acid related to the expression control genes, such as the RNA, mRNA, exons, introns, or 5' or 3' upstream or downstream sequence, or DNA or gene can be used or identified in any of the methods or with any of the compositions disclosed herein.
2. Molecules for detecting genes, gene expression products, proteins encoded by genes
70. The disclosed methods involve using specific intrinsic genes or gene sets or expression control genes or gene sets such that they are detected in some way or their expression product is detected in some way. Typically the expression of a gene or its expression product will be detected by a primer or probe as disclosed herein. However, it is understood that they can also be detected by any means, such as in a microarray analysis or a specific monoclonal antibody or other visualization technique. Often, the expression of the genes of interest (control "housekeeper" genes or intrinsic classifier genes) can be detected after or during an amplification process, such as RT-PCR, including quantitative PCR. 3. Method of diagnosing or prognosing cancer
Microarrays have shown that gene expression patterns can be used to molecularly classify various types of cancers into distinct and clinically significant groups. In order to translate these profiles into routine diagnostics, a microarray breast cancer classification system has been recapitulated using real-time quantitative (q)RT-PCR (Example 2). Statistical analyses were performed on multiple independent microarray datasets to select an "intrinsic" gene set that can classify breast tumors into four different subtypes designated as Luminal, Normal-like, HER2+/ER-, and Basal-like. Intrinsic genes, as described in Perou et al. (Nature (2000) 406:747-752), are statistically selected to have low variation in expression between biological sample replicates from the same individual and high variation in expression across samples from different individuals. Thus, intrinsic genes are the classifier genes for breast cancer classification and each classifier gene can be normalized to the housekeeper (or control) genes in order to make the classification. A minimal gene set from the microarray "intrinsic" list, and additional genes important for outcome (e.g., proliferation genes), were used to develop a real-time qRT-PCR assay comprised of 53 classifiers and 3 housekeepers. The expression data and classifications from microarray and real-time qRT-PCR were respectively compared using 123 unique breast samples (117 invasive carcinomas, 1 fibroadenoma and 5 normal tissues) and 3 cells lines. The overall correlation for the 50 genes in common between microarray and qRT-PCR was 0.76. There was 91% (114/126) concordance in the hierarchical clustering classification of the real-time qRT-PCR minimal "intrinsic" gene set (37 genes) and the larger (550 genes) microarray intrinsic gene set from which the PCR list was derived. As expected, the Luminal tumors (ER+) had a significantly better outcome than the HER2+/ER- (p=0.043) and Basal-like tumors (p=0.001). High expression of the proliferation genes GTBP4 (P=O-OIl), HSP A14 (p=0.023), and STK6 (ρ=0.027) were significant predictors of relapse free survival (RFS) independent of grade and stage. It has been shown that genomic microarray data can be translated into a qRT-PCR diagnostic assay that improves the standard of care in breast cancer.
The overlap in the minimized gene set discussed above and in Example 2 versus those in Example 3 is 14 out of 40. There are 108 genes in common between the larger intrinsic gene sets, which included 427 in Perreard et al versus 1300 used in Example 3. Example 2 illustrates how intrinsic gene sets can be minimized from microarray data and used on fresh tissue in a qRT-PCR assay to recapitulate the microarray classifications. It also shows the importance of the 'proliferation' genes in risk stratifying Luminal (ER+) breast tumors. Example 3 discusses a version of the intrinsic gene set from Hu et a an s ows again ow t can be minimized to provide intrinsic classifications on both fresh and FFPE tissue and using microarray or qRT-PCR data. Validated primer sequences from FFPE tissues for 212 genes important for breast cancer diagnostics are presented in Table 21.
71. A major challenge in the clinical care of cancer has been providing an accurate diagnosis for appropriate management of breast cancer. For over 50 years, medicine has relied on morphological features (histopathology) and anatomic staging (Tumor size/Node involvement/Metastasis) for classification of tumors (Greenough, R.B. J Cancer Res 9:452- 463; Bloom et al. (1957) British Journal of Cancer 9:359-377). The TNM staging system provides information about the extent of disease and has been the "gold standard" for prognosis (Henson, et al. (1991) Cancer 68:2142-2149; Fitzgibbons, et al (2000) Arch Pathol Lab Med 124:966-978).
72. hi addition to TNM, the grade of the tumor is also prognostic for relapse free survival (RFS) and overall survival (OS) (Elston et al. (1991) Histopathology 19:403-410). Tumor grade is determined from histological assessment of tubule formation, nuclear pleomorphism, and mitotic count. Due to the subjective nature of grading and difficulties standardizing methods, there has been less than optimal agreement between pathologists (Dalton et al. (1994) Cancer 73:2765-2770). Applying the Nottingham combined histological grade has made scoring more quantitative and improved agreement between observers (Frierson (1995) Am J Clin Pathol 103:195-198), however, more objective methods are still needed before grade is integrated into the TNM classification (Singletary (2003) Surg Clin North Am 83:803-819). For instance, most studies show significance in outcome between Grade 1 (low/least aggressive) and Grade 3 (high/most aggressive), but Grade 2 (intermediate) tumors show variability in outcome and are commonly not classified the same across institutions (Kollias et al. (1999) Eur J Cancer 35:908-912; Robbins et al. (1995) Hum Pathol 26:873-879; Genestie et al. (1998) Anticancer Res 18:571-576.). Alternatively, proliferation assays, such as S-phase fraction and mitotic index, have shown to be independent prognostic indicators and could be used in conjunction with, or instead of grade (Michels et al. (2004) Cancer 100:455-464; CaIy et al. (2004) Anticancer Res 24:3283-3288). It has been shown that proliferation genes can be used in a qRT-PCR assay and the genes can be averaged to produce a proliferation meta-gene that correlates with grade but is more prognostic (Figure 17).
73. Women with the same stage of breast cancer can have widely different clinical outcomes due to differences in tumor biology (van 't Veer et al. (2002) Nature 415:530-536; van αe Vϊjvfer et a'l.'porø) N Engl J Med 347:1999-2009 . The use of gene expression markers in breast pathology can provide addition clinical information that complements the TNM system for prognosis and is important for making therapeutic decisions (van 't Veer et al. (2002) Nature 415:530-536; van de Vijver et al. (2002) N Engl J Med 347:1999-2009; Paik et al. (2004) N Engl J Med 351:2817-2826; Sørlie et al. (2001) Proc Natl Acad Sci U S A 98:10869-10874; Sorlie et al. (2003) Proc Natl Acad Sci U S A 100:8418-8423). Undoubtedly, one of the greatest advancements in breast cancer medicine has been the identification and routine testing for the expression of the hormone receptors, namely the Estrogen Receptor (ER) and the Progesterone Receptor (PgR), which allows the clinician to offer endocrine blockade therapy that can significantly prolong survival in women with tumors expressing these proteins (Buzdar et al. (2003) J Clin Oncol 21:1007-1014; Fisher et al (1989) N Engl J Med 320:479- 484).
74. Although ER expression is a predictive marker, it also serves as a surrogate marker for describing a tumor biology that is characteristically less aggressive (e.g. lower grade) than ER-negative tumors (Fisher et al. (1981) Breast Cancer Res Treat 1:37-41). Microarrays have elucidated the richness and diversity in the biology of breast cancer and have identified many genes that associate with ER-positive and ER-negative tumors (Perou et al. (2000) Nature 406:747-752; West et al. (2001) Proc Natl Acad Sci U S A 98:11462-11467; Gruvberger et al. (2001) Cancer Res 61:5979-5984). When microarray data from invasive breast carcinomas are analyzed by hierarchical clustering, samples are separated primarily based on ER status (Sotiriou et al. (2003) Proc Natl Acad Sci U S A 100:10393-10398).
75. Breast tumors of the "Luminal" subtype are ER positive and have a similar keratin expression profile as the epithelial cells lining the lumen of the breast ducts (Taylor- Papadimitriou et al. (1989) J Cell Sci 94:403-413; Perou et al. (2000) New Technologies for life sciences: A Trends Guide:67-76). Conversely, ER-negative tumors can be broken into two main subtypes, namely those that overexpress (and are DNA amplified for) HER2 and GRB7 (HER2+/ER-), and "Basal-like" tumors that have an expression profile similar to basal epithelium and express Keratin 5, 6B and 17. Both these tumor subtypes are aggressive and typically more deadly than Luminal tumors; however, there are subtypes of Luminal tumors that lead to poor outcome despite being ER-positive. For instance, Sorlie et al. identified a Luminal B subtype with similar outcomes to the HER2+/ER- and Basal-like subtypes, and Sotiriou et al. showed that there are 3 different types of Luminal tumors with different outcomes. The Luminal tumors with poor outcomes consistently share the histopathological ieattrøWbeing Higher" grade and the molecular feature of highly expressing proliferation genes.
76. The so called "proliferation genes" show periodicity in expression through the cell cycle and have a variety of functions necessary for cell growth, DNA replication, and mitosis (Whitfield et al. (2002) MoI Biol Cell 13:1977-2000; Ishida et al. MoI Cell Biol 21:4684- 4699). Despite their diverse functions, proliferation genes have similar gene expression profiles when analyzed by hierarchical clustering. As might be expected, proliferation genes correlate with grade, the mitotic index ( Perou et al. (1999) Proc Natl Acad Sci U S A 96:9212- 9217), and outcome ( Sørlie et al. (2001) Proc Natl Acad Sci U S A 98:10869-10874). Proliferation genes are often selected when supervised analysis is used to find genes that correlate with patient outcome. For example, the SAM264 "survival" list presented in Sorlie et al., the 231 "prognosis classifier" list in van't Veer et al., and the "485 prognostic gene" list in Sotiriou et al., identified common proliferation genes (PCNA, TOP2A, CENPF). This suggests that all these studies are likely tracking a similar phenotype.
77. Gene expression profiling using DNA microarrays is a powerful tool to discover genes for molecular classifications of cancer but the platforms are labor intensive, expensive and currently not amenable to routine clinical diagnostics. Real-time qRT-PCR is well-suited for solid tumor diagnostics since it is rapid, homogenous (amplification and quantification in a single vessel), and can be performed from archived (FFPE tissue) samples. Example 3 shows that FFPE samples can perform as well as fresh samples. It has been shown that "intrinsic" breast cancer classifications from microarray can be recapitulated by qRT-PCR using a minimal "intrinsic" gene set. In addition, by supplementing the "intrinsic" gene set with proliferation genes, a more objective measurement of grade has been developed. The assay disclosed herein adds prognostic information to the standard of care for breast cancer.
78. Microarray used in conjunction with RT-PCR provides a powerful system for discovering and translating genomic markers into the clinical laboratory for molecular diagnostics. Although these platforms are fundamentally very different, the quantitative data across the methods have a high correlation. In fact, the data across the methods is no more disparate then across different microarray platforms. By hierarchical clustering, it has been shown that a biological classification of breast cancer derived from microarray data can be recapitulated using real-time qRT-PCR. Biological classification by real-time qRT-PCR makes the important clinical distinction between ER positive and ER negative tumors and identifies additional subtypes that have prognostic (ie, correlate to outcome) and predictive value (ie, correlate to treatment response).
79. The benefit of using real-time qRT-PCR for cancer diagnostics is that new informative markers can be readily validated and implemented, making tests expandable and/or tailored to the individual. For instance, it has been shown that including proliferation genes serves a similar purpose to grade but is more prognostic. Since grade has been shown to be universal as a prognostic factor in cancer, it is likely that the same markers correlate to grade and are important for survival in other tumor types. Real-time qRT-PCR is attractive for clinical use because it is fast, reproducible, tissue sparing, and able to be automated. Although genomic profiling should currently be used for ancillary testing, the fact that normal tissues can be distinguished from tumor tissue shows that these molecular assays may eventually be used for cancer diagnostics without histological corroboration.
80. Disclosed is a method of classifying cancer in a subject, comprising: a) identifying intrinsic genes of the subject to be used to classify the cancer; b) obtaining a sample from the subject; c) amplifying and detecting levels of intrinsic genes in the subject; and d)classifying cancer based upon results of step c. The sample can be fresh, or can be an FFPE sample.
81. Also disclosed is a method of diagnosing cancer in a subject the method comprising: a) amplifying and detecting intrinsic genes; and b) diagnosing cancer based on expression levels of the gene within the subject. The methods disclosed herein can be used with any of the types of cancer listed herein. The cancer can be breast cancer, for example. The breast cancer can be classified into one of four or more groups: luminal, normal-like, HER2+/ER- and basal-like, for example. Again, the sample can be fresh, or can be an FFPE sample.
82. Disclosed are methods of analyzing nucleic acid expression levels in a sample, the methods comprising comparing expression levels of an intrinsic gene set to a test nucleic acid, wherein specific expression patterns of the test gene relative to the intrinsic gene set indicates a diagnoses, poor prognosis, likelihood of obtaining, predisposition to obtaining, or presence of a cancer. Also disclosed are methods wherein the step of comparing comprises identifying the expression levels of an intrinsic gene set and a test nucleic acid by interaction with a primer or probe.
83. Disclosed are methods where a specific expression pattern of a test nucleic acid relative to an intrinsic gene set indicates the presence of a cancer, a poor (or good) prognosis for a patient having a cancer, a predisposition of getting a cancer, or a iagnoses of cancer or a cancerous state.
84. It is understood that any method of assaying any gene discussed herein can be performed. For example methods of assaying gene copy number or mRNA expression copy number can be performed. For example, RT-PCR, PCR, quantitative PCR, and any other forms of nucleic acid amplification can be performed. Furthermore, methods of hybridization, such as blotting, such as Northern or Southern techniques, such as chip and microarray techniques and any other techniques involving hybridizing of nucleic acids.
4. A non-limiting list of Cancers which can be assayed with disclosed compositions and methods
85. The disclosed compositions can be used to diagnose or prognose any disease where uncontrolled cellular proliferation occurs such as cancers. A non-limiting list of different types of cancers is as follows: lymphomas (Hodgkins and non-Hodgkins), leukemias, carcinomas, carcinomas of solid tissues, squamous cell carcinomas, adenocarcinomas, sarcomas, gliomas, high grade gliomas, blastomas, neuroblastomas, plasmacytomas, histiocytomas, melanomas, adenomas, hypoxic tumours, myelomas, AIDS-related lymphomas or sarcomas, metastatic cancers, or cancers in general.
86. A representative but non-limiting list of cancers that the disclosed compositions can be used to diagnose or prognose is the following: lymphoma, B cell lymphoma, T cell lymphoma, mycosis fungoides, Hodgkin's Disease, myeloid leukemia, bladder cancer, brain cancer, nervous system cancer, head and neck cancer, squamous cell carcinoma of head and neck, kidney cancer, lung cancers such as small cell lung cancer and non-small cell lung cancer, neuroblastoma/glioblastoma, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, liver cancer, melanoma, squamous cell carcinomas of the mouth, throat, larynx, and lung, colon cancer, cervical cancer, cervical carcinoma, breast cancer, and epithelial cancer, renal cancer, genitourinary cancer, pulmonary cancer, esophageal carcinoma, head and neck carcinoma, large bowel cancer, hematopoietic cancers; testicular cancer; colon and rectal cancers, prostatic cancer, or pancreatic cancer.
87. Compounds disclosed herein may also be used for the diagnosis or prognosis of precancer conditions such as cervical and anal dysplasias, other dysplasias, severe dysplasias, hyperplasias, atypical hyperplasias, and neoplasias. 'fλ," " έ li ά of identifying a minimal intrinsic gene set
88. Disclosed are methods of identifying minimal intrinsic genes. These methods are described in detail above, and generally comprise the following: deriving a minimal intrinsic gene set for making biological classifications of cancer comprising: a) collecting data from multiple samples from the same or different individuals to identify potential intrinsic classifier genes (microarray data can be used in this step, for example); b) weighting intrinsic classifier genes of multiple individuals identified using the method of step a relative to each other and forming classification clusters (weighting can be done, for example, by forming hierarchical clusters); c) estimating the number of clusters formed in step b) and assigning individual samples to clusters; d) identifying genes that optimally distinguish the samples in the assigned groups of step c); e) performing iterative cross-validation with a nearest centroid classifier and overlapping gene sets of various sizes using the genes identified in step d); and f) choosing a gene set which provides the highest class prediction accuracy when compared to the classifications made in step b).
89. Also disclosed is a method of assigning a sample to an intrinsic subtype, ' comprising a) creating an intrinsic subtype average profile (centroid) for each subtype; b) individually comparing a new sample to each centroid; and c) assigning the new sample to the centroid that is most similar to the new sample. This is known as the Single Sample Predictor (SSP) method, and is described in further detail in Example 2.
90. Also disclosed are computerized implementing systems, as well as storage and retrieval systems, of biological information, comprising: a data entry means; a display means; a programmable central processing unit; and a data storage means having expression data for a gene electronically stored; wherein the stored sequences are used as input data for determining which sequence is the best intrinsic gene set for a specific tissue type.
C. Compositions
91. Disclosed are the components to be used to prepare the disclosed compositions as well as the compositions themselves to be used within the methods disclosed herein. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular expression control gene is disclosed and discussed and a number of modifications that can be made to a number of molecules including the expression control gene are discussed, specifically contemplated is each an every com nat on an permutat on o expression control gene and the modifications that are possible unless specifically indicated to the contrary. Thus, if a class of molecules A, B, and C are disclosed as well as a class of molecules D, E, and F and an example of a combination molecule, A-D is disclosed, then even if each is not individually recited each is individually and collectively contemplated meaning combinations, A-E, A-F, B-D, B-E, B-F, C-D, C-E, and C-F are considered disclosed. Likewise, any subset or combination of these is also disclosed. Thus, for example, the subgroup of A-E, B-F, and C-E would be considered disclosed. This concept applies to all aspects of this application including, but not limited to, steps in methods of making and using the disclosed compositions. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the disclosed methods. 1. Sequence similarities
92. It is understood that as discussed herein the use of the terms homology and identity mean the same thing as similarity. Thus, for example, if the use of the word homology is used between two non-natural sequences it is understood that this is not necessarily indicating an evolutionary relationship between these two sequences, but rather is looking at the similarity or relatedness between their nucleic acid sequences. Many of the methods for determining homology between two evolutionarily related molecules are routinely applied to any two or more nucleic acids or proteins for the purpose of measuring sequence similarity regardless of whether they are evolutionarily related or not.
93. In general, it is understood that one way to define any known variants and derivatives or those that might arise, of the disclosed genes and proteins herein, is through defining the variants and derivatives in terms of homology to specific known sequences. This identity of particular sequences disclosed herein is also discussed elsewhere herein. In general, variants of genes and proteins herein disclosed typically have at least, about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent homology to the stated sequence or the native sequence. Those of skill in the art readily understand how to determine the homology of two proteins or nucleic acids, such as genes. For example, the homology can be calculated after aligning the two sequences so that the homology is at its highest level.
94. Another way of calculating homology can be performed by published algorithms. Optimal alignment of sequences for comparison may be conducted by the local homology algorithm of Smith and Waterman Adv. Appl. Math. 2: 482 (1981), by the homology alignment algorithm of Needleman and Wunsch, J. MoL Biol. 48: 443 (1970), by the search for similarity method of Pearson and Lipman, Proc. Natl. Acad. Sci. U.S.A. 85: 2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by inspection.
95. The same types of homology can be obtained for nucleic acids by for example the algorithms disclosed in Zuker, M. Science 244:48-52, 1989, Jaeger et al. Proc. Natl. Acad. Sci. USA 86:7706-7710, 1989, Jaeger et al. Methods Enzymol 183:281-306, 1989 which are herein incorporated by reference for at least material related to nucleic acid alignment. It is understood that any of the methods typically can be used and that in certain instances the results of these various methods may differ, but the skilled artisan understands if identity is found with at least one of these methods, the sequences would be said to have the stated identity, and be disclosed herein.
96. For example, as used herein, a sequence recited as having a particular percent homology to another sequence refers to sequences that have the recited homology as calculated by any one or more of the calculation methods described above. For example, a first sequence has 80 percent homology, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent homology to the second sequence using the Zuker calculation method even if the first sequence does not have 80 percent homology to the second sequence as calculated by any of the other calculation methods. As another example, a first sequence has 80 percent homology, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent homology to the second sequence using both the Zuker calculation method and the Pearson and Lipman calculation method even if the first sequence does not have 80 percent homology to the second sequence as calculated by the Smith and Waterman calculation method, the Needleman and Wunsch calculation method, the Jaeger calculation methods, or any of the other calculation methods. As yet another example, a first sequence has 80 percent homology, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent homology to the second sequence using each of calculation methods (although, in practice, the different calculation methods will often result in different calculated homology percentages). . yb i ization/selective hybridization
97. The term hybridization typically means a sequence driven interaction between at least two nucleic acid molecules, such as a primer or a probe and a gene. Sequence driven interaction means an interaction that occurs between two nucleotides or nucleotide analogs or nucleotide derivatives in a nucleotide specific manner. For example, G interacting with C or A interacting with T are sequence driven interactions. Typically sequence driven interactions occur on the Watson-Crick face or Hoogsteen face of the nucleotide. The hybridization of two nucleic acids is affected by a number of conditions and parameters known to those of skill in the art. For example, the salt concentrations, pH, and temperature of the reaction all affect whether two nucleic acid molecules will hybridize.
98. Parameters for selective hybridization between two nucleic acid molecules are well known to those of skill in the art. For example, in some embodiments selective hybridization conditions can be defined as stringent hybridization conditions. For example, stringency of hybridization is controlled by both temperature and salt concentration of either or both of the hybridization and washing steps. For example, the conditions of hybridization to achieve selective hybridization may involve hybridization in high ionic strength solution (6X SSC or 6X SSPE) at a temperature that is about 12-25°C below the Tm (the melting temperature at which half of the molecules dissociate from their hybridization partners) followed by washing at a combination of temperature and salt concentration chosen so that the washing temperature is about 50C to 2O0C below the Tm. The temperature and salt conditions are readily determined empirically in preliminary experiments in which samples of reference DNA immobilized on filters are hybridized to a labeled nucleic acid of interest and then washed under conditions of different stringencies. Hybridization temperatures are typically higher for DNA-RNA and RNA-RNA hybridizations. The conditions can be used as described above to achieve stringency, or as is known in the art. (Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989; Kunkel et al. Methods Enzymol. 1987:154:367, 1987 which is herein incorporated by reference for material at least related to hybridization of nucleic acids). A preferable stringent hybridization condition for a DNA:DNA hybridization can be at about 680C (in aqueous solution) in 6X SSC or 6X SSPE followed by washing at 68°C. Stringency of hybridization and washing, if desired, can be reduced accordingly as the degree of complementarity desired is decreased, and further, depending upon the G-C or A-T richness of any area wherein variability is searched for. Likewise, stringency of hybridization and washing, if desired, can be increased accordingly as homology desired is increased, and further, depending upon the G- C or A-T richness of any area wherein high homology is desired, all as known in the art.
99. Another way to define selective hybridization is by looking at the amount (percentage) of one of the nucleic acids bound to the other nucleic acid. For example, in some embodiments selective hybridization conditions would be when at least about, 60, 65, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 percent of the limiting nucleic acid is bound to the non-limiting nucleic acid. Typically, the non-limiting primer is in for example, 10 or 100 or 1000 fold excess. This type of assay can be performed at under conditions where both the limiting and non-limiting primer are for example, 10 fold or 100 fold or 1000 fold below their kd, or where only one of the nucleic acid molecules is 10 fold or 100 fold or 1000 fold or where one or both nucleic acid molecules are above their kd.
100. Another way to define selective hybridization is by looking at the percentage of primer that gets enzymatically manipulated under conditions where hybridization is required to promote the desired enzymatic manipulation. For example, in some embodiments selective hybridization conditions would be when at least about, 60, 65, 70, 71, 72, 73, 74, 75, 76, 77,
78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 percent of the primer is enzymatically manipulated under conditions which promote the enzymatic manipulation, for example if the enzymatic manipulation is DNA extension, then selective hybridization conditions would be when at least about 60, 65, 70, 71, 72, 73, 74, 75, 76, 77, 78,
79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 percent of the primer molecules are extended. Preferred conditions also include those suggested by the manufacturer or indicated in the art as being appropriate for the enzyme performing the manipulation.
101. Just as with homology, it is understood that there are a variety of methods herein disclosed for determining the level of hybridization between two nucleic acid molecules. It is understood that these methods and conditions may provide different percentages of hybridization between two nucleic acid molecules, but unless otherwise indicated meeting the parameters of any of the methods would be sufficient. For example if 80% hybridization was required and as long as hybridization occurs within the required parameters in any one of these methods it is considered disclosed herein. 102'. IFϊs"und'eS:sfood that those of skill in the art understand that if a composition or method meets any one of these criteria for determining hybridization either collectively or singly it is a composition or method that is disclosed herein. 3. Nucleic acids
103. There are a variety of molecules disclosed herein that are nucleic acid based, including for example the nucleic acids that encode, for example, the intrinsic genes disclosed herein (Table 12), as well as various functional nucleic acids. The disclosed nucleic acids are made up of for example, nucleotides, nucleotide analogs, or nucleotide substitutes. Non- limiting examples of these and other molecules are discussed herein. It is understood that for example, when a vector is expressed in a cell, that the expressed mRNA will typically be made up of A, C, G, and U. Likewise, it is understood that if, for example, an antisense molecule is introduced into a cell or cell environment through for example exogenous delivery, it is advantagous that the antisense molecule be made up of nucleotide analogs that reduce the degradation of the antisense molecule in the cellular environment. a) Nucleotides and related molecules
104. A nucleotide is a molecule that contains a base moiety, a sugar moiety and a phosphate moiety. Nucleotides can be linked together through their phosphate moieties and sugar moieties creating an internucleoside linkage. The base moiety of a nucleotide can be adenin-9-yl (A), cytosin-1-yl (C), guanin-9-yl (G), uracil-1-yl (U), and thymin-1-yl (T). The sugar moiety of a nucleotide is a ribose or a deoxyribose. The phosphate moiety of a nucleotide is pentavalent phosphate. An non-limiting example of a nucleotide would be 3'- AMP (3'-adenosine monophosphate) or 5'-GMP (5'-guanosine monophosphate). b) Primers and probes
105. It is understood that primers and probes can be produced for the actual gene (DNA) or expression product (mRNA) or intermediate expression products which are not fully processed into mRNA. Discussion of a particular gene is also a disclosure of the DNA, mRNA, and intermediate RNA products associated with that particular gene.
106. Disclosed are compositions including primers and probes, which are capable of interacting with the intrinsic genes disclosed herein, as well as the any other genes or nucleic acids discussed herein, hi certain embodiments the primers are used to support DNA amplification reactions. Typically the primers will be capable of being extended in a sequence specific manner. Extension of a primer in a sequence specific manner includes any methods wherein the sequence and/or composition of the nucleic acid molecule to which the primer is hybridized or otherwise associated directs or influences the composition or sequence of the product produced by the extension of the primer. Extension of the primer in a sequence specific manner therefore includes, but is not limited to, PCR, DNA sequencing, DNA extension, DNA polymerization, RNA transcription, or reverse transcription. Techniques and conditions that amplify the primer in a sequence specific manner are preferred. In certain embodiments the primers are used for the DNA amplification reactions, such as PCR or direct sequencing. It is understood that in certain embodiments the primers can also be extended using non-enzymatic techniques, where for example, the nucleotides or oligonucleotides used to extend the primer are modified such that they will chemically react to extend the primer in a sequence specific manner. Typically the disclosed primers hybridize with the disclosed genes or regions of the disclosed genes or they hybridize with the complement of the disclosed genes or complement of a region of the disclosed genes.
107. The size of the primers or probes for interaction with the disclosed genes in certain embodiments can be any size that supports the desired enzymatic manipulation of the primer, such as DNA amplification or the simple hybridization of the probe or primer. A typical disclosed primer or probe would be at least 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 61, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1250, 1500, 1750, 2000, 2250, 2500, 2750, 3000, 3500, or 4000 nucleotides long.
108. In other embodiments the disclosed primers or probes can be less than or equal to 6, 7, 8, 9, 10, 11, 12 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1250, 1500, 1750, 2000, 2250, 2500, 2750, 3000, 3500, or 4000 nucleotides long.
109. The primers for the disclosed genes in certain embodiments can be used to produce an amplified DNA product that contains the desired region of the disclosed genes. In general, typically the size of the product will be such that the size can be accurately determined to within 10, 5, 4, 3, or 2 or 1 nucleotides. 1 10. In certain embodiments this product is at least 20, 21, 22, 23, 24, 25, 27, 2 8 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 61, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1250, 1500, 1750, 2000, 2250, 2500, 2750, 3000, 3500, or 4000 nucleotides long.
111. In other embodiments the product is less than or equal to 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 61, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1250, 1500, 1750, 2000, 2250, 2500, 2750, 3000, 3500, or 4000 nucleotides long.
112. In certain embodiments the primers and probes are designed such that they are targeting as specific region in one of the genes disclosed herein. It is understood that primers and probes having an interaction with any region of any gene disclosed herein are contemplated: In other words, primers and probes of any size disclosed herein can be used to target any region specifically defined by the genes disclosed herein. Thus, primers and probes of any size can begin hybridizing with nucleotide 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or any specific nucleotide of the genes or gene expression products disclosed herein. Furthermore, it is understood that the primers and probes can be of a contiguous nature meaning that they have continuous base pairing with the target nucleic acid for which they are complementary. However, also disclosed are primers and probes which are not contiguous with their target complementary sequence. Disclosed are primers and probes which have at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 500, or more bases which are not contiguous across the length of the primer or probe. Also disclosed are primers and probes which have less than or equal to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 500, or more bases which are not contiguous across the length of the primer or probe.
113. In certain embodiments the primers or probes are designed such that they are able to hybridize specifically with a target nucleic acid. Specific hybridization refers to the ability to bind a particular nucleic acid or set of nucleic acids preferentially over other nucleic acids. The level of specific hybridization of a particular probe or primer with a target nucleic acid can be affected by salt conditions, buffer conditions, temperature, length of time of hybridization, wash conditions, and visualization conditions. By increasing the specificity of hybridization means decreasing the number of nucleic acids that a given primer or probe hybridizes to typically under a given set of conditions. For example, at 20 degrees Celsius under a given set of conditions a given probe may hybridize with 10 nucleic acids in a sample. However, at 40 degrees Celsius with all other conditions being equal, the same probe may only hybridize with 2 nucleic acids in the same sample. This would be considered an increase in specificity of hybridization. A decrease in specificity of hybridization means an increase in the number of nucleic acids that a given primer or probe hybridizes to typically under a given set of conditions. For example, at 700 mM NaCl under a given set of conditions a particular probe or primer may hybridize with 2 nucleic acids in a sample, however when the salt concentration is increased to 1 Molar NaCl the primer or probe may hybridize with 6 nucleic acids in the same sample.
114. The salt can be any salt such as those made from the alkali metals: Lithium, Sodium, Potassium, Rubidium, Cesium, or Francium or the alkaline earth metals: Beryllium, Magnesium, Calcium, Strontium, Barium, or Radiumsodium, or the transition metals: Scandium, Titanium, Vanadium, Chromium, Manganese, Iron, Cobalt, Nickel, Copper, Zinc, Yttrium, Zirconium, Niobium, Molybdenum, Technetium, Ruthenium, Rhodium, Palladium, Silver, Cadmium, Hafnium, Tantalum, Tungsten, Rhenium, Osmium, Iridium, Platinum, Gold, Mercury, Rutherfordium, Dubniuni, Seaborgium, Bohrium, Hassium, Meitnerium, Ununnilium, Unununium or Unuribium at any molar strength to promoter the desired condition, such as 1, 0.7, .5, 0.3, 0.2, 0.1, 0.05, or 0.02 molar salt, ha general increasing salt concentration decreases the specificity of a given probe or primer for a given target nucleic acid and decreasing the salt concentration increases the specificity of a given probe or primer for a given target nucleic acid.
115. The buffer conditions can be any buffer such as TRIS at any pH, such as 5.0, 5.5, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.1, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.5, or 9.0. In general pHs above or below 7.0 increase the specificity of hybridization.
116. The temperature of hybridization can be any temperature. For example, the temperature of hybridization can occur at 20°, 21°, 22°, 23°, 24°, 25°, 26°, 27°, 28°, 29°, 31°, 32°, 33°, 34°, 35°, 36°, 37°, 38°, 39°, 40°, 41°, 42°, 43°, 44°, 45°, 46°, 47°, 48°, 49°, 50°, 51°, 52°, 53°, 54°, 55°, 56°, 57°, 58°, 59°, 60°, 61°, 62°, 63°, 64°, 65°, 66°, 67°, 68°, 69°, 70°, 81°, 82°, 83°, 84°, 85°, 86°,87°, 88°, 89°, 90°, 91°, 92°, 93°, 94°, 95°, 96°, 97°, 98°, or 99° Celsius.
117. The length of time of hybridization can be for any time. For example, the length of time can be for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 120, 150, 180, 210, 240, 270, 300, 360, minutes or 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 30, 36, 48 or more hours.
118. It is understood that any wash conditions can be used including no wash step. Generally the wash conditions occur by a change in one or more of the other conditions designed to require more specific binding, by for example increasing temperature or decreasing the salt or changing the length of time of hybridization.
119. It is understood that there are a variety of visualization conditions which have different levels of detection capabilities. Li general any type of visualization or detection system can be used. For example, radiolabeling or fluorescence labeling can be used and in general fluorescence labeling would be more sensitive, meaning a fewer number of absolute molecules would have to be present to be detected.
120. c) Sequences
121. There are a variety of sequences related to the intrinsic genes as well as the others disclosed herein and others are herein incorporated by reference in their entireties as well as for individual subsequences contained therein. A specific intrinsic gene set can be found in Table 12.
4. Kits
122. Disclosed are kits comprising nucleic acids which can be used in the methods disclosed herein and, for example, buffers, salts, and other components to be used in the methods disclosed herein. Disclosed are kits for identifying minimal intrinsic gene sets comprising nucleic acids, such as in a microarray. Also disclosed are specific minimal intrinsic genes used for classifying cancer, such as those found in Table 21. As described above, these intrinsic genes can be used in any combination or permutation, and any combination of permutation of these genes can be used in a kit. Also disclosed are kits comprising instructions. 5. Chips and micro arrays
123. Disclosed are chips where at least one address is the sequences or part of the sequences set forth in any of the nucleic acid sequences disclosed herein.
124. Also disclosed are chips where at least one address is a variant of the sequences or part of the sequences set forth in any of the nucleic acid sequences disclosed herein.
6. Computer readable mediums
125. Those of skill in the art understand how to display and express any nucleic acid or protein sequence in any of the variety of ways that exist, each of which is considered herein disclosed. Specifically contemplated herein is the display of these sequences on computer readable mediums, such as, commercially available floppy disks, tapes, chips, hard drives, compact disks, and video disks, or other computer readable mediums. Also disclosed are the binary code representations of the disclosed sequences. Those of skill in the art understand what computer readable mediums. Thus, computer readable mediums on which the nucleic acids or protein sequences are recorded, stored, or saved.
126. Disclosed are computer readable mediums comprising the sequences and information regarding the sequences set forth herein.
D. Methods of making the compositions
127. The compositions disclosed herein and the compositions necessary to perform the disclosed methods can be made using any method known to those of skill in the art for that particular reagent or compound unless otherwise specifically noted.
1. Nucleic acid synthesis
128. For example, the nucleic acids, such as, the oligonucleotides to be used as primers can be made using standard chemical synthesis methods or can be produced using enzymatic methods or any other known method. Such methods can range from standard enzymatic digestion followed by nucleotide fragment isolation (see for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y., 1989) Chapters 5, 6) to purely synthetic methods, for example, by the cyanoethyl phosphoramidite method using a Milligen or Beckman System lPlus DNA synthesizer (for example, Model 8700 automated synthesizer of Milligen-Biosearch, Burlington, MA or ABI Model 380B). Synthetic methods useful for making oligonucleotides are also described by Dcuta et al., Ann. Rev. Biochem. 53:323-356 (1984), (phosphotriester and phosphite-triester methods), and Narang et al., Methods EnzymoL, 65:610-620 (1980), jfiosp otf es er metKo ). Protein nucleic acid molecules can be made using known methods such as those described by Nielsen et ah, Bioconjug. Chem. 5:3-7 (1994).
E. Methods of using the compositions
1. Methods of using the compositions as research tools
129. The disclosed compositions can be used in a variety of ways as research tools. The compositions can be used for example as targets in combinatorial chemistry protocols or other screening protocols to isolate molecules that possess desired functional properties related to the disclosed genes.
130. The disclosed compositions can also be used diagnostic tools related to diseases, such as cancers, such as those listed herein.
131. The disclosed compositions can be used as discussed herein as either reagents in micro arrays or as reagents to probe or analyze existing microarrays. The disclosed compositions can be used in any known method for isolating or identifying single nucleotide polymorphisms. The compositions can also be used in any method for determining allelic analysis of for example, the genes disclosed herein. The compositions can also be used in any known method of screening assays, related to chip/micro arrays. The compositions can also be used in any known way of using the computer readable embodiments of the disclosed compositions, for example, to study relatedness or to perform molecular modeling analysis related to the disclosed compositions.
132. Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon.
F. Examples
133. The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the compounds, compositions, articles, devices and/or methods claimed herein are made and evaluated, and are intended to be purely exemplary and are not intended to limit the disclosure. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in °C or is at ambient temperature, and pressure is at or near atmospheric. 1. E xample 1 : Biological Classification of Breast Cancer by Real-Time Quantitative RT-PCR: Comparisons to Microarray and Histopathology a) Methods
134. Patient selection. An ethnically diverse cohort of patients were studied using samples collected from various locations throughout the United States. Tissues analyzed included 117 invasive breast cancers, 1 fibroadenoma, 5 "normal" samples (from reduction mammoplasty), and 3 cells lines. Patients were heterogeneously treated in accordance with the standard of care dictated by their disease stage, ER and HER2 status. Patients were censored for recurrence and/or death for up to 118 months (median 21.5 months). Clinical data presented in supplementary Table 7.
135. Sample preparation and first strand synthesis for qRT-PCR. Nucleic acids were extracted from fresh frozen tissue using RNeasy Midi Kit (Qiagen Inc., Valencia, CA). The quality of RNA was assessed using the Agilent 2100 Bioanalyzer with the RNA 6000 Nano LabChip Kit (Agilent Technologies, Palo Alto, CA). All samples used had discernable 18S and 28S ribosomal peaks. First strand cDNA was synthesized from approximately 1.5 mg total RNA using 500 ng Oligo(dT)12-18 and Superscript TΩ. reverse transcriptase (1st Strand Kit, Invitrogen, Carlsbad, CA). The reaction was held at 42°C for 50 min followed by a 15-min step at 7O0C. The cDNA was washed on a QIAquick PCR purification column and stored at - 80°C in TE' (25 mM Tris, 1 mM EDTA) at a concentration of 5 ng/ul (concentration estimated from the starting RNA concentration used in the reverse transcription).
136. Primer design. Genbank sequences were downloaded from Evidence viewer (NCBI website) into the Lightcycler Probe Design Software (Roche Applied Science, Indianapolis, IN). All primer sets were designed to have a Tm » 6O0C, GC content » 50% and to generate a PCR amplicon <200 bps. Finally, BLAT and BLAST searches were performed on primer pair sequences using the UCSC Genome Bioinformatics (http://genome.ucsc.edu/) and NCBI (http://www.ncbi.nhn.nih.gov/BLAST/) to check for uniqueness. Primer sets and identifiers are provided in supplementary Table 8.
137. Real-time PCR. For PCR, each 20 μL reaction included IX PCR buffer with 3 mM MgC12 (Idaho Technology Inc., Salt Lake City, UT), 0.2 mM each of dATP, dCTP, and dGTP, 0.1 mM dTTP, 0.3 mM dUTP (Roche, Indianapolis, IN), 10 ng cDNA and IU Platinum Taq (Invitrogen, Carlsbad, CA). The dsDNA dye SYBR Green I (Molecular Probes, Eugene, OR) was used for all quantification (1/50000 final). PCR amplifications were performed on the Lightcycler (Roche, Indianapolis, IN) using an initial denaturation step (94 0C, 90 sec) owe y 5 cycles: 'denaturation (940C, 3 sec), annealing (58°C, 5 sec with 20°C/s transition), and extension (72°C, 6 sec with 2° C/sec transition). Fluorescence (530 nm) from the dsDNA dye SYBR Green I was acquired each cycle after the extension step. Specificity of PCR was determined by post-amplification melting curve analysis. Reactions were automatically cooled to 60°C at a rate of 3°C/s and slowly heated at 0.1 °C/s to 95°C while continuously monitoring fluorescence.
138. Relative quantification by RT-PCR. Quantification was performed using the LightCycler 4.0 software. The crossing threshold (Ct) for each reaction was determined using the 2nd derivative maximum method (Wittwer et al. (2004) Washington, DC: ASM Press; Rasmussen (2001) Heidelberg: Springer Verlag. 21-34). Relative copy number was calculated using an external calibration curve to correct for PCR efficiency and a within run calibrator to correct for the variability between run. The calibrator is made from 4 equal parts of RNA from 3 cell lines (MCF7, SKBR3, ME16C) and Universal Human Reference RNA (Stratagene, La Jolla, CA, Cat #740000). Differences in cDNA input were corrected by dividing target copy number by the arithmetic mean of the copy number for 3 housekeeper genes (MRPLl 9, PSMC4, and PUMl) ( Szabo et al. (2004) Genome Biol 5:R59). The normalized relative gene copy number was Iog2 transformed and analyzed by hierarchical clustering using Cluster (Eisen et al. (1998) Proc Natl Acad Sci U S A 95:14863-14868). The clustering was visualized using Treeview software (Eisen Lab, http:/rana.lbl.gov/EisenSoftware.htm).
139. Microarray experiments. The same 126 samples used for qRT-PCR were analyzed by microarray (Agilent Human oligonucleotide). Total RNA was prepared and quality checked as described above. Labeling and hybridization of RNA for microarray was done using the Agilent low RNA input linear amplification kit
(http://www.chem.agilent.com/Scripts/PDS. asp?lPage=10003), but with one-half the recommended reagent volumes and using a Qiagen PCR purification kit to clean up the cRNA. Each sample was assayed versus a common reference sample that was a mixture of Stratagene's Human Universal Reference total RNA (lOOug) enriched with equal amounts of RNA (0.3 μg each) from MCF7 and ME16C cell lines. Microarray hybridizations were carried out on Agilent Human oligonucleotide microarrays (1 A-vl, 1 A-v2 and custom designed 1 A-vl based microarrays) using 2 μg each of Cy3-labeled "reference" and Cy5-labeled "experimental" sample. Hybridizations were done using the Agilent hybridization kit and a Robbins Scientific "22k chamber" hybridization oven. The arrays were incubated overnight and then washed once in 2X SSC and 0.0005% triton X-102 (10 min), twice in 0.1XSSC (5 πiih)"," aiidlnen immersed' into Agilent Stabilization and Drying solution for 20 seconds. All microarrays were scanned using an Axon Scanner 4000A. The image files were analyzed with GenePix Pro 4.1 and loaded into the UNC Microarray Database at the University of North Carolina at Chapel Hill (https://genome.unc.edu/ ) where a lowess normalization procedure was performed to adjust the Cy3 and Cy5 channels (Yang et al. (2002) Nucleic Acids Res 30:el5). All primary microarray data associated with this study are available at the UNC Microarray Database and have been deposited into the GEO (http://www.ncbi.nlm.nih.gov/geo/) under the accession number of GSE1992, series GSM34424-GSM34568.
140. Selecting genes for real-time qRT-PCR. A new "intrinsic" gene set for classifying breast tumors was derived using 45 before and after therapy samples from the combined data sets presented in Sorlie et al. (see Table 9 for the list of 45 pairs). The two-color DNA microarray data sets were downloaded from the internet and the R/G ratio (experimental/reference) for each spot was normalized and Iog2 transformed. Missing values were imputed using the k-NN imputation algorithm described by Troyanskaya et al. (Troyanskaya et al. (2001) Bioinformatics 17:520-525). The "intrinsic" analysis identified 550 gene elements.
141. Next, a completely independent data set was utilized (van't Veer et al. 2002) to derive an optimized version of the 550 intrinsic gene list. To allow across data set analyses, gene annotation from each dataset was translated to UniGene Cluster IDs (UCID) using the SOURCE database ( Diehn et al. (2003) Nucleic Acids Res 31:219-223). Following the alogorithm outlined by Tibshirani and colleagues ( Bair et al. (2004) PLoS Biol 2:E108; Bullinger et al. (2004) N Engl J Med 350:1605-1616), the 97 samples from the van't Veer et al. 2002 study were hierarchical clustered using a common set of 350 genes, and assigned an "intrinsic subtype of either Luminal, HER2+/ER-, Basal-like, or Normal-like to each sample. A feature/gene selection was then performed to identify genes that optimally distinguished these
4 classes using a version of the gene selection method first described by Dudoit et al. (Genome Biol 3:RESEARCH0036), where the best class distinguishers are identified according to the ratio of between-group to within-group sums of squares (a type of ANOVA). In addition to statistically selecting "intrinsic" classifiers proliferation genes (e.g., TOP2A, KI-67, PCNA) were also chosen, and other important prognostic markers (e.g., PgR) that have potential for diagnostics. In total, 53 differentially expressed biomarkers were used in the real-time qRT- PCR assay (Table 8). ""14'Z. Combining" microarray and qRT-PCR datasets. Distance We ghted Discrimination (DWD) was used to identify and correct systematic biases across the microarray and qRT-PCR datasets (Benito et al. (2004) Bioinformatics 20:105-114). Prior to DWD, each dataset was normalized by setting the mean to zero and the variance to one. Normalization was done within each microarray experiment and for genes profiled across many experimental runs for real-time qRT-PCR. After DWD, genes in common between the datasets were clustered using Spearman correlation and average linkage association.
143. Receiver operator curves. In order to determine agreement between protein expression (immunohistochemistry) and gene expression (qRT-PCR), a cut-off for relative gene copy number was selected by minimizing the sum of the observed false positive and false negative errors. That is, minimizing the estimated overall error rate under equal priors for the presence/absence of the protein. The sensitivity and specificity of the resulting classification rule were estimated via bootstrap adjustment for optimism (Efron et al. (1998) CRC Press LLC. p 247 pp).
144. Survival analyses. Survival curves were estimated by the Kaplan-Meier method and compared via a log-rank or stratified log-rank test as appropriate. Standard clinical pathological parameters of age (in years), node status (positive vs. negative), tumor size (cm, as a continuous variable), grade (1-3, as a continuous covariate), and ER status (positive vs. negative) were tested for differences in RFS and OS using Cox proportional hazards regression model. Pairwise log-rank tests were used to test for equality of the hazard functions among the intrinsic classes. Only the classes Luminal, HER2+/ER-, and Basal-like classes were included in the analyses because it was believed the Normal Breast-like subtype is not a pure tumor class and may result from normal breast contamination. Cox regression was used to determine predictors of survival from continuous expression data. All statistical analyses were performed using the R statistical software package (R Foundation for Statistical Computing). b) Results
145. Recapitulating microarray breast cancer classifications by qRT-PCR. 126 different breast tissue samples (117 invasive, 5 normal, 1 fibroadenoma, and 3 cell lines) were expression profiled using a real-time qRT-PCR assay comprised of 53 biological classifiers and 3 control/housekeepers genes. Genes were statistically selected to optimally identify the 4 main breast tumor intrinsic subtypes, and to create an objective gene expression predictor for cell proliferation and outcome (Ross et al. (2000) Nat Genet 24:227-235). . Tnere were 402 genes in common between this microarray dataset and the 550 "intrinsic" genes selected from the Sorlie et al. 2003 study. Two-way hierarchical clustering of the 402 genes in the microarray gave the same tumor subtypes as the minimal 37 "intrinsic" genes assayed by qRT-PCR (Figure 4). The samples were grouped into Luminal, HER2+/ER-, Normal-like, and Basal-like subtypes. Out of 123 breast samples compared across the platforms, 114 (93%) were classified the same. The minimal "intrinsic" gene set identified expression signatures within the 3 different cell lines that were characteristic of each tumor subtype: Luminal (MCF7), HER2+/ER- (SKBR3), and Basal-like (MEl 6C). The genes EGFR and PgR, which were added for their predictive and prognostic value in breast cancer Nielsen et al. (2004) Clin Cancer Res 10:5367-5374; Makretsov et al. (2004) Clin Cancer Res 10:6143- 6151), had opposite expression and were found to associate with either ER-positive tumors (high expression of PgR) or ER-negative tumors (high expression of EGFR) (Fig. 4C).
147. Proliferation and grade. Expression of the 14 "proliferation" genes (Fig. 4D) assayed by qRT-PCR showed that Luminal tumors have relatively low replication activity compared to HER2+/ER- and Basal-like tumors. As expected, the Normal-like samples showed the lowest expression of the "proliferation" genes. When correlating (Spearman correlation) the gene expression of all 53 genes with grade, it was found that the top 3 proliferation genes with a positive correlation (i.e., high expression correlates with high grade) were the proliferation genes CENPF (p=2.00E-07), BUBl (p=6.84E-07), and STK6 (p=2.67E- 06) (see supplementary Table 10). Interestingly, all thejproliferation genes, except PCNA, were at the top of the list for having a positive correlation to grade. Conversely, the top markers with significant negative correlations with grade (i.e., low expression correlates with high grade) were GATA3 (p=3.53E-07), XBPl (p=9.64E-06), and ESRl (p=4.53E-05).
148. Agreement between immunohistochemistry, qRT-PCR "intrinsic" classifications, and gene expression. Fifty out of fifty-five (91%) Luminal tumors with IHC data were scored positive for ER. Conversely, 50 out of 56 (89%) tumors classified as HER2+/ER- or Basal-like were negative for ER by IHC. Cluster analysis showed that the Luminal tumors co-express ER and estrogen responsive genes such as LIV1/SLC39A6, X-box binding protein 1 (XBPl), and hepatocyte nuclear factor 3a (HNF3A/FOXA1). The gene with the highest correlation in expression to ESRl was GATA3 (0.79, 95% CI: 0.71 - 0.85). It was found that the gene expression of ESRl alone had 88% sensitivity and 85% specificity for calling ER status by IHC, and GATA3 alone showed 79% sensitivity and 88% specificity (Figure 5A). In addition, gene expression of PgR correlated well with PR IHC status (sensitivity=89%, spec 'c y=82 ) F' g" B). The data showed a very high correlation in expression between HER2/ERBB2 and GRB7 (0.91, 95% CI: 0.87 - 0.94), which are physically located near one another and are commonly overexpressed and DNA amplified together (Pollack et al. (1999) Nature Genetics 23:41-46; Pollack et al. (2002) Proc Natl Acad Sci U S A 99:12963-12968). However, neither ERBB2 (sensitivity=91%, sρecificity=54%) nor GRB7 (sensitivity=52% specificity=78%) gene expression had both high sensitivity and specificity for predicting HER2 status by mC (Fig 5C).
149. Reproducibility ofqRT-PCR. The run-to-run variation in Cp (cycle number determined from fluorescence crossing point) for all 56 genes (53 classifiers and 3 housekeepers) was determined from 8 runs. The median CV (standard deviation/mean) for all the genes was 1.15% (0.28%-6.55%) and 51/56 genes (91%) had a CV <2%. The reproducibility of the classification method is illustrated from the observation that replicates of the same sample (UB57A&B and UB60A&B), cluster directly adjacent to one another. Notably, the replicates were from separate RNA/cDNA preparations done on different pieces of the same tumor.
150. Survival Predictors. The clinical significance of individual markers and "intrinsic" subtypes were analyzed using qRT-PCR data. Patients with Luminal tumors showed significantly better outcomes for relapse-free survival (RPS) and overall survival (OS) compared to HER2+/ER- (RFS: p=0.023; OS: p=0.003) and Basal-like (RFS: ρ=0.065; OS: p=0.002) tumors (Figure 6). This difference in outcome was significant for overall survival even after adjustment for stage (HER2+/ER-: p=0.043; Basal-like: p=0.001). There was no difference in outcome between patients with HER2+/ER- and Basal-like tumors. Analysis of the same cohort using standard clinical pathological information shows that stage, tumor size, node status, and ER status were prognostic for RFS and OS.
151. Using a Cox proportional hazards model to find biomarkers from the qRT-PCR data that predict survival, it was found that high expression of the proliferation genes GTBP4 (ρ=0.011), HSPA14 (p=0.023), and STK6 (ρ=0.027) were significant predictors of RFS independent of grade and stage (Figure 7). The only proliferation gene significant for OS after correction for grade and stage was GTBP4 (p=0.011). Overall, the best predictor for both RFS (p=0.004) and OS (ρ=0.004) independent of grade and stage was SMA3 (Table 10).
152. Co-clustering qRT-PCR and Microarray Data. In order to determine if qRT-PCR and microarray data could be analyzed together in a single dataset, DWD was used to combine data for 50 genes and 126 samples profiled on both platforms (252 samples total). Hierarchical clus r ng f tl ese ata s ow that 98% (124/126) of the paired samples classified in the same group and 83/126 (66%) clustered directly adjacent to their corresponding partner (Figure 10). Thus, DNA microarray and real-time qRT-PCR can be combined into a seamless dataset without sample segregation based on platform. Overall, the correlation between microarray and qRT-PCR expression data was 0.76 (95% CI: 0.75, 0.77) before DWD and 0.77 (95% CI: 0.76, 0.78) after DWD (Figure 5). The DWD does not significantly effect the correlation but corrects for systematic biases between the platforms. c) Discussion
153. Gene expression analyses can identify differences in breast cancer biology that are important for prognosis. However, a major challenge in using genomics for diagnostics is finding biomarkers that can be reproducibly measured across different platforms and that provide clinically significant classifications on different patient populations. Using microarray data, 402 "intrinsic" genes were identified that classify breast cancers based on vastly different expression patterns. This "intrinsic" gene set was shown to provide the same classifications when applied to a completely new and ethnically diverse population. Furthermore, the microarray dataset can be minimized to 37 "intrinsic" genes, translated into a real-time qRT- PCR assay, and provide the same classifications as the larger gene set. Molecular classifications using the "intrinsic" qRT-PCR assay agree with standard pathology and are clinically significant for prognosis. Thus, biological classifications based on "intrinsic" genes are robust, reproducible across different platforms, and can be used for breast cancer diagnostics.
154. The greatest contribution genomic assays have made towards clinical diagnostics in breast cancer has been in identifying risk of recurrence in women with early stage disease. For instance, MammaPrint™ is a microarray assay based on the 70 gene prognosis signature originally identified by van't Veer et al. On the test set validation, the 70 gene assay found that individuals with a poor prognostic signature had approximately a 50% chance of remaining free of distant metastasis at 10 years while those with a good-prognostic signature had a 85% chance of remaining free of disease. Another assay with similar utility is Oncotype Dx (Genomic Health Lie) - a real-time qRT-PCR assay that uses 16 classifiers to assess if patients with ER positive tumors are at low, intermediate, or high risk for relapse. While recurrence can be predicted with high and low risk tumors, patients in the intermediate risk group still have variable outcomes and need to be diagnosed more accurately. . _, f "55. In genef aij'tumors that have a low risk of early recurrence are low grade and have low expression of proliferation genes. Due to the correlation of proliferation genes with grade and their significance in predicting outcome, a group of 14 proliferation genes were assayed. While the classic proliferation markers TOP2A and MKI67 significantly correlated with grade in the cohort, they were not near the top of the list. Furthermore, PCNA did not significantly correlate with grade (p=0.11) in the cohort. This could result from PCR primer design or differences between RNA and protein stability. Nevertheless, the proliferation gene that was found had the highest correlation to grade was CENPF (mitosin); another commonly used mitotic marker that has been shown to correlate with grade and outcome in breast cancer (Clark et al. (1997) Cancer Res 57:5505-5508). Since tumor grade and the mitotic index have been shown to be important in predicting risk of relapse (Chia et al. (2004) J Clin Oncol 22:1630-1637; Manders et al. (2003) Breast Cancer Res Treat 77:77-84), it is not surprising that 4 (GTBP4, HSPAl 4, STK6/15, BUBl) out the top 5 predictors for RFS (independent of stage) were proliferation genes. The proliferation gene that was the best predictor of RFS was GTBP4, a GTP-binding protein implicated in chronic renal disease and shown to be upregulated after serum administration (i.e., serum response gene) (Laping et al. (2001) J Am Soc Nephrol 12:883-890). Overall, the best predictor for both RFS (ρ=0.004) and OS (p=0.004) independent of grade and stage was SMA3. The role of SMA3 in the pathogenesis of breast cancer is still unclear, although it has also been associated with the BCL2 anti- apoptotic pathway (Iwahashi et al. (1997) Nature 390:413-417).
2. Example 2: A New Breast Tumor Intrinsic Gene List Identifies Novel Characteristics that are Conserved Across Microarray Platforms
156. A training set of 105 tumors were used to derive a new breast tumor "intrinsic" gene list and validated it using a combined test set of 315 tumors compiled from three independent microarray studies. An unchanging Single Sample Predictor was also used, and applied to three additional test sets. The Mrinsic/UNC gene set identified a number of findings not seen in previous analyses including 1) significance in multivariate testing, 2) that the proliferation signature is an intrinsic property of tumors, 3) the high expression of many Kallikrein genes in Basal-like tumors, and 4) the expression of the Androgen Receptor within the HER2+/ER- and Luminal tumor subtypes. The Single Sample Predictor that was based upon subtype average profiles, was able to identity groups of patients within a test set of local therapy only patients, and two independent tamoxifen-treated patient sets, which showed significant differences in outcomes. The analyses demonstrates that the "intrinsic" subtypes add valtL'et6''the ex'ϊstϊri'g fep*ertoire of clinical markers used for breast cancer patients. The computation approach also provides a means for quickly validating gene expression profiles using publicly available data.
157. Breast cancers represent a spectrum of diseases comprised of different tumor subtypes, each with a distinct biology and clinical behavior. Despite this heterogeneity, global analyses of primary breast tumors using microarrays have identified gene expression signatures that characterize many of the essential qualities important for biological and clinical classification. Using cDNA microarrays, five distinct subtypes of breast tumors arising from at least two distinct cell types (basal-like and luminal epithelial cells) were previously identified (Perou et al. 2000; Sorlie et al. 2001; Sorlie et al. 2003). This molecular taxonomy was based upon an "intrinsic" gene set, which was identified using a supervised analysis to select genes that showed little variance within repeated samplings of the same tumor, but which showed high variance across tumors (Perou et al. 2000). An intrinsic gene set reflects the stable biological properties of tumors and typically identifies distinct tumor subtypes that have prognostic significance, even though no knowledge of outcome was used to derive this gene set.
158. 315 breast tumor samples compiled from publicly available microarray data were generated on different microarray platforms. These analyses show for the first time, that the breast tumor intrinsic subtypes are significant predictors of outcome when correcting for standard clinical parameters, and that common patterns of expression and outcome predictions can be identified when comparing data sets generated by independent labs. a) METHODS
159. Tissue samples, RNA preparations and microarray protocols. 105 fresh frozen breast tumor samples and 9 normal breast tissue samples were used as the training set and were obtained from 4 different sources using IRB approved protocols from each participating institution: the University of North Carolina at Chapel Hill, The University of Utah, Thomas Jefferson University and the University of Chicago. Thus, this sample set represents an ethnically diverse cohort from different geographic regions in the US with the clinical and microarray data for samples provided in Table 11. Patients were heterogeneously treated in accordance with the standard of care dictated by their disease stage, ER and HER2 status. The 105 patient training data set had a median follow up of 19.5 months, while the 315 sample combined test set had a median follow up of 74.5 months. Finally, another 16 tamoxifen- treated patient "tumbr'safnples were included that were used for the Single Sample Predictor additional test set analysis (tamoxifen-treated set #2).
160. Total RNA was purified from each sample using the Qiagen RNeasy Kit according to the manufacturer's protocol (Qiagen, Valencia CA) and using 10-50 milligram of tissue per sample. The integrity of the RNA was determined using the RNA 6000 Nano LabChip Kit and an Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, CA). The total RNA labeling and hybridization protocol used is described in the Agilent low RNA input linear amplification kit (http://www.chem.agilent.com/Scripts/PDS. asp?lPage=10003 ) with the following modifications: 1) a Qiagen PCR purification kit was used to clean up the cRNA and 2) all reagent volumes were cut in half. Each sample was assayed versus a common reference sample that was a mixture of Stratagene's Human Universal Reference total RNA (Novoradovskaya et al. 2004) (100ug) enriched with equal amounts of RNA (0.3 μg each) from MCF7 and ME16C cell lines. Microarray hybridizations were carried out on Agilent Human oligonucleotide microarrays (lA-vl, 1A-V2 and custom designed lA-vl based microarrays) using 2μg of Cy3-labeled Reference and 2μg of Cy5-labeled experimental sample. Hybridizations were done using the Agilent hybridization kit and a Robbins Scientific "22k chamber" hybridization oven. The arrays were incubated overnight and then washed once in 2X SSC and 0.0005% triton X-105 (10 min), twice in 0.1XSSC (5 min), and then immersed into Agilent Stabilization and Drying solution for 20 seconds. AU microarrays were scanned using an Axon Scanner GenePix 4000B. The image files were analyzed with GenePix Pro 4.1 and loaded into the UNC Microarray Database at the University of North Carolina at Chapel Hill (https://genome.unc.edu/) where a Lowess normalization procedure was performed to adjust the Cy3 and Cy5 channels(Yang et al. 2002). All primary microarray data associated with this study are available at https://genome.unc.edu/pubsup/breastTumor/ and have been deposited into the GEO (http://www.ncbi.nlm.nih.gov/geo/) under the accession number of GSE1992, series GSM34424-GSM34568.
161. Intrinsic gene set analysis. A new breast tumor intrinsic gene set was derived, called the "Intrinsic/UNC" list using 105 patients (146 total arrays) and 15 repeated tumor samples that were different physical pieces (and RNA preparations) of the same tumor, 9 tumor-metastasis pairs and 2 normal sample pairs (26 paired samples in total, Table 11). This sample size was chosen based upon Basal-like, Luminal A, Luminal B, HER2+/ER-, and Normal-like samples, which occur at a frequency of 15%, 40%, 15%, 20%, and 10%, respectively; and it was estimated that most clinically relevant classes would constitute at least lU%"of the affected population, and it was hoped to acquire at least 10 samples from each class in the new data set. Therefore, a sample size of 100 tumors was deemed adequate to identify most classes that might be present in breast cancer patients.
162. The background subtracted, Lowess normalized Iog2 ratio of Cy5 over Cy3 intensity values were first filtered to select genes that had a signal intensity of at least 30 units above background in both the Cy5 and Cy3 channels. Only genes that met these criteria in at least 70% of the 146 microarrays were included for subsequent analysis. Next, an "intrinsic" analysis was performed as described in Sorlie et al. 2003 (Sorlie et al. 2003) using the 26 paired samples and 86 additional microarrays. An intrinsic analysis identifies genes that have low variability in expression within paired samples and high variability in expression across different tumors; for an intrinsic analysis, each gene receives a score that is the average "within-pair variance" (the average square before/after difference), as well as the "between- subject variance" (the variance of the pair averages across subjects). The ratio D=(within-pair variance)/(between-subject variance) was then computed, and those genes with a small value of D (i.e. cut-off) declared to be "intrinsic". The choice of a value of D was set at one standard deviation below the mean intrinsic score of all genes. This analysis resulted in the selection of 1410 microarray elements representing 1300 genes. Ih order to obtain an estimate of the number of false-positive intrinsic genes, the sample labels were permuted to generate 26 random pairs and 86 non-paired samples. This permutation was performed 100 times and the intrinsic scores were calculated for each. These permuted scores were used to determine a threshold on the intrinsic score corresponding to a false discovery rate less than 1%. The selected threshold resulted in 1410 microarray features being called significant with a FDR=0.3% and the 90th percentile FDR=0.5%. (See Tusher et al. for a complete description of this calculation (Tusher et al. 2001)).
163. These 1410 microarray elements were then used to perform a two-way average linkage hierarchical cluster analysis using a centered Pearson correlation metric and the program "Cluster" (Eisen et al. 1998), with the data being displayed relative to the median expression for each gene (i.e. median centering of the rows/genes). The cluster results were then visualized using "Treeview".
164. Combined test set analysis. The two-color DNA microarray data sets of Sorlie et al. 2001 and 2003 van't Veer et al. and Sotiriou et al. (Sotiriou et al. 2003) were each downloaded from the internet and pre-processed similarly. Briefly, pre-processing included Iog2 transformation of the R/G ratio and then Lowess normalization of the data set (Yang et al. 2002 J. Next, missing values were imputed using the k-NN imputation algorithm described by Troyanskaya et al. (Troyanskaya et al. 2001). Gene annotation from each dataset was translated to UniGene Cluster IDs (UCTD) using the SOURCE database (Diehn et al. 2003), which gave a common gene set of approximately 2800 genes that were present across all four data sets. UniGene was chosen because a majority of the identifiers from each dataset could be easily mapped to a UniGene identifier (Build 161). Multiple occurrences of a UCDD were collapsed by taking the median value for that E) within each experiment and platform. Next, Distance Weighted Discrimination was performed in a pair-wise fashion by first combining the Sorlie et al. data set with the Sotiriou et al. data set, and then combining this with the van't Veer et al. data to make a single data set. hi the final step of pre-processing, each individual experiment (microarray) was normalized by setting the mean to zero and its variance to one. The data for 306 of the 1300 Intrinsic/UNC genes was present in the combined test set and was used in a two-way average linkage hierarchical cluster analysis across the set of 315 microarrays as described above.
165. Single Sample Predictor. The Single Sample Predictor/SSP is based upon the Nearest Centroid method presented in (Hastie et al. 2001). More specifically, the combined test set was utilized, and 306 Litrinsic/UNC gene set hierarchical cluster presented in Figure 14, as the starting point to create five Subtype Mean Centroids. A mean vector (centroid) for each of the five intrinsic subtypes (LumA, LumB, HER2+/ER-, Basal-like and Normal Breast-like) was created by averaging the gene expression profiles for the samples clearly assigned to each group (which limited the analysis to 249 samples total); the hierarchical clustering dendrogram in Figure 14 were used as a guide for deciding those samples to group together. Next, using the 249 samples and 306 genes as a new training set (see Figure 11), the SSP was applied back onto this data set (only the 249 samples) using Spearman correlation (which will calculate a training set error rate) and assigned a sample to the subtype to which it was most similar. This analysis showed 92% concordance with the clustering based subtype assignments.
166. Three additional test data sets were then analyzed: First the 60 sample data set of Ma et al. (Ma et al. 2004) was taken, which is an already pre-processed data set of Iog2 transformed ratios (GEO GSE1379), and performed a DWD correction using the 278 genes that were in common between the Ma et al. data set and the set of 306 Mrinsic/UNC genes used in the SSP. The SSP was applied to the 60 Ma et al. samples and, using Spearman correlation, each of the 60 samples were assigned to an intrinsic subtype based upon the highest correlation value to a centroid. Next, 220 samples from Chang et al.(Chang et al. 2005) were analyzed and 16 additional samples from UNC that were not used in the training set. The 220 samples represent an extension of the sample set presented in van't Veer et al.(van 't Veer et al. 2002), and the combination of these two are the data used in van de Vijver et al.(van de Vijver et al. 2002). Each sample was column-standardized and then performed DWD to combine the 249 SSP samples (306 intrinsic genes) with the 220 samples from Chang et al. and the 16 UNC additional test set samples. Next, each sample's correlation to each centroid was calculated using a Spearman correlation and a sample was assigned to the centroid it was closest to, and the test set was then split into a local only therapy test set, and a tamoxifen- treated test set. Finally, the SSP was applied to the 105 sample original training set after DWD normalization.
167. Survival analyses. Univariate Kaplan-Meier analysis using a log-rank test was performed using WinSTAT for excel (R. Fitch Software). Standard clinical pathological parameters of age (in decades), node status (positive vs. negative), tumor size (categorical variable of T1-T4), grade (I vs. II and I vs. ID), and ER status (positive vs. negative) were tested for differences in RFS, OS and DSS using a proportional hazards regression model. The likelihood ratio test was used to test for equality of the hazard functions among the intrinsic classes after adjusting for the covariates listed above. For the intrinsic subtype analyses, the coding was such that LuniA was the reference group to which the other classes were compared. SAS (SAS Institute Inc., SAS/STAT User's Guide, Version 8, 1999, Cary, NC) was used for proportional hazards modeling.
168. Immunohistochemistry. Five micron sections from formalin-fixed, paraffin- embedded tumors were cut and mounted onto Probe On Plus slides (Fisher Scientific). Following deparaffinization in xylene, slides were rehydrated through a graded series of alcohol and placed in running water. Endogenous peroxidase activity was blocked with 3% hydrogen peroxidase and methanol. Samples were steamed for antigen retrieval with 10 mM citrate buffer (pH 6.0) for 30 min. Following protein block, slides were incubated with biotinylated antibody for the Androgen Receptor (Zymed, 08-1292) and incubated with streptavidin conjugated HRP using Vectastain ABC kit protocol (Vector Laboratories). 3,3'- diaminobenzidine tetrahydrochloride (DAB) chromogen (the substrate) was used for the visualization of the antibody/enzyme complex. Slides were counterstained with hematoxylin (Biomeda-MlO) and examined by light microscopy. b) RESULTS
169. Overview. The goals were to create a new breast tumor intrinsic list and validate this list using multiple test data sets so that new biology could be identified, and the clinical significance of "intrinsic" classifications shown. A new intrinsic list was created using paired samples that were similarly treated (note that these were different "intrinsic" pairs than previously used since they were not before and after therapy pairs). In deriving the "new" list microarrays containing many more thousands of genes than was used before were used. A diagram representing the flow of data sets used here, and the different analysis methods, is presented in Figure 11. First, a new 1300 gene "Ihtrinsic/UNC" list was created using 26 paired samples and a "training set" of 105 patients. Second, a large "combined test set" of 315 samples was created by combining three publicly available data sets. A reduced version of the Intrinsic/UNC gene set (reduced to an overlapping set of 306 genes) was applied onto this pure test set and show significance in a multivariate analysis. Finally, using the "combined test set", a Single Sample Predictor (SSP) was created from the subtype average profiles (i.e. centroids) and assign subtype designation onto three "additional test sets". Thus, the "combined test set" becomes the training set for the SSP, which is then used to predict subtype, and ultimately outcome, on the "additional test sets".
170. Identification of the Intrinsic/UNC gene set. A new breast tumor intrinsic gene set was created, called the "Intrinsic/UNC" list, using 26 paired samples comprised of 15 paired primary tumors that were different physical pieces (and RNA preparations) of the same tumor, 9 primary tumor-metastasis pairs, and 2 normal breast sample pairs. In total, 105 biologically diverse breast tumor specimens and 9 normal breast samples (146 microarrays, see Table 11) were assayed on Agilent oligo DNA microarrays representing 17,000 genes (GEO accession number GSE 1992). This intrinsic analysis identified 1410 microarray elements that represented 1300 genes. When this new gene list was used in a two-way hierarchical clustering analysis on the training set (Figure 12), the experimental sample dendrogram (Figure 12B) showed four groups corresponding to the previously defined HER2+/ER-, Basal-like, Luminal and Normal Breast-like groups (Perou et al. 2000). AU 26 tumor pairs were paired in this clustering analysis, including the 5 primary tumor-local metastasis pairs and the 4 distant metastasis pairs (Figure 12); thus, the individual portraits of tumors are maintained even in their metastasis samples (Weigelt et al. 2003).
171. The biology of the intrinsic subtypes is rich and extensive, and the current analysis identified new biologically important features. A HER2+ expression cluster was observed that contained genes from the 17ql 1 amplicon including HER2/ERBB2 and GRB7 (Figure 12D). The HER2+ expression subtype (pink dendrogram branch in Figure 12B) was predominantly ER-negative (i.e. HER2+/ER-), but showed expression of the Androgen Receptor (AR) gene. To determine if this finding extended to the protein level, immunohistochemistry for AR was performed, and it was confirmed that the HER2+/ER- and many Luminal tumors, expressed AR at moderate to high levels (Figure 13); in some cases, high nuclear expression was observed (Figure 13B).
172. A Basal-like expression cluster was also present and contained genes characteristic of basal epithelial cells such as SOX9, CKl 7, c-KIT, FOXCl and P-Cadherin (Figure 13E). These analyses extend the Basal-like expression profile to contain four Kallikrein genes (KLK5-8), which are a family of serine proteases that have diverse functions and proven utility as biomarkers (e.g. KLK3/PSA); however, it should be noted that KLK3/PSA was not part of the basal profile. Finally, a Luminal/ER+ cluster was present and contained ER, XBPl, FOXAl and GAT A3 (Figure 12C). GATA3 has recently been shown to be somatically mutated in some ER+ breast tumors (Usary et al. 2004), and some of the genes in Figure 12C are GAT A3 -regulated (FOXAl, TFF3 and AGR2). In addition, the Luminal/ER+ cluster contained many new biologically relevant genes such as AR (Figure 12C), FBPl (a key enzyme in gluconeogenesis pathway) and BCMPl 1.
173. The subtype defining genes from this analysis showed similarity to the previous breast tumor intrinsic lists (i.e. hitrinsic/Stanford) described in (Perou et al. 2000; Sorlie et al. 2003), except there was a significant increase in gene numbers likely due to the increased number of genes present on the current microarrays, and another significant difference was that the new Intrinsic/UNC list contained a large proliferation signature (Figure 12F) (Perou et al. 1999; Chung et al. 2002; Whitfield et al. 2002). The inclusion of proliferation genes in the Intrinsic/UNC gene set, but not in the previous Intrinsic/Stanford lists, is likely due to the fact that the Intrinsic/Stanford lists were based upon before and after chemotherapy paired samples of the same tumor, while the Intrinsic/UNC list was based upon identically treated paired samples. This finding suggests that tumor cell proliferation rates did vary before and after chemotherapy, and that proliferation is a reproducible feature of a tumor's expression profile. Thus, the new Intrinsic/UNC list likely encompasses most features of the previous lists, adds new genes to each subtype's defining gene set and adds a biological and clinically relevant feature that is the proliferation signature. 174. Combined test set analysis. Another difference between the intrinsic subtypes found in the 105 sample training data set versus those presented in Sorlie et al. 2001 and 2003 (Sorlie et al. 2001; Sorlie et al. 2003), was that the training set did not have a clear Luminal B (LumB) group as determined by hierarchical clustering analysis. The lack of a LumB group in the training set cluster analysis could be due to few LumB tumors being present in this data set, an artifact of the clustering analysis, or the lack of LumB defining genes in the Intrinsic/UNC gene list. To address this question, a "combined test set" of 315 breast samples was made (311 tumors and 4 normal breast samples) that was a single data set created by combining together the data from Sorlie et al. 2001 and 2003 (cDNA microarrays), van't Veer et al. 2002 (custom Agilent oligo microarrays) and Sotiriou et al. 2003 (cDNA microarrays).
175. A single data table of these three sets was created by first identifying the common genes present across all four microarray data sets (2800 genes). Next, Distance Weighted Discrimination (DWD) was used to combine these three data sets together (Benito et al. 2004); DWD is a multivariate analysis tool that is able to identify systematic biases present in separate data sets and then make a global adjustment to compensate for these biases. Finally, it was determined that 306 of the 1300 unique Intrinsic/UNC genes were present in the combined test set. Figure 14 shows the 315 sample combined test set and the 306 Intrinsic/UNC genes in a two-way hierarchical cluster analysis (see Supplementary Figure 12 for the complete cluster diagram). As expected, this analysis identified the same expression patterns seen in Figure 12 and more. For example, there was a Luminal/ER+ cluster containing ER, GATA3 and GAT A3 -regulated genes (Figure 14C), aHER2+ cluster (Figure 15D), a Basal-like cluster (Figure 14F) and a prominent proliferation signature (Figure 14). The sample-associated dendrogram (Figure 14B) showed the major subtypes seen in Sorlie et al. 2003 including a LumB group, and a potential new tumor group (Luminal T) characterized by the high expression of Interferon (IFN)-regulated genes (Figure 14E). The IFN-regulated cluster contained STATl, which is likely the transcription factor that regulates expression of these IFN-regulated genes (Bromberg et al. 1996; Matikainen et al. 1999). The IFN cluster was one of the first expression patterns to be identified in breast tumors (Perou et al. 1999), and since has been linked to positive lymph node metastasis status and a poor prognosis (Huang et al. 2003; Chung et al. 2004). The effectiveness of the DWD normalization is evident upon close examination of the sample associated dendrogram, which shows that every subtype is populated by samples from each data set (i.e. significant inter-data set mixing). ff6'. Evenithδugh there was limited overlap between the new Mrinsic/UNC list and the Intrinsic/Stanford list of Sorlie et al. 2003 (108 genes in common), there was high agreement in sample classification. For example, it was found 85% concordance in subtype assignments for the 416 tumor data set (combined samples from training and combined test set) that were analyzed independently using the Intrinsic/Stanford and Intrinsic/UNC lists, and both lists showed significance in univariate survival analyses (data not shown). This analysis suggests that, even though the exact constituent genes may vary, the different lists are tracking the same phenotypes and the same "portraits" are seen. However, since the Intrinsic/UNC list contained many more genes and a biologically relevant pattern of expression not seen in the Intrinsic/Stanford lists (i.e. proliferation signature), therefore, it can be more biologically representative of breast tumors. The Intrinsic/UNC list can also be more valuable because it provides a larger number of genes for performing across data set analyses and thus, classifications made across different platforms are less susceptible to artifactual groupings as a result of gene attrition.
177. Multivariate analyses. In the training set and combined test set, the standard clinical parameters of ER status, node status, grade, and tumor size were all significant predictors of Relapse-Free Survival (RFS, where an event is either a recurrence or death) using univariate Kaplan-Meier analysis (Figure 15 for combined test set analysis). In addition, the Intrinsic/UNC gene set identified tumor groups/subtypes that were predictive of RFS on both the training (Figure 16A) and combined test set (Figure 16B). As before, the Luminal group had the best outcome and the HER2+/ER- and Basal-like groups had the worst. The Intrinsic/UNC gene list was also predictive of Overall Survival (OS) on the training and combined test set. As previously seen, patients of the LumB classification showed worse outcomes that LumA, despite being clinically ER+ tumors (Figure 16B). Finally, the new class of Luml showed similar outcomes to LumB, and both showed elevated proliferation rates when compared to LumA tumors (Figure 14G).
178. When the five standard clinical parameters were tested on the 315 sample combined test set using a proportional hazards regression model and RFS, OS or Disease- Specific Survival (DSS) as endpoints, tumor size, grade and ER status were the significant predictors with node status being close to significant (p = 0.06-0.07); however, node status was still prognostic in a univariate analysis (Figure 15B). The next objective was to test for differences in survival among the intrinsic subtypes on the combined test set after adjusting for the clinical covariates of age, ER, node status, grade and tumor size. The approach used was a proportional hazards regression model for RFS (or time to distant metastasis for the van't Veer et al. samples), OS and DSS (which was limited to the Sorlie et al. and Sotiriou et al. data sets). P-values of 0.05 (RFS), 0.009 (OS) and 0.04 (DSS) were obtained when the intrinsic subtypes were tested in a model that included the clinical covariates, which showed that the classifications have significantly different hazard functions, and thus, different survival curves after taking into account (or adjusting for) the effects of age, node status, size, grade, and ER status (Table 11, example for RFS). In this analysis, the Basal-like, LumB and HER2+/ER- subtypes were significantly different from the LumA group (the reference group), while Luml was not. Similar findings were also obtained for the other endpoints except for the LumB subtype, which was not significantly different from LumA in OS (p = 0.36) or DSS (p=0.08).
179. Single Sample Predictions using three additional test sets. A major limitation of using hierarchical clustering as a classifications tool, is its' dependence upon the sample/gene set used for the analysis (Simon et al. 2003). That is, new samples cannot be analyzed prospectively by simply adding them to an existing dataset because it may alter the initial classification of a few previous samples. If an assay is going to be used in the clinical setting, it must be robust and unchanging. To address this concern, a Single Sample Predictor (SSP) was developed using the "combined test set" and its 306 Mrinsic/UNC genes (See Figure 11); the SSP is based upon "Subtype Mean Centroids" and a nearest centroid predictor (Hastie et al. 2001) (see Methods). For the SSP, an intrinsic subtype average profile (centroid) was created for each subtype using the combined test set presented in Figure 14, and then a new sample is individually compared to each centroid and assigned to the subtype/centroid that it is the most similar to using Spearman correlation. Using this method, an intrinsic subtype can be assigned to any sample, from any data set, one at a time.
180. Using the combined test set, five centroids representing the LumA, LumB, Basal- like, HER2+/ER- and Normal Breast-like groups were created). The SSP was tested on three "additional test sets", the first of which was the Ma et al. data set of ER+ patients that were homogenously treated with tamoxifen (Ma et al. 2004). Using the 60 whole tissue samples of Ma et al., the SSP called 2 Basal-like, 2 HER2+/ER-, 12 Normal Breast-like, 34 LumA, and 9 LumB. Since this patient set had RFS data, the SSP classifications were tested in terms of outcomes (the 2 Basal-like and 2 HER2+/ER- samples by SSP analysis were excluded). The SSP assignments were a significant predictor for this group of adjuvant tamoxifen treated patients (p=0.04, Figure 16C). 181. Next, the SSP was applied onto a 96 sample test set of local only (surgery) treated patients from Chang et al. (Chang et al. 2005), which showed highly significant results (Figure 16D, p=0.0006). The final additional test set analyzed was a second adjuvant tamoxifen-treated patient set created by combining similarly treated patients from Chang et al. 2005 plus 16 patients from UNC (which were not included within the 105 patient training data set); for the 45 patient tamoxifen treated data set #2, the SSP called 3 Normal-like, 2 Basal-like and 2 HER2+/ER-, and these samples were excluded from the survival analyses. Again, the SSP- based assignments were a statistically significant predictor of outcomes (Figure 16E for tamoxifen-treated set #2, p=0.02). Finally, if the SSP was applied back onto the original training data set of 105 samples, it was noted that 17 tumors were called LumB (Figure 12) and that the survival analysis showed that these tumors did show a poor outcome (Figure 16F, p=0.02). Thus, the SSP that was based upon hundreds of samples, was able to define clinically relevant distinctions that the hierarchical clustering analysis of 105 samples missed, which further demonstrates the usefulness and objectivity of the SSP. c) DISCUSSION
182. This study identified a number of new biologically relevant "intrinsic" features of breast tumors and methods that are important for the microarray community. These new biological features include the 1) demonstration that proliferation is a stable and intrinsic feature of breast tumors, 2) identification of the high expression of many Kallikrein genes in Basal-like tumors, and 3) demonstration that there are multiple types of "HER2-positive" tumors; the HER2-positive tumors falling into the "HER2+/ER-" intrinsic subtype were also shown to associate with the expression of the Androgen Receptor, while those not falling into this group were present in the LumB or Luml subtypes and usually showed better outcomes . relative to the HER2+/ER- tumors. Recent studies in prostate cancer have shown that HER2 signaling enhances AR signaling under low androgen levels (Mellinghoff et al. 2004). When this finding is coupled to the observation that some HER2+/ER- tumors showed nuclear AR expression (Figure 13B), this suggests that active AR signaling maybe occurring and that anti- androgen therapy can be helpful in these HER2+ (i.e. amplified) and AR+ patients.
183. Microarray studies are often criticized for a lack of reproducibility and limited validation due to small sample sizes (Simon et al. 2003; Ioannidis 2005). By using DWD, multiple microarray data sets have been comboned together to create a single and large combined test set, and it has been shown that the same "intrinsic" patterns can be identified in different data sets in a coordinated analysis, even though entirely different patient populations ^ were'investϊgated'όh different microarray platforms. The analysis of the 315 sample combined test set showed that the "intrinsic" subtypes based upon the Mrinsic/UNC list, were independent prognostic variables, and thus, were providing new clinical information.
184. To be of routine clinical use, a gene expression-based test must be based upon an unchanging assay that is capable of making a prediction on a single sample. Therefore, a Single Sample Predictor/SSP was created that was able to classify samples from three additional test sets of similarly treated patients. In particular, the new Intrinsic/UNC list and the SSP, recapitulated the finding that the intrinsic subtypes are truly prognostic on a test set of local only treated patients (Figure 16D), and it was shown on two additional test sets that LumB patient fair worse than LumA patient in the presence of tamoxifen (Figure 16C and 16E). It should be noted that the distinction of LumA versus LumB closely mirrors the "Recurrence Score" predictor of Paik et al. (Paik et al. 2004), where outcome predictions for tamoxifen- treated ER+ tumors were stratified based mostly on the expression of genes in the HER2- amplicon (HER2 and GRB7), genes of proliferation (STKl 5 and MYBL2), and genes associated with positive ER status (ESRl and BCL2). In essence, high expression of HER2- amplicon and/or proliferation genes gives a high Recurrence Score (and correlates with LumB because most HER2+ and ER+ tumors fall into this subtype), while low expression of these genes and high expression of ER status genes gives a low Recurrence Score (and correlates with LumA).
185. This data shows that the breast tumor intrinsic subtypes identified using the hitrinsic/UNC gene list can be generalized to many different patient sets, both treated and untreated. The intrinsic portraits of breast tumors are recognizable patterns of expression that are of biological and clinical value, and the SSP-based classification tool represents an unchanging predictor to be used for individualized medicine.
3. Example 3: Agreement in Breast Cancer Classification between Microarray and qRT-PCR from Fresh-Frozen and Formalin-Fixed Paraffin- Embedded Tissues
186. Microarray analyses of breast cancers have identified different biological groups that are important for prognosis and treatment. In order to transition these classifications into the clinical laboratory, a real-time quantitative (q)RT-PCR assay has been developed for profiling breast tumors from formalin-fixed paraffin-embedded (FFPE) tissues and evaluate its performance relative to fresh-frozen (FF) RNA samples. 187. Micro array'data from 124 breast samples were used as a training set for classifying tumors into four different previously defined molecular subtypes of Luminal, HER2+/ER-, Basal-like, and Normal-like. Sample class predictors were developed from hierarchical clustering of microarray data using two different centroid-based algorithms: Prediction Analysis of Microarray and a Single Sample Predictor. The training set data was applied to predicting sample class on an independent test set of 35 breast tumors procured as both fresh-frozen and formalin-fixed, paraffin embedded tissues (70 samples). Classification of the test set samples was determined from microarray data using a large 1300 gene set, and using a minimized version of this gene list (40 genes). The minimized gene set was also used in a real-time qRT-PCR assay to predict sample subtype from the fresh-frozen and formalin- fixed, paraffin embedded tissues. Agreement between primer set performance on fresh-frozen and formalin-fixed, paraffin embedded tissues was evaluated using diagonal bias, diagonal correlation, diagonal standard deviation, concordance correlation coefficient, and subtype assignment.
188. The centroid-based algorithms (Prediction Analysis of Microarray and Single Sample Predictor) had complete agreement in classification from formalin-fixed, paraffin- embedded tissues using qRT-PCR and the minimized 'intrinsic' gene set (40 classifiers). There was 94% (33/35) concordance between the diagnostic algorithms when comparing subtype classification from fresh-frozen tissue using microarray (large and minimized gene set) and qRT-PCR data. By qRT-PCR alone, there was 97% (34/35) concordance between fresh-frozen and formalin-fixed, paraffin embedded tissues using Prediction Analysis of Microarray and 91% (32/35) concordance using Single Sample Predictor. Finally, we used several analytical techniques to assess primer set performance between fresh-frozen and formalin-fixed, paraffin- embedded tissues and found that the ratio of the diagonal standard deviation to the dynamic range was the best method for assessing agreement on a gene-by-gene basis.
189. Determining agreement in classification between platforms and procurement methods requires a variety of methods. It has been shown that centroid-based algorithms are robust classifiers for breast cancer subtype assignment across platforms (microarray and qRT- PCR data) and procurement conditions (fresh-frozen and formalin-fixed, paraffin-embedded tissues). In addition, the standard deviation, dynamic range, and concordance correlation coefficient are important parameters to assess individual primer set performance across procurement methods. The strategy for primer set validation and classification have applications in routine clinical practice for stratifying breast cancers and other tumor types. l"9'0'. Expression-based classifications are important for determining risk of relapse and making treatment decisions in breast cancer (Fan et a. N Engl J Med 2006, 355:560-569; Paik et al. N Engl J Med 2004, 351:2817-2826; Perou et al. Nature 2000, 406:747-752; van 't Veer et al. Nature 2002, 415:530-536). Classifications are often developed using microarray data and then further validated on the same or different platforms using minimized gene sets. For instance, van't Veer and van de Vijer used microarray data in training and test sets to validate a 70-gene signature that predicts relapse in early stage ER-positive and ER-negative tumors (van 't Veer et al. Nature 2002, 415:530-536; van de Vijver et al. N Engl J Med 2002, 347:1999- 2009). In addition, Paik et al developed a 16-gene classifier that predicts relapse in ER-positive tumors using qRT-PCR on formalin-fixed, paraffin embedded (FFPE) tissues. Furthermore, Perou and Sorlie showed that hierarchical clustering of microarray data separates breast tumors into different 'biological' subtypes (Luminal, HER2+/ER-, Basal-like, and Normal-like) and that these subtypes are prognostic (Sorlie et al. Proc Natl Acad Sci U S A 2001, 98:10869- 10874). The biological classification has been validated on multiple patient cohorts using cross-platform microarray analyses and qRT-PCR (Hu et al. BMC Genomics 2006, 7:96; Perreard et al. Breast Cancer Res 2006, 8:R23; Sorlie et al. Proc Natl Acad Sci U S A 2003; 100:8418-8423).
191. Although there are few genes in common between those used to determine the biological subtypes and those used in other classifications for breast cancer prognosis, the different tests identify similar properties that predict tumor behavior (Fan et al. N Engl J Med 2006, 355:560-569). A major difference between the classification for biological subtypes and other classifications for breast cancer is that it is based on hierarchical clustering. The unsupervised nature of hierarchical clustering is effective for discovery (Eisen et al. Proc Natl Acad Sci U S A 1998, 95:14863-14868), but it is not suitable for predicting a new sample's class since dendrogram associations can change when new data is introduced. However, it is possible to classify samples within the framework of hierarchical clustering using centroid- based methods (Tibshirani et al. Proc Natl Acad Sci U S A 2002, 99:6567-6572; Bair et al. PLoS Biol 2004, 2:E108; Bullinger et al. N Engl J Med 2004, 350:1605-1616). For instance, Tibshirani et al has shown that the nearest shrunken centroid method, used in Prediction Analysis of Microarray (PAM), can classify samples as accurately as statistical approaches like artificial neural networks. In addition, Hu et al employed another simple centroid method called Single Sample Predictor (SSP) to classify subtypes of breast cancer (Hu et al. 2006). "a) ""'Materials and Methods
(1) Tissue Procurement and Processing
192. All tissues and data used in this study were collected and handled in compliance with federal and institutional guidelines. Breast samples received in pathology were flash frozen in liquid nitrogen and stored at -8O0C. Samples were procured at the University of North Carolina at Chapel Hill, Thomas Jefferson University, University of Chicago, and University of Utah. The 159 breast samples analyzed included a 124-sample microarray training set and a 35 -sample test set profiled by microarray and real-time qRT-PCR (FF and FFPE). Total RNA from FF samples was isolated using the RNeasy Midi Kit (Qiagen, Valencia, CA) and treated on-column with DNase I to eliminate contaminating DNA. The RNA was stored at -8O0C until used for cDNA synthesis.
193. Each FF sample in the test set was compared to the clinical FFPE tissue block. An H&E slide was used to confirm the presence of >50% tumor and 20 micron cuts were prepared using a microtome. Tissue blocks were 1-5 years in age (i.e. early age FFPE). The FFPE cut was de-paraffinized in Hemo-De (Scientific Safety Solvents) and washed with 100% ethanol. Total RNA was isolated using the High Pure RNA Paraffin Kit (Roche Molecular Biochemicals, Mannheim, Germany). Manufacturer's instructions were followed for RNA extraction except that the reagents were increased 2-fold for the first proteinase K digestion. Samples were treated with TURBO DNA-free (Ambion, #1906) and stored at -8O0C until cDNA synthesis.
(2) First-Strand cDNA synthesis
194. cDNA synthesis for each sample was performed in 40μl total volume reaction containing 600ng total RNA. Total RNA was first mixed with 2μl gene specific cocktail containing 55 primers (each anti-sense primer at lpmol/μl) and 2μl 10 niM dNTP Mix (1OmM each dATP, dGTP, dCTP, dTTP at ρH7). Reagents were heated at 650C for 5 minutes in a PTC-100 Thermal Cycler (MJ Research, Inc.) and briefly centrifuged. The following reagents were added to each tube: 8μl 5X First-Strand Buffer, 2μl 0.1M DTT, 2μl RNase Out (Invitrogen), and 2μl Superscript DI polymerase (200units/μl). The reaction was thoroughly mixed by pipetting and incubated at 550C for 45 minutes followed by 15 minutes at 7O0C for enzyme inactivation. Following cDNA synthesis, samples were purified with the QIAquick PCR Purification Kit (Qiagen, Valencia, CA). Samples were adjusted to a final concentration of 1.25ng/μl cDNA with TE (1OmM Tris-HCl, pH 8.0, 0.1 mM EDTA). (3) Primer Design and Optimization
195. Primers were designed using Roche LightCycler Probe Design Software 2.0. Reference gene sequences were obtained through NCBI LocusLink and optimal primer sites were found with the aid of Evidence Viewer (http ://www.ncbi.nlm.nih. gov). Primers sets were selected to avoid known insertions/deletions and mismatches while including all isoforms possible. Amplicons were limited to 60-100bp in length due to the degraded condition of the FFPE mRNA. When possible, RNA specific amplicons were localized between exons spanning large introns (>1 kb). Finally, NCBI BLAST was used to verify gene target specificity of each primer set. Primer sequences are presented in Table 1. Primers were synthesized by Operon, Inc. (Huntsville, AL), re-suspended in TE to a final concentration of 6OuM, and stored at -8O0C. Each new FFPE primer set was assessed for performance through qRT-PCR runs with three serial 10-fold dilutions of reference cDNA in duplicate and two no template control reactions. Primers were verified for use when they fulfilled the following criteria: 1) target Cp < 30 in 10 ng reference cDNA; 2) PCR efficiency >1.75; 3) no primer- dimers in presence of template as determined through post amplification melting curve analysis; and 4) no primer-dimers in negative template control before cycle 40.
(4) Real-Time Quantitative (q)RT-PCR
196. PCR amplification was carried out on the Roche LightCycler 2.0. Each reaction contained 2μl cDNA (2.5ng) and 18μl of PCR master mix with the following final concentration of reagents: 1 U Platinum Taq, 5OmM Tris-HCl (pH 9.1), 1.6mM (NH4)2SO4, 0.4mg/μl BSA, 4mM MgCl2, 0.2mM dATP, 0.2mM dCTP, 0.2mM dGTP, 0.6mM dUTP, 1/40000 dilution of SYBR Green I dye (Molecular Probes, Eugene, OR, USA), and 0.4μM of both forward and reverse primers for the selected target. The PCR was done with an initial denaturation step at 940C for 90s and then 50 cycles of denaturation (940C, 3s), annealing (580C, 6s), and extension (720C, 6s). Fluorescence acquisition (530nm) was taken once each cycle at the end of the extension phase. After PCR, a post-amplification melting curve program was initiated by heating to 940C for 15 s, cooling to 580C for 15 seconds, and slowly increasing the temperature (0.1°C/s) to 950C while continuously measuring fluorescence.
197. Each PCR run contained a no template control, a calibrator reference in triplicate, and each sample in duplicate. The calibrator reference sample was comprised of 3 breast cancer cell lines (MCF7, SKBR3, and ME16C2) and Stratagene Universal Human Reference RNA (Stratagene, La Jolla, CA, USA) represented in equal parts. The crossing point (Cp) for each reaction was automatically calculated by the Roche LightCycler Software 4.0. Relative qϋMϊffl'cation was αoϊϊe by importing an external efficiency curve (Eff=1.89) and setting the calibrator at IOng for each gene. In order to correct for differences in sample quality and cDNA input, copy numbers were adjusted to the arithmetic mean of 5 'housekeeper' genes (ACTB, PSMC4, PUMl, MRPL19, SF3A1). Values from replicate samples were averaged and data was Iog2 transformed.
(5) Microarray
198. AU samples were analyzed by DNA microarray (Agilent Human Al, Agilent Human A2, and Agilent custom oligonucleotide microarrays). Labeling and hybridization of RNA for microarray analysis were performed using the Agilent low RNA input linear amplification kit (http://www.chem.agilent.com/Scripts/PDS. asp?lPage:=10003) as described in Hu et al (Hu et al. Biotechniques2005, 38:121-124). Each sample was assayed versus a common reference that was a mixture of Stratagene's Human Universal Reference total RNA (Stratagene, La Jolla, CA, USA) enriched with equal amounts of RNA from the MCF7 and ME16C cell lines. Microarray hybridizations were carried out on Agilent Human oligonucleotide microarrays using 2 μg Cy3-labeled 'reference' sample and 2 μg Cy5-labeled 'experimental' sample.
199. All microarrays were scanned using an Axon Scanner 4000B (Axon Instruments Inc, Foster City, CA, USA). The image files were analyzed with GenePix Pro 4.1 (Axon Instruments) and were uploaded into the UNC Microarray Database at the University of North Carolina at Chapel Hill (https://genome.unc.edu/), where a Lowess normalization procedure was performed to adjust the Cy3 and Cy5 channels (Yang et al. Nucleic Acids Res 2002' 30:el5).
(6) Clinical Lnmunohistochemistry and PCR
200. Samples were scored for protein expression at the time of diagnosis using standard operating procedures established at each institution. Greater than 10% positive staining nuclei was considered positive for the ER and PR. Staining and scoring criteria for HER2 were carried out according to the Dako HercepTest™ (Dako, Carpinteria, CA, USA). Quantitative PCR, used to determine DNA copy number of the ERBB2 gene, was done using a clinical assay from ARUP Laboratories Inc (cat# 00049390, Salt Lake City, UT, USA).
(7) Selecting Genes for Real-Time qRT-PCR
201. The real-time qRT-PCR assay consisted of 5 housekeeper genes (Szabo et al. Genome Biol 2004, 5:R59), 5 proliferation genes for risk stratification of the Luminal (ER- positive) tumors, and 40 'intrinsic' genes important for distinguishing biological subtypes of breast cancer. The minimal 40 'intrinsic' classifiers were statistically selected from a larger 1300 'intrinsic' gene set previously reported in Hu et al (2006). The larger gene set was minimized as described in Perreard et al (2006). Briefly, a semi-supervised classification method was used in which samples are hierarchical clustered and assigned subtypes based on the sample-associated dendrogram. Samples were designated as Luminal, HER2+/ER— , Basal- like, or Normal-like. The best class distinguishers were identified according to the ratio of between-group to within-group sums of squares. A 10-fold cross-validation was performed using a nearest centroid classifier and testing overlapping gene sets of varying sizes. The smallest gene set which provided the highest class prediction accuracy when compared to the classifications made by the complete microarray-based intrinsic gene set was selected.
(8) Assessing qRT-PCR Agreement between FF and FFPE Tissues
202. Thirty-five matched FF and FFPE samples (70 samples total) were analyzed by qRT-PCR using the same primer sets. Agreement in the quantitative data was determined using diagonal bias (m), diagonal spread (d), diagonal standard deviation (dsd), diagonal correlation (rd), and concordance correlation coefficient (CCC).
203. In diagonal bias, a best fitting line parallel to the diagonal (slope equals 1) is made from a plot of the qRT-PCR data (FF versus FFPE). Numerically, if (x,., y{) , i = l ,...., n denote the measurement pairs then the best fitting line parallel to the diagonal is given by the expression:
Figure imgf000059_0002
where x and y denote the sample means of the x and y measurements, respectively.
Then diagonal bias is calculated as:
Figure imgf000059_0003
The diagonal standard deviation was calculated as follows:
Figure imgf000059_0001
Let d represent:
Figure imgf000060_0001
Diagonal correlation was used to determine the spread of points around the diagonal line:
Figure imgf000060_0002
204. This method does not provide information about the extent of deviation but allows measurements with different units to be compared. Further, if we let p denote the correlation coefficient and Oχ and Oγ the respective standard deviations, then
Figure imgf000060_0003
205. That is, the diagonal correlation penalizes the correction coefficient if there is a scale shift (σx ≠ σγ). The combined effect of the bias and scale shift was measured using the concordance correlation coefficient (CCC) proposed by Lin et al (Lin et al. Biometrics 1989, 45:255-268):
Figure imgf000060_0004
(9) Assessing Agreement between Microarray and qRT-PCR for
Classification.
206. A breast cancer subtype predictor was developed in PAM (http://www- stat. Stanford. edu/~tibs/P AMΛ and SSP using 124 breast samples and the 'intrinsic' gene set identified in Hu et al (2006). The training set contained representative samples of Luminal (64 samples), HER2+/ER- (23 samples), Basal-like (28 samples), and Normal-like (9 samples) subtypes. Classification of an independent test set (35 matched FF and FFPE samples) was done using a large (1300 genes) and minimized (40 genes) version of the 'intrinsic' set. Subtypes were assigned based on Spearman correlation to the centroid. The qRT-PCR data from the test set was merged with the microarray data of the training set prior to classification using distance weighted discrimination (Benito et al. Bioinformatics 2004, 20:105-1.14). The gold standard for classification of the training and test samples was based on FF tissue RNA and using the classifications obtained when performing hierarchical clustering analysis using the 1300 gene intrinsic gene set from microarray data, b) Results
(1) Assessment of qRT-PCR Primer Set Performance by Comparing Agreement between FF and FFPE Tissues.
207. The data set of 35 matched FF and FFPE tissues (70 samples) was evaluated for 50 genes using the same PCR conditions. Agreement between FF and FFPE tissues was assessed for diagonal bias (m), diagonal correlation (rd) diagonal standard deviation (dsd), and concordance correlation coefficient (ccc). Figure 18 shows an agreement plot between FF and FFPE for the estrogen receptor gene (ESRl) after normalization to the 5 housekeepers. The large dynamic range of ESRl expression provides clear separation of the tumors from both FF and FFPE (Figure 20). ESRl alone measured from FF tissue has very high sensitivity and specificity using ER status by IHC as the gold standard (Perreard 2006).
208. For each gene, the agreement between FF and FFPE was analyzed using the raw data, housekeeper normalized data, and DWD adjusted normalized data. Scatter plots are provided in Figures 20-23 and values are presented in Table 14. The line graphs in 19 show the effects at each step of data processing. The raw (pre-normalized) data shows a negative bias for all genes likely due to lower RNA quality in the FFPE tissue (Fig 19A). Much of the bias was corrected by normalization to the 'housekeeper' genes and using DWD adjustment. As expected, DWD had a significant effect on bias (m) but did not effect other measurements of agreement (Fig 19B-D).
209. The median biases for the un-normalized, housekeeper normalized, and DWD adjusted normalized data were -1.5 (-3.1 to -0.033), 0.58 (-1.1 to 2) and 0.24 (-0.3 to 1.3), respectively. Normalization to the housekeeper genes had a relatively modest effect on the diagonal standard deviation with a change in the median from 1.1 (0.76-2) to 0.81 (0.38-1.8). While most genes had a similar standard deviation (e.g. ESRl) after applying the housekeepers, other genes such as C10orf7 and COX6C had nearly a 3-fold reduction in standard deviation after normalization. 210. In general, genes with the highest diagonal correlation between FF and FFPE also had the largest dynamic range in expression (e.g., ESRl, TFF3, COX6C, and FBPl). Housekeeper genes and other genes with low variability in expression (IGBPl) had the lowest diagonal correlation since they form more of a cloud than a line around the diagonal. The housekeeper genes all had high agreement in terms of having low variability in expression across samples in the FF and FFPE tissues.
211. The concordance correlation coefficient (CCC) considers both bias and scale shift when determining agreement. The median concordance correlation coefficient between FF and FFPE for the raw data of the 45 genes (housekeepers excluded) was 0.28. Normalization to housekeepers raised the CCC median to 0.48, and adjusting with DWD brought the median to 0.61. Only 27% of the genes had a CCC value greater than 0.5, whereas 47% of the genes were above that value in the normalized data, and 76% were above that when using DWD adjusted normalized data. A comparison of the CCC value to the ratio of the diagonal standard deviation over the dynamic range identified many of the same primer sets as good (or poor) performers from the FFPE derived samples.
(2) Breast Cancer Subtype Classification of Test Set using PAM and SSP.
212. Hierarchical clustering of the 124 sample training set using the "intrinsic" gene set identified in Hu et al shows 4 distinct classes representing Luminal, HER2+/ER-, Basal- like, and Normal-like (Figure 24). Centroid classifiers were developed from the microarray expression data using PAM and SSP (Hu et al. 2006, Tibshirani et al. 2002). Class predictions were made on the test set using microarray (large and minimized 'intrinsic' sets) and qRT-PCR data (15). Each individual microarray (large and minimized) and PCR datasets were DWD merged with the training set prior to subtype class prediction.
213. Agreement in classification between large and minimized microarray gene sets. Thirty-three out of 35 (94%) samples classified the same between PAM and SSP when using the large 'intrinsic' microarray dataset for classification. In both discrepant cases, IHC data agreed with the PAM classification. There was the same agreement (94%) when performing the analysis with the minimized version of the microarray data. Interestingly, there was one sample that was called HER2+/ER- by both PAM and SSP when using the large microarray dataset, but called Basal-like by both methods when using the minimized microarray dataset. Additional analysis of this sample by quantitative PCR showed no DNA amplification of HER2/ERBB2 amplicon. LI2'?4"." ABj-Bemehfm classification between FF and FFPE. By qRT-PCR, there was 97% (34/35) concordance between FF and FFPE using PAM, and 91% (32/35) concordance using SSP. There was 94% (33/35) concordance between the diagnostic algorithms from FF tissue and complete agreement in classification from FFPE tissue. Since the FFPE samples were obtained from the clinical block, it is likely that there was a higher tumor percentage in those samples than in the matched FF sample, which could affect the agreement. Indeed, 2 out of the 3 discrepancies in classification made by SSP were when the FF tissue sample was called Normal-like (microarray and PCR) and the FFPE sample was called Luminal (PCR). These samples were ER-positive by IHC and likely Luminal. The only discrepancy in PAM was in a sample classified as Normal-like from FF tissue and Luminal from FFPE.
215. Overall concordance across methods. Overall, PAM diagnosed 33 out of 35 samples (94%) the same across microarray and qRT-PCR, while SSP diagnosed 30 out of 35 samples (86%) the same across platforms and procurement methods. Discrepancies were of several types including Luminal tumors classified as Normal-like, HER2+/ER- tumors classified as Luminal, and Basal-like tumors classified as HER2+/ER-. c) Discussion
216. The transition of large-scale microarray experiments into a clinical test requires identifying a minimum set of genes for classification, translating the assay from microarray to qRT-PCR for routine diagnostics, and validating the assay using both FF and FFPE specimen types.
217. A previous qRT-PCR assay for identifying biological subtypes was based on an intrinsic gene set derived from first generation microarrays that contained 8,100 genes. In comparison, the current intrinsic set was derived from a different microarray platform (cDNA versus Agilent), contained a larger number of genes (427 vs. 1300), and used pre-treatment samples only (Hu et al. 2006. The overlap in the minimized gene set developed here versus the list in Perreard et al. was 14 out of 40, which is not surprising since there were only 108 genes in common between the larger intrinsic gene sets. It has been shown that the new intrinsic gene set reproducibly identifies the same breast cancer subtypes within independent datasets (i.e. pure training and test sets), and that the biological classification adds significant clinical information when used in a multivariate Cox analysis.
218. It has been shown that the centroid-based method called Single Sample Predictor can use microarray data to classify breast cancers into biological subtypes that predict survival in treated and untreated patients (Hu et al. 2006). Here PAM is directly compared to SSP using "ϊhfe''ϊirge"mϊcroarray daϊaset applied in Hu et al, and also tested a minimized version using microarray and qRT-PCR data. Both methods performed well.
219. This method of classification is considered semi-supervised since data from hierarchical clustering is initially used to develop a centroid or shrunken centroid from a training set and new samples are then classified based on the distance to the centroid. In this way, the training set is not only necessary for initial discovery and validation but the data continues to be used as a reference base for future classification of new samples. Similarly, the Oncotype Dx assay established cut points for risk of relapse from a training set and this classifier rule is applied to new samples to derive a recurrence score.
220. Determining agreement between methods is a complex issue that requires consideration of several factors before reaching a conclusion. Cronin et al used Pearson correlation to show that the genes with the highest correlation in microarray maintained their association with qRT-PCR. They used short amplicons and control 'housekeeper' genes in the qRT-PCR assay to correct biases between FF and FFPE tissues. Although correlation provides information about the linearity and slope (positive or negative correlation) of the data, it does not indicate the amount of bias, scale shift, or data spread. These additional measurements are helpful in determining whether the discrepancies in the data can be compensated for experimentally (e.g., housekeeper genes) or by software algorithms. For example, when the qRT-PCR data from FF and FFPE were compared, it was found that a significant bias could be corrected by normalization to the housekeepers and applying Distance Weighted Discrimination. Distance Weighted Discrimination corrected systematic biases but did not change other measurements of agreement. After correcting for systematic bias, it is then possible to evaluate variation due to noise that cannot be predicted or controlled.
221. It was found that the most useful analyses for assessing PCR primer set performance across FF and FFPE tissues were the concordance correlation coefficient, the diagonal standard deviation, and the dynamic range. Genes with a large dynamic range often had high correlation and were good classifiers across conditions, even with relatively large diagonal standard deviations. Although genes with a small dynamic range can be good classifiers, the measurement may not be as reproducible if there is a large amount of variation. Thus, it was found that the best assessment of a classifier was using a ratio of the diagonal standard deviation to the dynamic range. This allowed genes with smaller dynamic ranges to be considered as good classifiers, if they also had low diagonal standard deviations. The concordance correlation coefficient and the ratio of the diagonal standard deviation to the dynamic raήge selected many of the same genes as having similar performance from the FF and FFPE tissues.
222. Translating an assay from microarray to qRT-PCR provides a second level of gene validation and allows the test to be used on archived FFPE tissue blocks from clinical trials or on samples submitted for routine diagnostics (Paik et al. 2004; Cronin et al. Am J Pathol 2004, 164:35-42). qRT-PCR on formalin-fixed paraffin-embedded tissue can be effectively used for gene expression based diagnostics for translation into the clinical laboratory. The FFPE procured RNA provided accurate subtype classifications in breast cancer, and in some instances provided more tumor specific information than the FF derived samples. This study also developed methodologies that have wider application for developing qRT-PCR assays for subtype classification in a wide variety of cancer types. These gene expression based tests can provide powerful new prognostic clinical tools and aid in more appropriate individualized treatment decisions.
223'. Tables" 1 l'."Kegression model using RFS and the intrinsic classes from the 315 tumor sample Combined Test Set.
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
1876
PCT/US2006/044737
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
P3 minimized P2 minimized
Intrinsic gene list Intrinsic qene list
ASF1A ACADSB
BLVRA B3GNT5 BF
BTG3 COX6C C5ORF18 (=DP1)
C10orf7 ERBB2 CDK2AP1
C16orf45 ESR1 CX3CL1
CaMKIINalpha FOXC1 CYB5
CDH3 FZD7 DSC2 (ESTs)
CHI3L2 GATA3 EGFR
CSDA GRB7 FLJ14525
CTPS GSTP1 FOXA1
FABP7 KIT GARS
FBP1 KRT17 HSD17B4
FLJ10980 S100A11 KIAA0310
GSTM3 SLC39A6 KRT5
HIS1 XBP1 NAT1
ID4 PGR
IGBP1 PLOD1
INPP4B PTP4A2
SEMA3C RABEP1
SLC5A6 RARRES3
TCEAL1 SDC2
TFF3 SLPI
TMSB10 SMA3
TP53BP2 TAP1
VAV3 TRIM29
WWP1
Proliferation genes Proliferation genes
BUB1 BIRC5
MKI67 BUB1
MYBL2 CENPF
STK6 CKS2
T0P2A FAM54A (=DUFD1)
GTPBP4
Housekeeper genes HSPA14 ACTB MKI67
MRPL19 MYBL2
PSMC4 NEK2
PUM1 PCNA
SF3A1 STK6
TOP2A TTK
Housekeeper genes
MRPL19
PSMC4
PUM1
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
G. References
Akilesh S, Shaffer DJ, Roopenian D. "Customized molecular phenotyping by quantitative gene expression and pattern recognition analysis" Genome Res 13:1719-1727 (2003).
Bair, E., and Tibshirani, R. "Semi-supervised methods to predict patient survival from gene expression data" PLoS Biol 2:E108 (2004).
Bloom, H. J.G., and Richardson, W. W. "Histologic grading and prognosis in breast cancer" British Journal of Cancer 9:359-377 (1957).
Benito, M., Parker, J., Du, Q., Wu, J., Xiang, D., Perou, CM., and Marron, J.S. "Adjustment of systematic microarray data biases" Bioinformatics 20:105-114 (2004).
Bhatia P, Taylor WR, Greenberg AH, Wright JA. "Comparison of glyceraldehyde-3-phosphate dehydrogenase and 28S-ribosomal RNA gene expression as RNA loading controls for northern blot analysis of cell lines of varying malignant potential" Anal Biochem 216:223-226 (1994).
Bullinger, L., Dohner, K., Bair, E., Frohling, S., Schlenk, R.F., Tibshirani, R., Dohner, H., and Pollack, J.R. "Use of gene-expression profiling to identify prognostic subclasses in adult acute myeloid leukemia" N Engl J Med 350:1605-1616 (2004).
Buzdar, A., O'Shaughnessy, J.A., Booser, D.J., Pippen, J.E., Jr., Jones, S.E., Munster, P.N., Peterson, P., Melemed, A.S., Winer, E., and Hudis, C. "Phase JJ, randomized, double-blind study of two dose levels of arzoxifene in patients with locally advanced or metastatic breast cancer" J Clin Oncol 21:1007-1014 (2003).
CaIy, M., Genin, P., Ghuzlan, A.A., Elie, C, Freneaux, P., Klijanienko, J., Rosty, C, Sigal- Zafrani, B., Vincent-Salomon, A., Douggaz, A., et al. "Analysis of correlation between mitotic index, MIBl score and S-phase fraction as proliferation markers in invasive breast carcinoma. Methodological aspects and prognostic value in a series of 257 cases" Anticancer Res 24:3283- 3288 (2004).
Chia, S.K., Speers, C.H., Bryce, C.J., Hayes, M.M., and Olivotto, LA. "Ten-year outcomes in a population-based cohort of node-negative, lymphatic, and vascular invasion-negative early breast cancers without adjuvant systemic therapies" J Clin Oncol 22:1630-1637 (2004).
Clark, G.M., Allred, D.C., Hilsenbeck, S.G., Chamness, G.C., Osborne, C.K., Jones, D., and Lee, W.H. "Mitosin (a new proliferation marker) correlates with clinical outcome in node- negative breast cancer" Cancer Res 57:5505-5508 (1997).
Cronin, M., Pho, M., Dutta, D., Stephans, J.C., Shak, S., Kiefer, M.C., Esteban, J.M., and Baker, J.B. "Measurement of gene expression in archival paraffin-embedded tissues: development and performance of a 92-gene reverse transcriptase-polymerase chain reaction assay" Am J Pathol 164:35-42 (2004).
Dalton, L. W., Page, D.L., and Dupont, W.D. "Histologic grading of breast carcinoma. A reproducibility study" Cancer 73:2765-2770 (1994). Dhanasekarati SM, Barrette TR, Ghosh D, Shah R, Varambally S, Kurachi K, Pienta KJ, Rubin MA, Chinnaiyan AM. "Delineation of prognostic biomarkers in prostate cancer" Nature 412:822-826 (2001).
Diehn, M., Sherlock, G., Binkley, G., Jin, H., Matese, J.C., Hernandez-Boussard, T., Rees, C.A., Cherry, J.M., Botstein, D., Brown, P.O., et al. "SOURCE: a unified genomic resource of functional annotations, ontologies, and gene expression data" Nucleic Acids Res 31:219-223 (2003).
Dudoit, S., and Fridlyand, J. "A prediction-based resampling method for estimating the number of clusters in a dataset" Genome Biol 3:RESEARCH0036 (2002).
Efron, B., Tibshirani, RJ. "An Introduction to the Bootstrap" Boca Raton, Florida: CRC Press LLC. p 247 pp (1998).
Eggert A, Brodeur GM, Dcegaki N. "Relative quantitative RT-PCR protocol for TrkB expression in neuroblastoma using GAPD as an internal control" Biotechniques 28:681-682, 686, 688-691 (2000).
Eisen, M.B., Spellman, P.T., Brown, P.O., and Botstein, D. "Cluster analysis and display of genome-wide expression patterns" Proc Natl Acad Sci U S A 95:14863-14868 (1998).
Elston, C.W., and Ellis, LO. "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 19:403-410 (1991).
Fisher, E.R., Osborne, C.K., McGuire, W.L., Redmond, C, Knight, W.A., 3rd, Fisher, B., Bannayan, G., Walder, A., Gregory, E.J., Jacobsen, A., et al. "Correlation of primary breast cancer histopathology and estrogen receptor content" Breast Cancer Res Treat 1:37-41 (1981).
Fisher, B., Costantino, J., Redmond, C, Poisson, R., Bowman, D., Couture, J., Dimitrov, N. V., Wolmark, N., Wickerham, D.L., Fisher, E.R., et al. "A randomized clinical trial evaluating tamoxifen in the treatment of patients with node-negative breast cancer who have estrogen- receptor-positive tumors" N Engl J Med 320:479-484 (1989).
Fitzgibbons, P.L., Page, D.L., Weaver, D., Thor, A.D., Allred, D.C., Clark, G.M., Ruby, S.G., O'Malley, F., Simpson, J.F., Connolly, J.L., et al. "Prognostic factors in breast cancer. College of American Pathologists Consensus Statement 1999" Arch Pathol Lab Med 124:966-978 (2000).
Frank SG, Bernard, P. S. "Profiling Breast Cancer using Real-Time Quantitative PCR. In Rapid Cycle Real-Time PCR: Methods and Applications" Edited by S. Meuer W, C, Nakagawara, K. Heidelberg, Germany, Springer pp 95-106 (2003).
Frierson, H.F., Jr., Wolber, R.A., Berean, K.W., Franquemont, D.W., Gaffey, M.J., Boyd, J.C., and Wilbur, D.C. "Merobserver reproducibility of the Nottingham modification of the Bloom and Richardson histologic grading scheme for infiltrating ductal carcinoma" Am J Clin Pathol 103:195-198 (1995).
Genestie, C, Zafrani, B., Asselain, B., Fourquet, A., Rozan, S., Validire, P., Vincent-Salomon, A., and Sastre-Garau, X. "Comparison of the prognostic value of Scarff-Bloom-Richardson and Nottingham histological grades in a series of 825 cases of breast cancer: major importance of the mitotic count as a component of both grading systems" Anticancer Res 18:571-576 (1998).
Greenough, R.B. "Varying degrees of malignancy in cancer of the breast" J Cancer Res 9:452- 463 (1925).
Gruvberger, S., Ringner, M., Chen, Y., Panavally, S., Saal, L.H., Borg, A., Ferno, M., Peterson, C, and Meltzer, P. S. "Estrogen receptor status in breast cancer is associated with remarkably distinct gene expression patterns" Cancer Res 61:5979-5984 (2001).
Henson, D.E., Ries, L., Freedman, L.S., and Carriaga, M. "Relationship among outcome, stage of disease, and histologic grade for 22,616 cases of breast cancer. The basis for a prognostic index" Cancer 68:2142-2149 (1991).
Ishida, S., Huang, E., Zuzan, H., Spang, R., Leone, G., West, M., andNevins, J.R. "Role for E2F in control of both DNA replication and mitotic functions as revealed from DNA microarray analysis" MoI Cell Biol 21:4684-4699 (2001).
Iwahashi, H., Eguchi, Y., Yasuhara, N., Hanafusa, T., Matsuzawa, Y., and Tsujimoto, Y. "Synergistic anti-apoptotic activity between Bcl-2 and SMN implicated in spinal muscular atrophy" Nature 390:413-417 (1997).
Kollias, J., Murphy, C.A., Elston, C.W., Ellis, I.O., Robertson, J.F., and Blarney, R. W. "The prognosis of small primary breast cancers" Eur J Cancer 35:908-912 (1999).
Kristt D, Turner I, Koren R, Ramadan E, Gal R. "Overexpression of cyclin Dl mRNA in colorectal carcinomas and relationship to clinicopathological features: an in situ hybridization analysis" Pathol Oncol Res 6:65-70 (2000).
Laping, NJ., Olson, B. A., and Zhu, Y. "Identification of a novel nuclear guanosine triphosphate- binding protein differentially expressed in renal disease" J Am Soc Nephrol 12:883-890 (2001).
Manders, P., BuIt, P., Sweep, C.G., Tjan-Heijnen, V.C., and Beex, L. V. "The prognostic value of the mitotic activity index in patients with primary breast cancer who were not treated with adjuvant systemic therapy" Breast Cancer Res Treat 77:77-84 (2003).
Makretsov, N.A., Huntsman, D.G., Nielsen, T.O., Yorida, E., Peacock, M., Cheang, M.C., Dunn, S.E., Hayes, M., van de Rijn, M., Bajdik, C, et al. "Hierarchical clustering analysis of tissue microarray immunostaining data identifies prognostically significant groups of breast carcinoma" Clin Cancer Res 10:6143-6151 (2004).
Michels, JJ., Marnay, J., Delozier, T., Denoux, Y., and Chasle, J. "Proliferative activity in primary breast carcinomas is a salient prognostic factor" Cancer 100:455-464 (2004).
Miller CL, Yolken RH. "Methods to optimize the generation of cDNA from postmortem human brain tissue" Brain Res Brain Res Protoc 10:156-167 (2003).
Mischel PS, Nelson SF, Cloughesy TF. "Molecular analysis of glioblastoma: pathway profiling and its implications for patient therapy" Cancer Biol Ther 2:242-247 (2003).
Nielsen, T.O., Hsu, F.D., Jensen, K., Cheang, M., Karaca, G., Hu, Z., Hernandez-Boussard, T., Livasy, C, Cowan, D., Dressier, L., et al. "Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma" Clin Cancer Res 10:5367-5374 (2004).
Paik, S., Shak, S., Tang, G., Kim, C, Baker, J., Cronin, M., Baehner, F.L., Walker, M.G., Watson, D., Park, T., et al. "A multigene assay to predict recurrence of tamoxif en-treated, node- negative breast cancer" N Engl J Med 351:2817-2826 (2004).
Panaro NJ, Yuen PK, Sakazume T, Fortina P, Kricka LJ, Wilding P. "Evaluation of DNA fragment sizing and quantification by the agilent 2100 bioanalyzer" Clin Chem 46:1851-1853 (2000).
Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D. "Molecular portraits of human breast tumours" Nature 406:747-752 (2000).
Perou CM, Brown PO, Botstein D. "Tumor classification using gene expression patterns from DNA microarrays" New Technologies for life sciences: A Trends Guide pp 67-76 (2000).
Perou, CM., Jeffrey, S.S., van de Rijn, M., Rees, C.A., Eisen, M.B., Ross, D.T., Pergamenschikov, A., Williams, C.F., Zhu, S.X., Lee, J.C., et al. "Distinctive gene expression patterns in human mammary epithelial cells and breast cancers" Proc Natl Acad Sci U S A 96:9212-9217 (1999).
Pinheiro JC BD. "Mixed-effects models in S and S-PLUS" New York, Springer (2000).
Pollack, J.R., Sorlie, T., Perou, CM., Rees, CA., Jeffrey, S.S., Lonning, P.E., Tibshirani, R., Botstein, D., Borresen-Dale, A.L., and Brown, P.O. "Microarray analysis reveals a major direct role of DNA copy number alteration in the transcriptional program of human breast tumors" Proc Natl Acad Sci U S A 99:12963-12968 (2002).
Pollack, J.R., Perou, CM., Alizadeh, A.A., Eisen, M.B., Pergamenschikov, A., Williams, C.F., Jeffrey, S. S., Botstein, D., and Brown, P.O. "Genome-wide analysis of DNA copy-number changes using cDNA microarrays" Nature Genetics 23:41-46 (1999).
Rasmussen RP. "Quantification on the LightCycler. In Rapid Cycle Real-Time PCR: Methods and Applications" Edited by Wittwer CT, Meuer, S., Nakagawara, K. Heidelberg, Springer Verlag, pp 21-34 (2001).
Robbins, P., Pinder, S., de Klerk, N., Dawkins, H., Harvey, J., Sterrett, G., Ellis, L, and Elston, C "Histological grading of breast carcinomas: a study of interobserver agreement" Hum Pathol 26:873-879 (1995).
Ross, D.T., Scherf, U., Eisen, M.B., Perou, CM., Rees, C, Spellman, P., Iyer, V., Jeffrey, S.S., Van de Rijn, M., Waltham, M., et al. "Systematic variation in gene expression patterns in human cancer cell lines [see comments]" Nat Genet 24:227-235 (2000). Roux S, Pichaud F, Quinn J, Lalande A, Morieux C, Jullienne A, de Vernejoul MC. "Effects of prostaglandins on human hematopoietic osteoclast precursors" Endocrinology 138:1476-1482 (1997).
SantaLucia J. "A unified view of polymer, dumbbell, and oligonucleotide DNA nearest- neighbor thermodynamics" Proc Natl Acad Sci U S A 95:1460-1465 (1998).
Schena M, Sfialon D, Davis RW, Brown PO. "Quantitative monitoring of gene expression patterns with a complementary DNA microarray" Science 270:467-470 (1995).
Schwarz G. "Estimating the dimension of a model" The Annals of Statistics 6:461-464 (1978).
Singletary, S.E., Allred, C, Ashley, P., Bassett, L. W., Berry, D., Bland, K.I., Borgen, P.I., Clark, G.M., Edge, S.B., Hayes, D.F., et al. "Staging system for breast cancer" revisions for the 6th edition of the AJCC Cancer Staging Manual. Surg Clin North Am 83:803-819 (2003).
Sorlie, T., Tibshirani, R., Parker, J., Hastie, T., Matron, J.S., Nobel, A., Deng, S., Johnsen, H., Pesich, R., Geisler, S., et al. "Repeated observation of breast tumor subtypes in independent gene expression data sets" Proc Natl Acad Sci U S A 100:8418-8423 (2003). Sørlie, T., Perou, CM., Tibshirani, R., Aas, T., Geisler, S., Johnsen, H., Hastie, T., Eisen, M.B., van de Rijn, M., Jeffrey, S. S., et al. "Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications" Proc Natl Acad Sci U S A 98:10869-10874 (2001). .
Sotiriou, C, Neo, S.Y., McShane, L.M., Korn, EX., Long, P.M., Jazaeri, A., Martiat, P., Fox, S.B., Harris, A.L., and Liu, E.T. "Breast cancer classification and prognosis based on gene expression profiles from a population-based study" Proc Natl Acad Sci U S A 100:10393-10398 (2003).
Spanakis E. "Problems related to the interpretation of autoradiographic data on gene expression using common constitutive transcripts as controls" Nucleic Acids Res 21:3809-3819 (1993).
Suzuki T, Higgins PJ, Crawford DR. "Control selection for RNA quantitation" Biotechniques 29:332-337 (2000).
Szabo, A., Perou, CM., Karaca, M., Perreard, L., Quackenbush, J.F., and Bernard, P. S. "Statistical modeling for selecting housekeeper genes" Genome Biol 5:R59 (2004).
Taylor-Papadimitriou, J., Stampfer, M., Bartek, J., Lewis, A., Boshell, M., Lane, E.B., and Leigh, LM. "Keratin expression in human mammary epithelial cells cultured from normal and malignant tissue: relation to in vivo phenotypes and influence of medium" J Cell Sci 94:403-413 (1989).
Troyanskaya, O., Cantor, M., Sherlock, G., Brown, P., Hastie, T., Tibshirani, R., Botstein, D., and Altaian, R.B. "Missing value estimation methods for DNA microarrays" Bioinformatics 17:520-525 (2001).
Tubbs RR, Pettay JD, Roche PC, Staler MH, Jenkins RB, Grogan TM. "Discrepancies in clinical laboratory testing of eligibility for trastuzumab therapy: apparent immunohistochemical false-positives do not get the message" J Clin Oncol 19:2714-2721 (2001). van de Vijver MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, Parrish M, Atsma D, Witteveen A, Glas A, Delahaye L, van der Velde T, Bartelink H, Rodenhuis S, Rutgers ET, Friend SH, Bernards R. "A gene-expression signature as a predictor of survival in breast cancer" N Engl J Med 347:1999-2009 (2002).
van 't Veer, LJ., Dai, H., van de Vijver, M.J., He, Y.D., Hart, A.A., Mao, M., Peterse, H.L., van der Kooy, K., Marton, M.J., Witteveen, A.T., et al. "Gene expression profiling predicts clinical outcome of breast cancer" Nature 415:530-536 (2002).
Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F. "Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes" Genome Biol 3:RESEARCH0034 (2002).
Welsh JB, Zarrinkar PP, Sapinoso LM, Kern SG, Behling CA, Monk BJ, Lockhart DJ, Burger RA, Hampton GM. "Analysis of gene expression profiles in normal and neoplastic ovarian tissue samples identifies candidate molecular markers of epithelial ovarian cancer" Proc Natl Acad Sci U S A 98:1176-1181 (2001).
West, M., Blanchette, C, Dressman, H., Huang, E., Ishida, S., Spang, R., Zuzan, H., Olson, J.A., Jr., Marks, J.R., and Nevins, J.R. "Predicting the clinical status of human breast cancer by using gene expression profiles" Proc Natl Acad Sci U S A 98: 11462-11467 (2001).
Whitfield, MX., Sherlock, G., Saldanha, AJ., Murray, J.I., Ball, C.A., Alexander, K.E., Matese, J.C., Perou, CM., Hurt, M.M., Brown, P.O., et al. "Identification of genes periodically expressed in the human cell cycle and their expression in tumors" MoI Biol Cell 13:1977-2000 (2002).
Wittwer CT, a.K., N. "Real-time PCR. Jh Molecular Microbiology" T. Persing DH, FC, Versalovic, J, Tang, YW, Unger, ER, Relman, DA, and White, TJ, editor. Washington, DC: ASM Press (2004).
Yang, Y.H., Dudoit, S., Luu, P., Lin, D.M., Peng, V., Ngai, J., and Speed, T.P. "Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation" Nucleic Acids Res 30:el5 (2002).
Yu, K., Lee, C.H., Tan, P.H., and Tan, P. "Conservation of breast cancer molecular subtypes and transcriptional patterns of tumor progression across distinct ethnic populations" Clin Cancer Res 10:5508-5517 (2004).

Claims

VL CLAIMSWhat is claimed is:
1. A method of diagnosing cancer, the method comprising comparing expression levels of a combination of genes from Table 21 to test nucleic acids, wherein specific expression patterns of the test nucleic acids indicates a cancerous state.
2. The method of claim 1, wherein the combination of genes includes at least 10 genes from Table 21.
3. The method of claim 1, wherein the combination of genes includes at least 25 genes from Table 21.
4. The method of claim 1, wherein the combination of genes includes at least 50 genes from Table 21.
5. The method of claim 1, wherein the combination of genes includes at least 75 genes from Table 21.
6. A method of quantitating level of expression of a test nucleic acid comprising: a) comparing gene expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes; and b) quantitating level of expression of the test nucleic acid.
7. A method determining prognosis based on expression patterns in a subject diagnosed with cancer comprising: a) comparing expression levels of a combination of genes from Table 21 to test nucleic acids corresponding to the same combination of genes, b) identifying a subtype of cancer of the subject, and c) determining prognosis based on expression patterns in the subject.
8. A method of classifying cancer in a subject, comprising: a) identifying intrinsic genes of the subject to be used to classify the cancer; b) obtaining a sample from the subject; c) amplifying and detecting levels of intrinsic genes in the subject; and d)classifying cancer based upon results of step c.
9. A method of diagnosing cancer in a subject the method comprising: a) amplifying and detecting intrinsic genes; and b) diagnosing cancer based on expression levels of the gene within the subject.
10. A method of deriving a minimal intrinsic gene set for making biological classifications of cancer comprising: a) collecting data from multiple samples from the same individual to identify potential intrinsic classifier genes; b) weighting intrinsic classifier genes of multiple individuals identified using the method of step a relative to each other and forming classification clusters; c) estimating the number of clusters formed in step b) and assigning individual samples to classification clusters; d) identifying genes that optimally distinguish the samples in the assigned groups of step c); e) performing iterative cross-validation with a nearest centroid classifier and overlapping gene sets of various sizes using the genes identified in step d); and f) choosing a gene set which provides the highest class prediction accuracy when compared to the classifications made in step b).
11. The method of claim 10, wherein the cancer is selected from the group consisting of breast cancer, colon cancer, or melanoma.
.
12. The method of any one of claims 1-10, wherein the genes are derived from fresh samples.
13. The method of any one of claims 1-10, wherein the genes are derived from formalin-fixed paraffin embedded (FFPE) samples.
14. The method of claim 10, wherein sample comprises mRNA.
15. The method of claim 10, wherein the sample is amplified by PCR.
16. The method of claim 15, wherein the PCR is real time PCR.
17. The method of claim 11 , wherein the breast cancer is classified into luminal, normal-like, HER2+/ER-, and basal-like.
18. The method of claim 10, wherein the intrinsic gene set is identified using a microarray.
19. The method of claim 10, wherein the intrinsic gene set is modified from a microarray.
20. The method of claim 19, wherein the intrinsic gene set includes at least one housekeeper gene.
21. A method of assigning a sample to an intrinsic subtype, comprising: a) creating an intrinsic subtype average profile (centroid) for each subtype; b) individually comparing a new sample to each centroid; and c) assigning the new sample to the centroid that is most similar to an expression profile of the new sample.
PCT/US2006/044737 2005-11-23 2006-11-17 Methods and compositions involving intrinsic genes WO2007061876A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA002630974A CA2630974A1 (en) 2005-11-23 2006-11-17 Methods and compositions involving intrinsic genes
US12/094,898 US20090299640A1 (en) 2005-11-23 2006-11-17 Methods and Compositions Involving Intrinsic Genes
EP06844413A EP1954708A4 (en) 2005-11-23 2006-11-17 Methods and compositions involving intrinsic genes
US13/959,575 US20140087959A1 (en) 2005-11-23 2013-08-05 Methods And Compositions Involving Intrinsic Genes
US15/043,022 US20170044618A1 (en) 2005-11-23 2016-02-12 Methods and compositions involving intrinsic genes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US73915505P 2005-11-23 2005-11-23
US60/739,155 2005-11-23

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/094,898 A-371-Of-International US20090299640A1 (en) 2005-11-23 2006-11-17 Methods and Compositions Involving Intrinsic Genes
US13/959,575 Continuation US20140087959A1 (en) 2005-11-23 2013-08-05 Methods And Compositions Involving Intrinsic Genes

Publications (3)

Publication Number Publication Date
WO2007061876A2 true WO2007061876A2 (en) 2007-05-31
WO2007061876A3 WO2007061876A3 (en) 2007-09-20
WO2007061876A8 WO2007061876A8 (en) 2007-10-25

Family

ID=38067789

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/044737 WO2007061876A2 (en) 2005-11-23 2006-11-17 Methods and compositions involving intrinsic genes

Country Status (4)

Country Link
US (3) US20090299640A1 (en)
EP (1) EP1954708A4 (en)
CA (1) CA2630974A1 (en)
WO (1) WO2007061876A2 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2215254A1 (en) * 2007-10-05 2010-08-11 Pacific Edge Biotechnology Ltd Proliferation signature and prognosis for gastrointestinal cancer
WO2011056963A1 (en) * 2009-11-04 2011-05-12 The University Of North Carolina At Chapel Hill Methods and compositions for predicting survival in subjects with cancer
US8026060B2 (en) 2006-01-11 2011-09-27 Genomic Health, Inc. Gene expression markers for colorectal cancer prognosis
EP2664679A1 (en) 2008-05-30 2013-11-20 The University of North Carolina at Chapel Hill Gene expression profiles to predict breast cancer outcomes
WO2013177245A2 (en) 2012-05-22 2013-11-28 Nanostring Technologies, Inc. Nano46 genes and methods to predict breast cancer outcome
WO2014075067A1 (en) * 2012-11-12 2014-05-15 Nanostring Technologies, Inc. Methods to predict breast cancer outcome
US8906625B2 (en) 2006-03-31 2014-12-09 Genomic Health, Inc. Genes involved in estrogen metabolism
WO2015035377A1 (en) 2013-09-09 2015-03-12 British Columbia Cancer Agency Branch Methods and kits for predicting outcome and methods and kits for treating breast cancer with radiation therapy
JP2015530072A (en) * 2012-06-29 2015-10-15 ナノストリング テクノロジーズ,インコーポレイティド Breast cancer treatment with gemcitabine therapy
CN107058523A (en) * 2017-03-21 2017-08-18 温州迪安医学检验所有限公司 A kind of genetic test primer of breast carcinoma recurring risk assessment 21 and its application
US10179936B2 (en) 2009-05-01 2019-01-15 Genomic Health, Inc. Gene expression profile algorithm and test for likelihood of recurrence of colorectal cancer and response to chemotherapy
US11746378B2 (en) 2016-05-06 2023-09-05 Oxford BioDynamics PLC Detection of chromosome interaction relevant to breast cancer

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009046141A2 (en) * 2007-10-01 2009-04-09 Isis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
BRPI0913578A2 (en) 2008-05-14 2017-06-06 Dermtech Int diagnosis of melanoma and solar lentigo by nucleic acid analysis
US20120041274A1 (en) * 2010-01-07 2012-02-16 Myriad Genetics, Incorporated Cancer biomarkers
EP2513340B1 (en) * 2009-12-14 2016-06-29 North Carolina State University Mean dna copy number of chromosomal regions is of prognostic significance in cancer
US10378064B1 (en) 2010-04-16 2019-08-13 Chronix Biomedical Analyzing circulating nucleic acids to identify a biomarker representative of cancer presented by a patient population
PL2558854T3 (en) 2010-04-16 2019-04-30 Chronix Biomedical Breast cancer associated circulating nucleic acid biomarkers
GB201012590D0 (en) * 2010-07-27 2010-09-08 Queen Mary & Westfield College Methods for diagnosing cancer
CA2830240A1 (en) 2011-03-15 2012-09-20 The University Of North Carolina At Chapell Hill Methods of treating breast cancer with anthracycline therapy
PT2739153T (en) 2011-07-29 2018-11-28 Univ Colorado Regents Treatment of breast cancer
US9773091B2 (en) 2011-10-31 2017-09-26 The Scripps Research Institute Systems and methods for genomic annotation and distributed variant interpretation
EP2785873A4 (en) 2011-11-30 2015-11-11 Univ North Carolina Methods of treating breast cancer with taxane therapy
US11342048B2 (en) 2013-03-15 2022-05-24 The Scripps Research Institute Systems and methods for genomic annotation and distributed variant interpretation
US10235496B2 (en) 2013-03-15 2019-03-19 The Scripps Research Institute Systems and methods for genomic annotation and distributed variant interpretation
US9418203B2 (en) 2013-03-15 2016-08-16 Cypher Genomics, Inc. Systems and methods for genomic variant annotation
WO2015073870A1 (en) 2013-11-15 2015-05-21 North Carolina State University Chromosomal assessment to diagnose urogenital malignancy in dogs
WO2015160916A1 (en) 2014-04-15 2015-10-22 North Carolina State University Chromosomal assessment to differentiate histiocytic malignancy from lymphoma and hemangiosarcoma in dogs
US10513738B2 (en) 2014-07-24 2019-12-24 North Carolina State University Method to diagnose malignant melanoma in the domestic dog
SG11201704098VA (en) 2014-11-21 2017-06-29 Nanostring Technologies Inc Enzyme- and amplification-free sequencing
AU2015353747B2 (en) 2014-11-24 2021-02-25 Nanostring Technologies, Inc. Methods and apparatuses for gene purification and imaging
EP3604556A1 (en) 2014-12-12 2020-02-05 Medivation Prostate Therapeutics LLC Method for predicting response to breast cancer therapeutic agents and method of treatment of breast cancer
CN109715825B (en) 2016-05-16 2023-01-06 纳米线科技公司 Method for detecting target nucleic acid in sample
US10934590B2 (en) * 2016-05-24 2021-03-02 Wisconsin Alumni Research Foundation Biomarkers for breast cancer and methods of use thereof
CA3025776A1 (en) 2016-05-31 2017-12-07 North Carolina State University Methods of mast cell tumor prognosis and uses thereof
SG10202100951SA (en) 2016-11-21 2021-03-30 Nanostring Technologies Inc Chemical compositions and methods of using same
CN110520887A (en) * 2017-02-17 2019-11-29 气象预报公司 System and method for using the statistical analysis prediction economic trend of weather data
US11094397B2 (en) * 2017-05-12 2021-08-17 Noblis, Inc. Secure communication of sensitive genomic information using probabilistic data structures
CN107245523A (en) * 2017-07-18 2017-10-13 深圳市亿立方生物技术有限公司 Molecular probe and detection kit for breast cancer fluorescence in situ hybridization detection
WO2019152504A1 (en) * 2018-01-30 2019-08-08 Board Of Regents, The University Of Texas System Multi-color fish test for bladder cancer detection
CA3090912A1 (en) * 2018-02-13 2019-08-22 Genecentric Therapeutics, Inc. Methods for subtyping of bladder cancer
US11976332B2 (en) 2018-02-14 2024-05-07 Dermtech, Inc. Gene classifiers and uses thereof in non-melanoma skin cancers
CA3099909A1 (en) 2018-05-14 2019-11-21 Nanostring Technologies, Inc. Chemical compositions and methods of using same
JP2022524641A (en) 2019-03-26 2022-05-09 ダームテック,インク. New gene classifiers and their use in skin cancer

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US739155A (en) 1903-01-03 1903-09-15 Harold A Ley Trackway for bicycle-riders or other performers.

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006010150A2 (en) * 2004-07-15 2006-01-26 University Of Utah Research Foundation Housekeeping genes and methods for identifying the same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US739155A (en) 1903-01-03 1903-09-15 Harold A Ley Trackway for bicycle-riders or other performers.

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BERTUCCI ET AL., PATHOLOGIE BIOLOGIE, vol. 54, 2006, pages 49 - 54
CALZA ET AL., BREAST CANCER RESEARCH, vol. 8, no. 4, 2006, pages R34
OH ET AL., J CLIN ONCOL, vol. 24, no. 11, 2006, pages 1656 - 1664
PERREARD ET AL., BREAST CANCER RESEARCH, vol. 8, no. 4, 2006, pages R23
ROUZIER ET AL., CLIN. CANCER RES., vol. 11, no. 16, 2005, pages 5678 - 5685
See also references of EP1954708A4

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8273537B2 (en) 2006-01-11 2012-09-25 Genomic Health, Inc. Gene expression markers for colorectal cancer prognosis
US8026060B2 (en) 2006-01-11 2011-09-27 Genomic Health, Inc. Gene expression markers for colorectal cancer prognosis
US8029995B2 (en) 2006-01-11 2011-10-04 Genomic Health, Inc. Gene expression markers for colorectal cancer prognosis
US8153378B2 (en) 2006-01-11 2012-04-10 Genomic Health, Inc. Gene expression markers for colorectal cancer prognosis
US8153380B2 (en) 2006-01-11 2012-04-10 Genomic Health, Inc. Gene expression markers for colorectal cancer prognosis
US8153379B2 (en) 2006-01-11 2012-04-10 Genomic Health, Inc. Gene expression markers for colorectal cancer prognosis
US8198024B2 (en) 2006-01-11 2012-06-12 Genomic Health, Inc. Gene expression markers for colorectal cancer prognosis
US8367345B2 (en) 2006-01-11 2013-02-05 Genomic Health Inc. Gene expression markers for colorectal cancer prognosis
US8906625B2 (en) 2006-03-31 2014-12-09 Genomic Health, Inc. Genes involved in estrogen metabolism
EP2215254A4 (en) * 2007-10-05 2012-07-18 Pacific Edge Biotechnology Ltd Proliferation signature and prognosis for gastrointestinal cancer
EP2215254A1 (en) * 2007-10-05 2010-08-11 Pacific Edge Biotechnology Ltd Proliferation signature and prognosis for gastrointestinal cancer
JP2010539973A (en) * 2007-10-05 2010-12-24 パシフィック エッジ バイオテクノロジー リミティド Signs of growth and prognosis in gastrointestinal cancer
EP2664679A1 (en) 2008-05-30 2013-11-20 The University of North Carolina at Chapel Hill Gene expression profiles to predict breast cancer outcomes
US9631239B2 (en) 2008-05-30 2017-04-25 University Of Utah Research Foundation Method of classifying a breast cancer instrinsic subtype
US10179936B2 (en) 2009-05-01 2019-01-15 Genomic Health, Inc. Gene expression profile algorithm and test for likelihood of recurrence of colorectal cancer and response to chemotherapy
WO2011056963A1 (en) * 2009-11-04 2011-05-12 The University Of North Carolina At Chapel Hill Methods and compositions for predicting survival in subjects with cancer
WO2013177245A2 (en) 2012-05-22 2013-11-28 Nanostring Technologies, Inc. Nano46 genes and methods to predict breast cancer outcome
JP2015530072A (en) * 2012-06-29 2015-10-15 ナノストリング テクノロジーズ,インコーポレイティド Breast cancer treatment with gemcitabine therapy
EP2867370A4 (en) * 2012-06-29 2016-06-29 Nanostring Technologies Inc Methods of treating breast cancer with gemcitabine therapy
WO2014075067A1 (en) * 2012-11-12 2014-05-15 Nanostring Technologies, Inc. Methods to predict breast cancer outcome
WO2015035377A1 (en) 2013-09-09 2015-03-12 British Columbia Cancer Agency Branch Methods and kits for predicting outcome and methods and kits for treating breast cancer with radiation therapy
US11746378B2 (en) 2016-05-06 2023-09-05 Oxford BioDynamics PLC Detection of chromosome interaction relevant to breast cancer
CN107058523A (en) * 2017-03-21 2017-08-18 温州迪安医学检验所有限公司 A kind of genetic test primer of breast carcinoma recurring risk assessment 21 and its application

Also Published As

Publication number Publication date
WO2007061876A3 (en) 2007-09-20
EP1954708A4 (en) 2009-05-13
US20140087959A1 (en) 2014-03-27
CA2630974A1 (en) 2007-05-31
EP1954708A2 (en) 2008-08-13
US20090299640A1 (en) 2009-12-03
WO2007061876A8 (en) 2007-10-25
US20170044618A1 (en) 2017-02-16

Similar Documents

Publication Publication Date Title
EP1954708A2 (en) Methods and compositions involving intrinsic genes
Perreard et al. Classification and risk stratification of invasive breast carcinomas using a real-time quantitative RT-PCR assay
US8868352B2 (en) Predicting response to chemotherapy using gene expression markers
US8906625B2 (en) Genes involved in estrogen metabolism
US8349555B2 (en) Methods and compositions for predicting death from cancer and prostate cancer survival using gene expression signatures
EP2195467B1 (en) Tumor grading and cancer prognosis in breast cancer
US20070128636A1 (en) Predictors Of Patient Response To Treatment With EGFR Inhibitors
US20100292933A1 (en) Methods, systems, and compositions for classification, prognosis, and diagnosis of cancers
US20190085407A1 (en) Methods and compositions for diagnosis of glioblastoma or a subtype thereof
EP2140020A2 (en) Gene expression markers for prediction of patient response to chemotherapy
KR20140105836A (en) Identification of multigene biomarkers
WO2006010150A2 (en) Housekeeping genes and methods for identifying the same
JP2006516897A (en) Gene expression markers for breast cancer prognosis
US9890430B2 (en) Copy number aberration driven endocrine response gene signature
EP1668151B1 (en) Materials and methods relating to breast cancer diagnosis
KR20180002882A (en) Gene expression profile and its use for breast cancer
Abruzzo et al. Identification and validation of biomarkers of IgVH mutation status in chronic lymphocytic leukemia using microfluidics quantitative real-time polymerase chain reaction technology
US20140086911A1 (en) Methods and Products for Predicting CMTC Class and Prognosis in Breast Cancer Patients
Musella et al. Use of formalin-fixed paraffin-embedded samples for gene expression studies in breast cancer patients
Parker Clinical implementation of breast cancer genomics

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2630974

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006844413

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12094898

Country of ref document: US