WO2012006447A2 - Gene signatures for cancer prognosis - Google Patents

Gene signatures for cancer prognosis Download PDF

Info

Publication number
WO2012006447A2
WO2012006447A2 PCT/US2011/043228 US2011043228W WO2012006447A2 WO 2012006447 A2 WO2012006447 A2 WO 2012006447A2 US 2011043228 W US2011043228 W US 2011043228W WO 2012006447 A2 WO2012006447 A2 WO 2012006447A2
Authority
WO
WIPO (PCT)
Prior art keywords
genes
cancer
protein
expression
test
Prior art date
Application number
PCT/US2011/043228
Other languages
French (fr)
Other versions
WO2012006447A3 (en
Inventor
Steve Stone
Alexander Gutin
Susanne Wagner
Julia Reid
Original Assignee
Myriad Genetics, Inc.
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 Myriad Genetics, Inc. filed Critical Myriad Genetics, Inc.
Priority to CA2804391A priority Critical patent/CA2804391A1/en
Priority to EP11804357.9A priority patent/EP2591126B1/en
Priority to EP20210783.5A priority patent/EP3812469A1/en
Publication of WO2012006447A2 publication Critical patent/WO2012006447A2/en
Publication of WO2012006447A3 publication Critical patent/WO2012006447A3/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12Q2600/156Polymorphic or mutational markers
    • 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
    • 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
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • the invention generally relates to a molecular classification of disease and particularly to molecular markers for cancer prognosis and methods of use thereof.
  • Cancer is a major public health problem, accounting for roughly 25% of all deaths in the United States. Though many treatments have been devised for various cancers, these treatments often vary in severity of side effects. It is useful for clinicians to know how aggressive a patient's cancer is in order to determine how aggressively to treat the cancer.
  • the present invention is based in part on the surprising discovery that the expression of those genes whose expression closely tracks the cell cycle (“cell-cycle genes” or “CCGs” as further defined below) is particularly useful in classifying selected types of cancer and determining the prognosis of these cancers.
  • a method for determining gene expression in a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer.
  • the method includes at least the following steps: (1) obtaining a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer; (2) determining the expression of a panel of genes in said tumor sample including at least 4 cell-cycle genes; and (3) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide said test value, wherein at least 50%, at least 75% or at least 90% of said plurality of test genes are cell-cycle genes.
  • the plurality of test genes includes at least 8 cell- cycle genes, or at least 10, 15, 20, 25 or 30 cell-cycle genes. Preferably, all of the test genes are cell- cycle genes.
  • the step of determining the expression of the panel of genes in the tumor sample comprises measuring the amount of mRNA in the tumor sample transcribed from each of from 4 to about 200 cell-cycle genes; and measuring the amount of mRNA of one or more housekeeping genes in the tumor sample.
  • a method for determining the prognosis of prostate cancer, lung cancer, bladder cancer or brain cancer which comprises determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of at least 6, 8 or 10 cell-cycle genes, wherein overexpression of said at least 6, 8 or 10 cell-cycle genes indicates a poor prognosis or an increased likelihood of recurrence of cancer in the patient.
  • the prognosis method comprises (1) determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of a panel of genes in said tumor sample including at least 4 or at least 8 cell- cycle genes; and (2) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from the panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide the test value, wherein at least 50%, at least 75% or at least 85% of the plurality of test genes are cell-cycle genes, and wherein an increased level of overall expression of the plurality of test genes indicates a poor prognosis, whereas if there is no increase in the overall expression of the test genes, it would indicate a good prognosis or a low likelihood of recurrence of cancer in the patient.
  • the prognosis method further includes a step of comparing the test value provided in step (2) above to one or more reference values, and correlating the test value to a risk of cancer progression or risk of cancer recurrence. Optionally an increased likelihood of poor prognosis is indicated if the test value is greater than the reference value.
  • the present invention also provide a method of treating cancer in a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer, comprising: (1) determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of a panel of genes in the tumor sample including at least 4 or at least 8 cell-cycle genes; (2) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide said test value, wherein at least 50% or 75% or 85% of the plurality of test genes are cell-cycle genes, wherein an increased level of expression of the plurality of test genes indicates a poor prognosis, and an un- increased level of expression of the plurality of test genes indicates a good prognosis; and recommending, prescribing or administering a treatment regimen or watchful waiting based on the prognosis provided in step (2).
  • the present invention further provides a diagnostic kit for prognosing cancer in a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, comprising, in a compartmentalized container, a plurality of oligonucleotides hybridizing to at least 8 test genes, wherein less than 10%, 30% or less than 40% of all of the at least 8 test genes are non-cell-cycle genes; and one or more oligonucleotide hybridizing to at least one housekeeping gene.
  • the oligonucleotides can be hybridizing probes for hybridization with the test genes under stringent conditions or primers suitable for PCR amplification of the test genes.
  • the kit consists essentially of, in a compartmentalized container, a first plurality of PCR reaction mixtures for PCR amplification of from 5 or 10 to about 300 test genes, wherein at least 50%, at least 60% or at least 80% of such test genes are cell-cycle genes, and wherein each reaction mixture comprises a PCR primer pair for PCR amplifying one of the test genes; and a second plurality of PCR reaction mixtures for PCR amplification of at least one housekeeping gene.
  • the present invention also provides the use of (1) a plurality of
  • oligonucleotides hybridizing to at least 4 or at least 8 cell-cycle genes; and (2) one or more oligonucleotides hybridizing to at least one housekeeping gene, for the manufacture of a diagnostic product for determining the expression of the test genes in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, to predict the prognosis of cancer, wherein an increased level of the overall expression of the test genes indicates a poor prognosis or an increased likelihood of recurrence of cancer in the patient, whereas if there is no increase in the overall expression of the test genes, it would indicate a good prognosis or a low likelihood of recurrence of cancer in the patient.
  • the oligonucleotides are PCR primers suitable for PCR amplification of the test genes. In other embodiments, the oligonucleotides are probes hybridizing to the test genes under stringent conditions. In some embodiments, the plurality of oligonucleotides are probes for hybridization under stringent conditions to, or are suitable for PCR amplification of, from 4 to about 300 test genes, at least 50%, 70% or 80% or 90% of the test genes being cell-cycle genes.
  • the plurality of oligonucleotides are hybridization probes for, or are suitable for PCR amplification of, from 20 to about 300 test genes, at least 30%, 40%, 50%, 70% or 80% or 90% of the test genes being cell-cycle genes.
  • the present invention further provides a system for determining gene expression in a tumor sample, comprising: (1) a sample analyzer for determining the expression levels of a panel of genes in a tumor sample including at least 4 cell-cycle genes, wherein the sample analyzer contains the tumor sample which is from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer, or cDNA molecules from mRNA expressed from the panel of genes; (2) a first computer program means for (a) receiving gene expression data on at least 4 test genes selected from the panel of genes, (b) weighting the determined expression of each of the test genes with a predefined coefficient, and (c) combining the weighted expression to provide a test value, wherein at least 50%, at least at least 75% of at least 4 test genes are cell-cycle genes; and optionally (3) a second computer program means for comparing the test value to one or more reference values each associated with a predetermined degree of risk of cancer recurrence or progression of the prostate cancer, lung cancer, bladder cancer or brain cancer.
  • a sample analyzer
  • the system further comprises a display module displaying the comparison between the test value to the one or more reference values, or displaying a result of the comparing step.
  • Figure 1 is an illustration of the predictive power over nomogram for
  • Figure 2 is an illustration of CCGs predicting time to recurrence.
  • Figure 3 is an illustration of nomogram predicting time to recurrence.
  • Figure 4 is an illustration of the non-overlapping recurrence predicted by nomogram and a CCG signature.
  • Figure 5 is an illustration of time to recurrence for several patient populations defined by nomogram and/or CCG status.
  • Figure 6 is an illustration of an example of a system useful in certain aspects and embodiments of the invention.
  • Figure 7 is a flowchart illustrating an example of a computer-implemented method of the invention.
  • Figure 8 a scatter plot comparing clinical parameters and CCG score as predictors of recurrence from Example 5.
  • Figure 9 illustrates, from Example 5, the CCG threshold derived from analysis of the training cohort to the validation data set, with the CCG signature score effectively subdividing patients identified as low-risk using clinical parameters into patients with very low recurrence rates and a higher risk of recurrence.
  • Figure 10 illustrates the predicted recurrence rate versus CCG score for patients in the validation cohort of Example 5.
  • Figure 11 illustrates the predicted recurrence rate versus CCG score for patients in the validation cohort of Example 5.
  • Figure 12 illustrates the distribution of clinical risk score in 443 patients studied in Example 5.
  • the dark vertical line represents the threshold chosen by KM means to divide low- and high-risk patients and used throughout this study.
  • Figure 13 illustrates the correlation between CCP score and survival in brain cancer.
  • Figure 14 illustrates illustrates the correlation between CCP score and survival in bladder cancer.
  • Figure 15 illustrates illustrates the correlation between CCP score and survival in breast cancer.
  • Figure 16 illustrates the correlation between CCP score and survival in lung cancer.
  • Figure 17 is an illustration of the predictive power over nomogram for CCG panels of different sizes.
  • the present invention is based in part on the discovery that genes whose expression closely tracks the cell cycle (“cell-cycle genes” or “CCGs”) are particularly powerful genes for classifying selected cancers including prostate cancer, lung cancer, bladder cancer, brain cancer and breast cancer, but not other types of cancer such as colorectal cancer.
  • CCG gene and “CCG” herein refer to a gene whose expression level closely tracks the progression of the cell through the cell-cycle. See, e.g., Whitfield et al, MOL. BIOL. CELL (2002) 13: 1977-2000.
  • the term "cell-cycle progression" or “CCP” will also be used in this application and will generally be interchangeable with CCG (i.e., a CCP gene is a CCG; a CCP score is a CCG score). More specifically, CCGs show periodic increases and decreases in expression that coincide with certain phases of the cell cycle— e.g., STK15 and PLK show peak expression at G2/M. Id.
  • CCGs have clear, recognized cell-cycle related function—e.g., in DNA synthesis or repair, in chromosome condensation, in cell-division, etc.
  • some CCGs have expression levels that track the cell-cycle without having an obvious, direct role in the cell- cycle— e.g., UBE2S encodes a ubiquitin-conjugating enzyme, yet its expression closely tracks the cell-cycle.
  • a CCG according to the present invention need not have a recognized role in the cell-cycle.
  • Exemplary CCGs are listed in Tables 1, 2, 3, and 4.
  • Whether a particular gene is a CCG may be determined by any technique known in the art, including that taught in Whitfield et al, MOL. BIOL. CELL (2002) 13: 1977-2000.
  • a sample of cells e.g., HeLa cells
  • this is done by arresting the cells in each phase— e.g., cells may be arrested in S phase by using a double thymidine block or in mitosis with a thymidine-nocodazole block. See, e.g., Whitfield et al., MOL. CELL. BIOL. (2000) 20:4188-4198.
  • RNA is extracted from the cells after arrest in each phase and gene expression is quantitated using any suitable technique—e.g. , expression microarray (genome-wide or specific genes of interest), real-time quantitative PCRTM (RTQ-PCR). Finally, statistical analysis (e.g., Fourier Transform) is applied to determine which genes show peak expression during particular cell- cycle phases. Genes may be ranked according to a periodicity score describing how closely the gene's expression tracks the cell-cycle— e.g., a high score indicates a gene very closely tracks the cell cycle. Finally, those genes whose periodicity score exceeds a defined threshold level (see Whitfield et al, MOL. BIOL. CELL (2002) 13: 1977-2000) may be designated CCGs.
  • CCGs A large, but not exhaustive, list of nucleic acids associated with CCGs (e.g., genes, ESTs, cDNA clones, etc.) is given in Table 1. See Whitfield et al, MOL. BIOL. CELL (2002) 13:1977-2000. All of the CCGs in Table 2 below form a panel of CCGs ("Panel A") useful in the methods of the invention.
  • PAICS* 10606 Hs00272390_ml NM 001079525.1;
  • ABI Assay ID means the catalogue ID number for the gene expression assay commercially available from Applied Biosystems Inc. (Foster City, CA) for the particular gene.
  • the method includes at least the following steps: (1) obtaining a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer; (2) determining the expression of a panel of genes in the tumor sample including at least 2, 4, 6, 8 or 10 cell-cycle genes; and (3) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide said test value, wherein at least 20%, 50%>, at least 75% or at least 90%> of said plurality of test genes are cell- cycle genes.
  • Gene expression can be determined either at the R A level (i.e., m NA or noncoding RNA (ncRNA)) (e.g., miRNA, tRNA, rRNA, snoRNA, siRNA and piRNA) or at the protein level.
  • R A level i.e., m NA or noncoding RNA (ncRNA)
  • ncRNA noncoding RNA
  • levels of proteins in a tumor sample can be determined by any known techniques in the art, e.g., HPLC, mass spectrometry, or using antibodies specific to selected proteins (e.g., IHC, ELISA, etc.).
  • the amount of RNA transcribed from the panel of genes including test genes is measured in the tumor sample.
  • the amount of RNA of one or more housekeeping genes in the tumor sample is also measured, and used to normalize or calibrate the expression of the test genes.
  • normalizing genes and “housekeeping genes” are defined herein below.
  • the plurality of test genes includes at least 2, 3 or 4 cell-cycle genes, which constitute at least 50%>, 75% or 80%> of the plurality of test genes, and preferably 100% of the plurality of test genes.
  • the plurality of test genes includes at least 5, 6, 7, or at least 8 cell-cycle genes, which constitute at least 20%>, 25%, 30%>, 40%>, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes.
  • a panel of genes is a plurality of genes. Typically these genes are assayed together in one or more samples from a patient.
  • the plurality of test genes includes at least 8, 10,
  • tumor sample means any biological sample containing one or more tumor cells, or one or more tumor derived R A or protein, and obtained from a cancer patient.
  • tissue sample obtained from a tumor tissue of a cancer patient is a useful tumor sample in the present invention.
  • the tissue sample can be an FFPE sample, or fresh frozen sample, and preferably contain largely tumor cells.
  • a single malignant cell from a cancer patient's tumor is also a useful tumor sample.
  • a malignant cell can be obtained directly from the patient's tumor, or purified from the patient's bodily fluid such as blood and urine.
  • a bodily fluid such as blood, urine, sputum and saliva containing one or tumor cells, or tumor-derived RNA or proteins, can also be useful as a tumor sample for purposes of practicing the present invention.
  • telomere PCRTM quantitative realtime PCRTM
  • immunoanalysis e.g., ELISA
  • the activity level of a polypeptide encoded by a gene may be used in much the same way as the expression level of the gene or polypeptide. Often higher activity levels indicate higher expression levels and while lower activity levels indicate lower expression levels. Thus, in some embodiments, the invention provides any of the methods discussed above, wherein the activity level of a polypeptide encoded by the CCG is determined rather than or in adition to the expression level of the CCG. Those skilled in the art are familiar with techniques for measuring the activity of various such proteins, including those encoded by the genes listed in Tables 1, 2, 3, and 4. The methods of the invention may be practiced independent of the particular technique used.
  • the expression of one or more normalizing genes is also obtained for use in normalizing the expression of test genes.
  • normalizing genes referred to the genes whose expression is used to calibrate or normalize the measured expression of the gene of interest (e.g., test genes).
  • the expression of normalizing genes should be independent of cancer outcome/prognosis, and the expression of the normalizing genes is very similar among all the tumor samples. The normalization ensures accurate comparison of expression of a test gene between different samples. For this purpose, housekeeping genes known in the art can be used.
  • Housekeeping genes are well known in the art, with examples including, but are not limited to, GUSB (glucuronidase, beta), HMBS (hydroxymethylbilane synthase), SDHA (succinate dehydrogenase complex, subunit A, flavoprotein), UBC (ubiquitin C) and YWHAZ (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide).
  • GUSB glucose curonidase, beta
  • HMBS hydroxymethylbilane synthase
  • SDHA succinate dehydrogenase complex, subunit A, flavoprotein
  • UBC ubiquitin C
  • YWHAZ tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide.
  • One or more housekeeping genes can be used.
  • at least 2, 5, 10 or 15 housekeeping genes are used to provide a combined normalizing gene
  • RNA levels for the genes In the case of measuring RNA levels for the genes, one convenient and sensitive approach is real-time quantitative PCR (qPCR) assay, following a reverse transcription reaction.
  • qPCR quantitative PCR
  • a cycle threshold C t is determined for each test gene and each normalizing gene, i.e., the number of cycle at which the fluoescence from a qPCR reaction above background is detectable.
  • the overall expression of the one or more normalizing genes can be represented by a "normalizing value" which can be generated by combining the expression of all normalizing genes, either weighted eaqually (straight addition or averaging) or by different predefined coefficients.
  • the normalizing value Cm can be the cycle threshold (C t ) of one single normalizing gene, or an average of the C t values of 2 or more, preferably 10 or more, or 15 or more normalizing genes, in which case, the predefined coefficient is 1/N, where N is the total number of normalizing genes used.
  • QH (Qm + Qm + " C t Hn) N.
  • the methods of the invention generally involve determining the level of expression of a panel of CCGs. With modern high-throughput techniques, it is often possible to determine the expression level of tens, hundreds or thousands of genes. Indeed, it is possible to determine the level of expression of the entire transcriptome (i.e., each transcribed sequence in the genome). Once such a global assay has been performed, one may then
  • transcripts i.e., panels or, as often used herein, pluralities of test genes.
  • one may analyze (e.g., informatically) the expression of a panel or plurality of test genes comprising primarily CCGs according to the present invention by combining the expression level values of the individual test genes to obtain a test value.
  • the test value provided in the present inveniton represents the overall expression level of the plurality of test genes composed substantially of cell-cycle genes.
  • the test value representing the overall expression of the plurality of test genes can be provided by combining the normalized expression of all test genes, either by straight addition or averaging (i.e., weighted eaqually) or by a different predefined coefficient.
  • test value (AC tl + AC t2 + ' " + AC tn )/n.
  • the plurality of test genes comprises the top 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40 or more CCGs listed in Tables 9, 11, 23, 24 or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 of the following genes: ASPM, BIRC5, BUB1B, CCNB2, CDC2, CDC20, CDCA8, CDKN3, CENPF, DLGAP5, FOXM1, KIAA0101, KIF11, KIF2C, KIF4A, MCM10, NUSAP1, PRC1,
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, or nine or all of gene numbers 2 & 3, 2 to 4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, or 2 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, or nine or all of gene numbers 2 & 3, 2 to 4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, or 2 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, or eight or all of gene numbers 3 & 4, 3 to 5, 3 to 6, 3 to 7, 3 to 8, 3 to 9, or 3 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, or eight or all of gene numbers 3 & 4, 3 to 5, 3 to 6, 3 to 7, 3 to 8, 3 to 9, or 3 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, 10, 11, 12, 13, 14, or 15 or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to
  • a method for prognosing cancer selected from prostate cancer, lung cancer, bladder cancer or brain cancer which comprises determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of at least 2, 4, 5, 6, 7 or at least 8, 9, 10 or 12 cell-cycle genes, wherein overexpression of the at least 4 cell-cycle genes indicates a poor prognosis or an increased likelihood of recurrence of cancer in the patient.
  • the expression can be determined in accordance with the method described above.
  • the prognosis method includes (1) obtaining a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer; (2) determining the expression of a panel of genes in the tumor sample including at least 2, 4,
  • test value by (a) weighting the determined expression of each of a plurality of test genes selected from the panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide said test value, wherein at least 20%, 50%>, at least 75%) or at least 90%> of said plurality of test genes are cell-cycle genes, and wherein an increased level of expression of the plurality of test genes indicates a poor prognosis or an increased likelihood of cancer recurrence.
  • the test value representing the overall expression of the plurality of test genes is compared to one or more reference values (or index values), and optionally correlated to a risk of cancer progression or risk of cancer recurrence.
  • a risk of cancer progression or risk of cancer recurrence is optionally correlated to a risk of cancer progression or risk of cancer recurrence.
  • an increased likelihood of poor prognosis is indicated if the test value is greater than the reference value.
  • the index value may represent the gene expression levels found in a normal sample obtained from the patient of interest, in which case an expression level in the tumor sample significantly higher than this index value would indicate, e.g., a poor prognosis or increased likelihood of cancer recurrence or a need for aggressive treatment.
  • the index value may represent the average expression level of for a set of individuals from a diverse cancer population or a subset of the population. For example, one may determine the average expression level of a gene or gene panel in a random sampling of patients with cancer (e.g., prostate, bladder, brain, breast, or lung cancer). This average expression level may be termed the "threshold index value,” with patients having CCG expression higher than this value expected to have a poorer prognosis than those having expression lower than this value.
  • cancer e.g., prostate, bladder, brain, breast, or lung cancer
  • the index value may represent the average expression level of a particular gene marker in a plurality of training patients ⁇ e.g., prostate cancer patients) with similar outcomes whose clinical and follow-up data are available and sufficient to define and categorize the patients by disease outcome, e.g., recurrence or prognosis. See, e.g., Examples, infra.
  • a "good prognosis index value" can be generated from a plurality of training cancer patients characterized as having "good outcome", e.g., those who have not had cancer recurrence five years (or ten years or more) after initial treatment, or who have not had progression in their cancer five years (or ten years or more) after initial diagnosis.
  • a “poor prognosis index value” can be generated from a plurality of training cancer patients defined as having “poor outcome”, e.g., those who have had cancer recurrence within five years (or ten years, etc.) after initial treatment, or who have had progression in their cancer within five years (or ten years, etc.) after initial diagnosis.
  • a good prognosis index value of a particular gene may represent the average level of expression of the particular gene in patients having a "good outcome”
  • a poor prognosis index value of a particular gene represents the average level of expression of the particular gene in patients having a "poor outcome.”
  • one aspect of the invention provides a method of classifying cancer comprising determining the status of a panel of genes comprising at least two CCGs, in tissue or cell sample, particularly a tumor sample, from a patient, wherein an abnormal status indicates a negative cancer classification.
  • determining the status of a gene refers to determining the presence, absence, or extent/level of some physical, chemical, or genetic characteristic of the gene or its expression product(s). Such characteristics include, but are not limited to, expression levels, activity levels, mutations, copy number, methylation status, etc.
  • characteristics include expression levels (e.g., mR A or protein levels) and activity levels. Characteristics may be assayed directly (e.g., by assaying a CCG's expression level) or determined indirectly (e.g., assaying the level of a gene or genes whose expression level is correlated to the expression level of the CCG).
  • some embodiments of the invention provide a method of classifying cancer comprising determining the expression level, particularly mRNA level of a panel of genes comprising at least two CCGs, in a tumor sample, wherein elevated expression indicates a negative cancer classification, or an increased risk of cancer recurrence or progression, or a need for aggressive treatment.
  • Abnormal status means a marker's status in a particular sample differs from the status generally found in average samples (e.g., healthy samples or average diseased samples). Examples include mutated, elevated, decreased, present, absent, etc.
  • An "elevated status” means that one or more of the above characteristics (e.g., expression or mRNA level) is higher than normal levels. Generally this means an increase in the characteristic (e.g., expression or mRNA level) as compared to an index value.
  • a “low status” means that one or more of the above characteristics (e.g., gene expression or mRNA level) is lower than normal levels. Generally this means a decrease in the characteristic (e.g., expression) as compared to an index value.
  • a "negative status" generally means the characteristic is absent or undetectable.
  • PTEN status is negative if PTEN nucleic acid and/or protein is absent or undetectable in a sample.
  • negative PTEN status also includes a mutation or copy number reduction in PTEN.
  • the methods comprise determining the expression of one or more CCGs and, if this expression is "increased," the patient has a poor prognosis.
  • "increased" expression of a CCG means the patient's expression level is either elevated over a normal index value or a threshold index (e.g. , by at least some threshold amount) or closer to the "poor prognosis index value" than to the "good prognosis index value.”
  • index values may be determined thusly:
  • a threshold value will be set for the cell cycle mean.
  • the optimal threshold value is selected based on the receiver operating characteristic (ROC) curve, which plots sensitivity vs (1 - specificity).
  • ROC receiver operating characteristic
  • the sensitivity and specificity of the test is calculated using that value as a threshold.
  • the actual threshold will be the value that optimizes these metrics according to the artisans requirements (e.g., what degree of sensitivity or specificity is desired, etc.).
  • Example 5 demonstrates determination of a threshold value determined and validated experimentally.
  • Panels of CCGs can accurately predict prognosis, as shown in Example 3.
  • Those skilled in the art are familiar with various ways of determining the expression of a panel of genes (i.e., a plurality of genes).
  • Increased expression in this context will mean the average expression is higher than the average expression level of these genes in normal patients (or higher than some index value that has been determined to represent the average expression level in a reference population such as patients with the same cancer).
  • a panel of genes by determining the average expression level (normalized or absolute) of at least a certain number (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30 or more) or at least a certain proportion (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100%) of the genes in the panel.
  • a certain proportion e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100%
  • the expression of a panel of genes by determining the absolute copy number of the mRNA (or protein) of all the genes in the panel and either total or average these across the genes.
  • classifying a cancer and “cancer classification” refer to determining one or more clinically-relevant features of a cancer and/or determining a particular prognosis of a patient having said cancer.
  • classifying a cancer includes, but is not limited to: (i) evaluating metastatic potential, potential to metastasize to specific organs, risk of recurrence, and/or course of the tumor; (ii) evaluating tumor stage; (iii) determining patient prognosis in the absence of treatment of the cancer; (iv) determining prognosis of patient response (e.g. , tumor shrinkage or progression- free survival) to treatment (e.g.
  • a "negative classification” means an unfavorable clinical feature of the cancer (e.g., a poor prognosis).
  • examples include (i) an increased metastatic potential, potential to metastasize to specific organs, and/or risk of recurrence; (ii) an advanced tumor stage; (iii) a poor patient prognosis in the absence of treatment of the cancer; (iv) a poor prognosis of patient response (e.g. , tumor shrinkage or progression- free survival) to a particular treatment (e.g.
  • a recurrence- associated clinical parameter or a high nomogram score
  • increased expression of a CCG indicate a negative classification in cancer (e.g., increased likelihood of recurrence or progression).
  • the invention provides a method of classifying cancer comprising determining the status of a panel of genes comprising at least two CCGs, wherein an abnormal status indicates an increased likelihood of recurrence or progression.
  • the status to be determined is gene expression levels.
  • the invention provides a method of determining the prognosis of a patient's cancer comprising determining the expression level of a panel of genes comprising at least two CCGs, wherein elevated expression indicates an increased likelihood of recurrence or progression of the cancer.
  • a patient has an "increased likelihood" of some clinical feature or outcome (e.g., recurrence or progression) if the probability of the patient having the feature or outcome exceeds some reference probability or value.
  • the reference probability may be the probability of the feature or outcome across the general relevant patient population. For example, if the probability of recurrence in the general prostate cancer population is X% and a particular patient has been determined by the methods of the present invention to have a probability of recurrence of Y%, and if Y > X, then the patient has an "increased likelihood" of recurrence.
  • a threshold or reference value may be determined and a particular patient's probability of recurrence may be compared to that threshold or reference.
  • predicting prognosis will often be used herein to refer to either or both.
  • a “poor prognosis” will generally refer to an increased likelihood of recurrence, progression, or both.
  • the invention provides a method of predicting prognosis comprising determining the expression of at least one CCG listed in Table 1 or Panels A through G.
  • Example 3 also shows that panels of CCGs (e.g., 2, 3, 4, 5, or 6 CCGs) can accurately predict prognosis.
  • the invention provides a method of classifying a cancer comprising determining the status of a panel of genes (e.g., a plurality of test genes) comprising a plurality of CCGs.
  • a panel of genes e.g., a plurality of test genes
  • increased expression in a panel of genes (or plurality of test genes) may refer to the average expression level of all panel or test genes in a particular patient being higher than the average expression level of these genes in normal patients (or higher than some index value that has been determined to represent the normal average expression level).
  • increased expression in a panel of genes may refer to increased expression in at least a certain number (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30 or more) or at least a certain proportion (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100%) of the genes in the panel as compared to the average normal expression level.
  • a certain number e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30 or more
  • a certain proportion e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100%
  • the panel comprises at least 3, 4, 5, 6, 7, 8, 9, 10, 15,
  • the panel comprises at least 10, 15, 20, or more CCGs. In some embodiments the panel comprises between 5 and 100 CCGs, between 7 and 40 CCGs, between 5 and 25 CCGs, between 10 and 20 CCGs, or between 10 and 15 CCGs. In some embodiments CCGs comprise at least a certain proportion of the panel. Thus in some embodiments the panel comprises at least 25%, 30%>, 40%>, 50%>, 60%>, 70%>, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% CCGs.
  • the panel comprises at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 70, 80, 90, 100, 200, or more CCGs, and such CCGs constitute of at least 50%, 60%, 70%, preferably at least 75%, 80%, 85%, more preferably at least 90%, 95%, 96%, 97%, 98%, or 99% or more of the total number of genes in the panel.
  • the CCGs are chosen from the group consisting of the genes in Table 1 and Panels A through G.
  • the panel comprises at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 20, 25, 30, or more of the genes in any of Table 1 and Panels A through G.
  • the invention provides a method of predicting prognosis comprising determining the status of the CCGs in Panels A through G, wherein abnormal status indicates a poor prognosis.
  • elevated expression indicates an increased likelihood of recurrence or progression.
  • the invention provides a method of predicting risk of cancer recurrence or progression in a patient comprising determining the status of a panel of genes, wherein the panel comprises between about 10 and about 15 CCGs, the CCGs constitute at least 90% of the panel, and an elevated status for the CCGs indicates an increased likelihood or recurrence or progression.
  • Several panels of CCGs (Table 2, supra, and Tables 3 & 4, infra) have been evaluated for their ability to predict prognosis in several different cancers. The results of these studies are described in Examples 1 through 6 below.
  • CCG signatures the particular CCGs assayed is often not as important as the total number of CCGs.
  • the number of CCGs assayed can vary depending on many factors, e.g., technical constraints, cost considerations, the classification being made, the cancer being tested, the desired level of predictive power, etc.
  • Increasing the number of CCGs assayed in a panel according to the invention is, as a general matter, advantageous because, e.g., a larger pool of mR As to be assayed means less "noise" caused by outliers and less chance of an assay error throwing off the overall predictive power of the test.
  • cost and other considerations will generally limit this number and finding the optimal number of CCGs for a signature is desirable.
  • P is the predictive power (i.e., P Sil is the predictive power of a signature with n genes and P Sil+i is the predictive power of a signature with n genes plus one) and Co is some optimization constant.
  • Predictive power can be defined in many ways known to those skilled in the art including, but not limited to, the signature's p-value.
  • Co can be chosen by the artisan based on his or her specific constraints. For example, if cost is not a critical factor and extremely high levels of sensitivity and specificity are desired, Co can be set very low such that only trivial increases in predictive power are disregarded.
  • Co can be set higher such that only significant increases in predictive power warrant increasing the number of genes in the signature.
  • a graph of predictive power as a function of gene number may be plotted (as in FIG.l) and the second derivative of this plot taken. The point at which the second derivative decreases to some predetermined value (Co') may be the optimal number of genes in the signature.
  • Examples 1 & 3 and FIGs 1 & 17 illustrate the empirical determination of optimal numbers of CCGs in CCG panels of the invention. Randomly selected subsets of the 31 CCGs listed in Table 3 were tested as distinct CCG signatures and predictive power (i.e., p-value) was determined for each. As FIG. 1 shows, p-values ceased to improve significantly between about 10 and about 15 CCGs, thus indicating that an optimal number of CCGs in a prognostic panel is from about 10 to about 15.
  • some embodiments of the invention provide a method of predicting prognosis in a patient having prostate cancer comprising determining the status of a panel of genes, wherein the panel comprises between about 10 and about 15 CCGs and an elevated status for the CCGs indicates a poor prognosis.
  • the panel comprises between about 10 and about 15 CCGs and the CCGs constitute at least 90% of the panel.
  • the panel comprises CCGs plus one or more additional markers that significantly increase the predictive power of the panel (i.e., make the predictive power significantly better than if the panel consisted of only the CCGs). Any other combination of CCGs (including any of those listed in Table 1 or Panels A through G) can be used to practice the invention.
  • CCGs have been determined to be accurate in predicting recurrence in prostate cancer (Examples 1 through 5). Further, CCGs can determine prognosis in bladder, brain, breast and lung cancers, as summarized in Example 6 and Tables 21 and 22 below.
  • the invention provides a method comprising determining the status of a panel of genes comprising at least two CCGs, wherein an abnormal status indicates a poor prognosis.
  • the panel comprises at least 2 genes chosen from the group of genes in at least one of Panels A through G.
  • the panel comprises at least 10 genes chosen from the group of genes in at least one of Panels A through G.
  • the panel comprises at least 15 genes chosen from the group of genes in at least one of Panels A through G.
  • the panel comprises all of the genes in at least one of Panels A through G.
  • the invention also provides a method of determining the prognosis of bladder cancer, comprising determining the status of a panel of genes comprising at least two CCGs (e.g., at least two of the genes in any of Panels B, C, & F), wherein an abnormal status indicates a poor prognosis.
  • the invention also provides a method of determining the prognosis of brain cancer, comprising determining the status of a panel of genes comprising at least two CCGs (e.g., at least two of the genes in any of Panels B, C, & F), wherein an abnormal status indicates a poor prognosis.
  • the invention further provides a method of determining the prognosis of breast cancer, comprising determining the status of a panel of genes comprising at least two CCGs (e.g., at least two of the genes in any of Panels B, C, & F), wherein an abnormal status indicates a poor prognosis.
  • the invention also provides a method of determining the prognosis of lung cancer, comprising determining the status of a panel of genes comprising at least two CCGs (e.g., at least two of the genes in any of Panels B, C, & F), wherein an abnormal status indicates a poor prognosis.
  • the panel comprises at least 3, 4, 5, 6, 7, 8, 9, 10, 15,
  • the panel comprises between 5 and 100 CCGs, between 7 and 40 CCGs, between 5 and 25 CCGs, between 10 and 20 CCGs, or between 10 and 15 CCGs.
  • CCGs comprise at least a certain proportion of the panel.
  • the panel comprises at least 25%, 30%>, 40%>, 50%>, 60%>, 70%>, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% CCGs.
  • the CCGs are chosen from the group consisting of the genes listed in Tables 1, 9 & 11 and Panels A through G.
  • the panel comprises at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more genes chosen from the group of genes in any of Tables 1, 9 or 11 or Panels A through G. In some embodiments the panel comprises all of the genes in any of Tables 1, 9, or 11 or Panels A through G.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises the top 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40 or more CCGs listed in Table 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 of the following genes: ASPM, BIRC5, BUBIB, CCNB2, CDC2, CDC20, CDCA8, CDKN3, CENPF, DLGAP5, FOXMl, KIAAOlOl, KIFll, KIF2C, KIF4A, MCM10, NUSAP1, PRC1, RACGAP1, and TPX2.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • ASPM BIRC5, BUBIB, CCNB2, CDC2, CDC20, CDCA8, CDKN3, CENPF, DLGAP5, FOXMl, KIAAOlOl, KIF
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, or nine or all of gene numbers 2 & 3, 2 to 4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, or 2 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, or nine or all of gene numbers 2 & 3, 2 to 4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, or 2 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, or eight or all of gene numbers 3 & 4, 3 to 5, 3 to 6, 3 to 7, 3 to 8, 3 to 9, or 3 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, or eight or all of gene numbers 3 & 4, 3 to 5, 3 to 6, 3 to 7, 3 to 8, 3 to 9, or 3 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, 10, 11, 12, 13, 14, or 15 or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, 1 to 10, 1 to 11, 1 to 12, 1 to 13, 1 to 14, or 1 to 15 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, 10, 11, 12, 13, 14, or 15 or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, 1 to 10, 1 to 11, 1
  • prostate cancer for example, it has been discovered that a high level of gene expression of any one of the genes in Panels C through F is associated with an increased risk of prostate cancer recurrence or progression in patients whose clinical nomogram score indicates a relatively low risk of recurrence or progression. Because evaluating CCG expression levels can thus detect increased risk not detected using clinical parameters alone, the invention generally provides methods combining evaluating at least one clinical parameter with evaluating the status of at least one CCG.
  • Example 3 shows, even individual CCGs add to clinical parameters in predicting cancer recurrence.
  • one aspect of the invention provides an in vitro diagnostic method comprising determining at least one clinical parameter for a cancer patient and determining the status of at least one CCG in a sample obtained from the patient.
  • assessing the status of multiple CCGs improves predictive power even more (also shown in Example 1).
  • the status of a plurality of CCGs e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 50 or more
  • abnormal status indicates an increased likelihood of recurrence or progression.
  • the patient has prostate cancer.
  • the patient has lung cancer.
  • clinical parameter and “clinical measure” refer to disease or patient characteristics that are typically applied to assess disease course and/or predict outcome.
  • cancer generally include tumor stage, tumor grade, lymph node status, histology, performance status, type of surgery, surgical margins, type of treatment, and age of onset.
  • stage defined by size of tumor and evidence of metastasis
  • Gleason score similar to concept of grade.
  • important clinical parameters in prostate cancer include margin and lymph node status.
  • breast cancer clinicians often use size of index lesion in cm, invasion, number of nodes involved, and grade.
  • certain clinical parameters are correlated with a particular disease character. For example, in cancer generally as well as in specific cancers, certain clinical parameters are correlated with, e.g., likelihood of recurrence or metastasis, prognosis for survival for a certain amount of time, likelihood of response to treatment generally or to a specific treatment, etc. In prostate cancer some clinical parameters are such that their status (presence, absence, level, etc.) is associated with increased likelihood of recurrence.
  • recurrence-associated parameters examples include high PSA levels (e.g., greater than 4ng/ml), high Gleason score, large tumor size, evidence of metastasis, advanced tumor stage, nuclear grade, lymph node involvement, early age of onset.
  • Other types of cancer may have different parameters correlated to likelihood of recurrence or progression, and CCG status, as a measure of proliferative activity, adds to these parameters in predicting prognosis in these cancers.
  • "recurrence-associated clinical parameter” has its conventional meaning for each specific cancer, with which those skilled in the art are quite familiar.
  • Example 5 shows how CCG status can add to one particular grouping of clinical parameters used to determine risk of recurrence in prostate cancer.
  • Clinical parameters in Example 5 include binary variables for organ- confined disease and Gleason score less than or equal to 6, and a continuous variable for logarithmic PSA (Table 14).
  • This model includes all of the clinical parameters incorporated in the post-RP nomogram (i.e., Kattan- Stephenson nomogram) except for Year of RP and the two components of the Gleason score.
  • at least two clinical parameters are assessed along with the expression level of at least one CCG.
  • nomograms are representations (often visual) of a correlation between one or more parameters and one or more patient or disease characters.
  • An example of a prevalent clinical nomogram used in determining a prostate cancer patient's likelihood of recurrence is described in Kattan et al., J. CLIN. ONCOL.
  • This nomogram evaluates a patient by assigning a point value to each of several clinical parameters (year of RP, surgical margins, extracapsular extension, seminal vesicle invasion, lymph node involvement, primary Gleason score, secondary Gleason score, and preoperative PSA level), totalling the points for a patient into a nomogram score, and then predicting the patient's likelihood of being recurrence-free at varying time intervals (up to 10 years) based on this nomogram score.
  • NPI Nottingham Prognostic Index
  • Table 3 above provides an exemplary panel of 31 CCGs (Panel C) and a subset panel of 26 CCGs (Panel D, shown with ) determined in Example 2 to show predictive synergy with the Kattan- Stephenson nomogram in prostate cancer prognosis. It has also been discovered that determining the status of a CCG in a sample obtained from a breast cancer patient, along with the patient's NPI score, is a better prognostic predictor than NPI score alone. See, e.g., Example 6, infra.
  • a clinical nomogram score ⁇ e.g., Kattan- Stephenson or NPI nomogram score
  • Example 3 illustrates the empirical determination of the predictive power of individual CCGs and of several CCG panels of varying size over the Kattan-Stephenson nomogram. Randomly selected subsets of the 31 CCGs listed in Table 3 were tested as distinct CCG signatures and predictive power (i.e., p-value) was determined for each. As FIG. l shows, CCG signatures of 2, 3, 4, 5, 6, 10, 15, 20,
  • the invention provides a method of determining whether a prostate cancer patient has an increased likelihood of recurrence comprising determining the status of a panel of genes comprising at least 2, 3, 4, 5, 6, 10, 15, 20, 25,
  • the method further comprises determining a clinical nomogram score of the patient.
  • the invention further provides a method of determining whether a breast cancer patient has an increased likelihood of recurrence comprising determining the status of a panel of genes comprising at least 2, 3, 4, 5, 6, 10, 15, 20, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 40, 45, 50, 60, 70, 80, 90, or 100 or more CCGs, wherein an elevated status (e.g., increased expression) for the CCGs indicates an increased likelihood of recurrence.
  • the method further comprises determining a clinical nomogram score of the patient.
  • the invention provides a method of determining whether a cancer patient has an increased likelihood of recurrence or progression comprising determining a clinical nomogram score for the patient and determining the status of at least one CCG in a sample obtained from the patient, wherein a high nomogram score and/or an elevated CCG status indicate the patient has an increased likelihood of recurrence or progression.
  • the cancer is prostate cancer.
  • the cancer is lung cancer.
  • this assessment is made before radical prostatectomy
  • a sample of one or more cells are obtained from a prostate cancer patient before or after treatment for analysis according to the present invention.
  • Prostate cancer treatment currently applied in the art includes, e.g., prostatectomy, radiotherapy, hormonal therapy (e.g., using GnRH antagonists, GnRH agonists, antiandrogens), chemotherapy, and high intensity focused ultrasound.
  • one or more prostate tumor cells from prostate cancer tissue are obtained from a prostate cancer patient during biopsy or prostatectomy and are used for analysis in the method of the present invention.
  • the present invention is also based on the discovery that PTEN status predicts aggressive prostate cancer.
  • PTEN status adds to both clinical parameters (e.g., Kattan- Stephenson nomogram) and CCGs (e.g., the genes in Table 1 or Panels A through G).
  • CCGs e.g., the genes in Table 1 or Panels A through G.
  • Negative PTEN status was found to be a significant predictor for risk of recurrence (p-value 0.031).
  • PTEN remained a significant predictor of recurrence after adjusting for post-surgery clinical parameters and the CCG signature shown in Table 3 (p-value 0.026).
  • the combination of PTEN and the CCG signature seems to be a better predictor of recurrence than post-surgery clinical parameters (p-value 0.0002).
  • one aspect of the invention provides a method of predicting a patient's likelihood of prostate cancer recurrence comprising determining PTEN status in a sample from the patient, wherein a low or negative PTEN status indicates the patient has a high likelihood of recurrence.
  • PTEN status can be determined by any technique known in the art, including but not limited to those discussed herein.
  • PTEN adds to CCG status in predicting prostate cancer recurrence
  • another aspect of the invention provides an in vitro method comprising determining PTEN status and determining the status of a plurality of CCGs in a sample obtained from a patient.
  • IHC immunohistochemistry
  • qPCRTM quantitative polymerase chain reaction
  • Some embodiments of the invention provide a method of determining a prostate cancer patient's likelihood of recurrence comprising determining PTEN status in a sample obtained from the patient, determining the status of a plurality of CCGs in a sample obtained from the patient, wherein low or negative PTEN status and/or elevated CCG status indicate the patient has an increased likelihood of recurrence.
  • yet another aspect of the invention provides an in vitro method comprising determining PTEN status and determining at least one clinical parameter for a cancer patient. Often the clinical parameter is at least somewhat independently predictive of recurrence and the addition of PTEN status improves the predictive power.
  • the invention provides a method of determining whether a cancer patient has an increased likelihood of recurrence comprising determining the status of PTEN in a sample obtained from the patient and determining a clinical nomogram score for the patient, wherein low or negative PTEN status and/or a high nomogram score indicate the patient has an increased likelihood of recurrence.
  • some embodiments of the invention provide a method of determining whether a cancer patient has an increased likelihood of recurrence comprising determining the status of PTEN in a sample obtained from the patient, determining a clinical nomogram score for the patient and determining the status of at least one CCG in a sample obtained from the patient, wherein low or negative PTEN status, a high nomogram score and an elevated CCG status indicate the patient has an increased likelihood of recurrence.
  • results of any analyses according to the invention will often be communicated to physicians, genetic counselors and/or patients (or other interested parties such as researchers) in a transmittable form that can be communicated or transmitted to any of the above parties.
  • a transmittable form can vary and can be tangible or intangible.
  • the results can be embodied in descriptive statements, diagrams, photographs, charts, images or any other visual forms. For example, graphs showing expression or activity level or sequence variation information for various genes can be used in explaining the results. Diagrams showing such information for additional target gene(s) are also useful in indicating some testing results.
  • statements and visual forms can be recorded on a tangible medium such as papers, computer readable media such as floppy disks, compact disks, etc., or on an intangible medium, e.g., an electronic medium in the form of email or website on internet or intranet.
  • results can also be recorded in a sound form and transmitted through any suitable medium, e.g., analog or digital cable lines, fiber optic cables, etc., via telephone, facsimile, wireless mobile phone, internet phone and the like.
  • the information and data on a test result can be produced anywhere in the world and transmitted to a different location.
  • the information and data on a test result may be generated, cast in a transmittable form as described above, and then imported into the United States.
  • the present invention also encompasses a method for producing a transmittable form of information on at least one of (a) expression level or (b) activity level for at least one patient sample.
  • the method comprises the steps of (1) determining at least one of (a) or (b) above according to methods of the present invention; and (2) embodying the result of the determining step in a transmittable form.
  • the transmittable form is the product of such a method.
  • the present invention further provides a system for determining gene expression in a tumor sample, comprising: (1) a sample analyzer for determining the expression levels of a panel of genes in a tumor sample including at least 2, 4, 6, 8 or 10 cell-cycle genes, wherein the sample analyzer contains the tumor sample which is from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer, or cDNA molecules from mR A expressed from the panel of genes; (2) a first computer program means for (a) receiving gene expression data on at least 4 test genes selected from the panel of genes, (b) weighting the determined expression of each of the test genes, and (c) combining the weighted expression to provide a test value, wherein at least 20%, 50%, at least 75% or at least 90% of the test genes are cell-cycle genes; and optionally (3) a second computer program means for comparing the test value to one or more reference values each associated with a predetermined degree of risk of cancer recurrence or progression of the prostate cancer, lung cancer, bladder cancer or brain cancer.
  • the amount of R A transcribed from the panel of genes including test genes is measured in the tumor sample.
  • the amount of RNA of one or more housekeeping genes in the tumor sample is also measured, and used to normalize or calibrate the expression of the test genes, as described above.
  • the plurality of test genes includes at least 2, 3 or 4 cell-cycle genes, which constitute at least 50%, 75% or 80%> of the plurality of test genes, and preferably 100% of the plurality of test genes.
  • the plurality of test genes includes at least 5, 6 or 7, or at least 8 cell-cycle genes, which constitute at least 20%>, 25%, 30%>, 40%, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes.
  • the plurality of test genes includes at least 8, 10,
  • cell-cycle genes which constitute at least 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes.
  • the sample analyzer can be any instruments useful in determining gene expression, including, e.g., a sequencing machine, a real-time PCR machine, and a microarray instrument.
  • the computer-based analysis function can be implemented in any suitable language and/or browsers. For example, it may be implemented with C language and preferably using object-oriented high-level programming languages such as Visual Basic, SmallTalk, C++, and the like.
  • the application can be written to suit environments such as the Microsoft WindowsTM environment including WindowsTM 98, WindowsTM 2000, WindowsTM NT, and the like.
  • the application can also be written for the MacintoshTM, SUNTM, UNIX or LINUX environment.
  • the functional steps can also be implemented using a universal or platform-independent programming language.
  • multi-platform programming languages include, but are not limited to, hypertext markup language (HTML), JAVATM, JavaScriptTM, Flash programming language, common gateway interface/structured query language (CGI/SQL), practical extraction report language (PERL), AppleScriptTM and other system script languages, programming language/structured query language (PL/SQL), and the like.
  • Java - or JavaScript -enabled browsers such as HotJavaTM, MicrosoftTM ExplorerTM, or NetscapeTM can be used.
  • active content web pages may include JavaTM applets or ActiveXTM controls or other active content technologies.
  • the analysis function can also be embodied in computer program products and used in the systems described above or other computer- or internet-based systems. Accordingly, another aspect of the present invention relates to a computer program product comprising a computer-usable medium having computer-readable program codes or instructions embodied thereon for enabling a processor to carry out gene status analysis. These computer program instructions may be loaded onto a computer or other programmable apparatus to produce a machine, such that the instructions which execute on the computer or other programmable apparatus create means for implementing the functions or steps described above. These computer program
  • instructions may also be stored in a computer-readable memory or medium that can direct a computer or other programmable apparatus to function in a particular manner, such that the instructions stored in the computer-readable memory or medium produce an article of manufacture including instruction means which implement the analysis.
  • the computer program instructions may also be loaded onto a computer or other programmable apparatus to cause a series of operational steps to be performed on the computer or other programmable apparatus to produce a computer implemented process such that the instructions which execute on the computer or other
  • programmable apparatus provide steps for implementing the functions or steps described above.
  • the system comprises (1) computer program means for receiving, storing, and/or retrieving a patient's gene status data (e.g. , expression level, activity level, variants) and optionally clinical parameter data (e.g. , Gleason score, nomogram score); (2) computer program means for querying this patient data; (3) computer program means for concluding whether there is an increased likelihood of recurrence based on this patient data; and optionally (4) computer program means for outputting/displaying this conclusion.
  • this means for outputting the conclusion may comprise a computer program means for informing a health care professional of the conclusion.
  • Computer system [600] may include at least one input module [630] for entering patient data into the computer system [600] .
  • the computer system [600] may include at least one output module [624] for indicating whether a patient has an increased or decreased likelihood of response and/or indicating suggested treatments determined by the computer system
  • Computer system [600] may include at least one memory module [606] in communication with the at least one input module [630] and the at least one output module [624] .
  • the at least one memory module [606] may include, e.g., a removable storage drive [608], which can be in various forms, including but not limited to, a magnetic tape drive, a floppy disk drive, a VCD drive, a DVD drive, an optical disk drive, etc.
  • the removable storage drive [608] may be compatible with a removable storage unit [610] such that it can read from and/or write to the removable storage unit [610].
  • Removable storage unit [610] may include a computer usable storage medium having stored therein computer-readable program codes or instructions and/or computer readable data.
  • removable storage unit [610] may store patient data.
  • Example of removable storage unit [610] are well known in the art, including, but not limited to, floppy disks, magnetic tapes, optical disks, and the like.
  • the at least one memory module [606] may also include a hard disk drive [612], which can be used to store computer readable program codes or instructions, and/or computer readable data.
  • the at least one memory module [606] may further include an interface [614] and a removable storage unit [616] that is compatible with interface [614] such that software, computer readable codes or instructions can be transferred from the removable storage unit [616] into computer system [600] .
  • interface [614] and removable storage unit [616] pairs include, e.g., removable memory chips (e.g., EPROMs or PROMs) and sockets associated therewith, program cartridges and cartridge interface, and the like.
  • Computer system [600] may also include a secondary memory module [618], such as random access memory (RAM).
  • RAM random access memory
  • Computer system [600] may include at least one processor module [602] . It should be understood that the at least one processor module [602] may consist of any number of devices.
  • the at least one processor module [602] may include a data processing device, such as a microprocessor or microcontroller or a central processing unit.
  • the at least one processor module [602] may include another logic device such as a DMA (Direct Memory Access) processor, an integrated communication processor device, a custom VLSI (Very Large Scale Integration) device or an ASIC (Application Specific Integrated Circuit) device.
  • the at least one processor module [602] may include any other type of analog or digital circuitry that is designed to perform the processing functions described herein.
  • the at least one memory module [606], the at least one processor module [602], and secondary memory module [618] are all operably linked together through communication infrastructure [620] , which may be a
  • Input interface [626] may operably connect the at least one input module [626] to the communication infrastructure [620] .
  • output interface [622] may operably connect the at least one output module [624] to the communication infrastructure [620] .
  • the at least one input module [630] may include, for example, a keyboard, mouse, touch screen, scanner, and other input devices known in the art.
  • the at least one output module [624] may include, for example, a display screen, such as a computer monitor, TV monitor, or the touch screen of the at least one input module [630]; a printer; and audio speakers.
  • Computer system [600] may also include, modems, communication ports, network cards such as Ethernet cards, and newly developed devices for accessing intranets or the internet.
  • the at least one memory module [606] may be configured for storing patient data entered via the at least one input module [630] and processed via the at least one processor module [602] .
  • Patient data relevant to the present invention may include expression level, activity level, copy number and/or sequence information for PTEN and/or a CCG.
  • Patient data relevant to the present invention may also include clinical parameters relevant to the patient's disease. Any other patient data a physician might find useful in making treatment decisions/recommendations may also be entered into the system, including but not limited to age, gender, and race/ethnicity and lifestyle data such as diet information.
  • Other possible types of patient data include symptoms currently or previously experienced, patient's history of illnesses, medications, and medical procedures.
  • the at least one memory module [606] may include a computer-implemented method stored therein.
  • the at least one processor module [602] may be used to execute software or computer-readable instruction codes of the computer-implemented method.
  • the computer- implemented method may be configured to, based upon the patient data, indicate whether the patient has an increased likelihood of recurrence, progression or response to any particular treatment, generate a list of possible treatments, etc.
  • the computer-implemented method may be configured to identify a patient as having or not having an increased likelihood of recurrence or progression. For example, the computer-implemented method may be configured to inform a physician that a particular patient has an increased likelihood of recurrence. Alternatively or additionally, the computer-implemented method may be configured to actually suggest a particular course of treatment based on the answers to/results for various queries.
  • FIG.7 illustrates one embodiment of a computer-implemented method [700] of the invention that may be implemented with the computer system [600] of the invention.
  • the method [700] begins with one of three queries ([710], [711], [712]), either sequentially or substantially simultaneously. If the answer to/result for any of these queries is "Yes” [720], the method concludes [730] that the patient has an increased likelihood of recurrence. If the answer to/result for all of these queries is "No" [721], the method concludes [731] that the patient does not have an increased likelihood of recurrence. The method [700] may then proceed with more queries, make a particular treatment recommendation ([740], [741]), or simply end.
  • the queries When the queries are performed sequentially, they may be made in the order suggested by FIG.7 or in any other order. Whether subsequent queries are made can also be dependent on the results/answers for preceding queries.
  • the method asks about clinical parameters [712] first and, if the patient has one or more clinical parameters identifying the patient as at increased risk for recurrence then the method concludes such [730] or optionally confirms by querying CCG status, while if the patient has no such clinical parameters then the method proceeds to ask about CCG status [711] .
  • the method may continue to ask about PTEN status
  • [700] is open-ended.
  • the apparent first step [710, 711, and/or 712] in FIG.7 may actually form part of a larger process and, within this larger process, need not be the first step/query. Additional steps may also be added onto the core methods discussed above.
  • Additional steps include, but are not limited to, informing a health care professional (or the patient itself) of the conclusion reached; combining the conclusion reached by the illustrated method [700] with other facts or conclusions to reach some additional or refined conclusion regarding the patient's diagnosis, prognosis, treatment, etc.; making a recommendation for treatment (e.g., "patient should/should not undergo radical prostatectomy”); additional queries about additional biomarkers, clinical parameters, or other useful patient information (e.g., age at diagnosis, general patient health, etc.).
  • the answers to the queries may be determined by the method instituting a search of patient data for the answer.
  • patient data may be searched for PTEN status (e.g., PTEN IHC or mutation screening), CCG status (e.g., CCG expression level data), or clinical parameters (e.g., Gleason score, nomogram score, etc.). If such a comparison has not already been performed, the method may compare these data to some reference in order to determine if the patient has an abnormal (e.g., elevated, low, negative) status.
  • PTEN status e.g., PTEN IHC or mutation screening
  • CCG status e.g., CCG expression level data
  • clinical parameters e.g., Gleason score, nomogram score, etc.
  • the method may present one or more of the queries [710, 711, 712] to a user (e.g., a physician) of the computer system [100] .
  • the questions [710, 711, 712] may be presented via an output module [624] .
  • the user may then answer "Yes” or "No” via an input module [630] .
  • the method may then proceed based upon the answer received.
  • the conclusions [730, 731] may be presented to a user of the computer-implemented method via an output module [624] .
  • the invention provides a method comprising: accessing information on a patient's CCG status, clinical parameters and/or PTEN status stored in a computer-readable medium; querying this information to determine at least one of whether a sample obtained from the patient shows increased expression of at least one CCG, whether the patient has a recurrence-associated clinical parameter, and/or whether the patient has a low/negative PTEN status,; outputting [or displaying] the sample's CCG expression status, the patient's recurrence-associated clinical parameter status, and/or the sample's PTEN status.
  • "displaying" means communicating any information by any sensory means.
  • Examples include, but are not limited to, visual displays, e.g., on a computer screen or on a sheet of paper printed at the command of the computer, and auditory displays, e.g., computer generated or recorded auditory expression of a patient's genotype.
  • some embodiments provide a computer-implemented method of determining whether a patient has an increased likelihood of recurrence comprising accessing information on a patient's PTEN status ⁇ e.g., from a tumor sample obtained from the patient) or clinical parameters and CCG status ⁇ e.g., from a tumor sample obtained from the patient) stored in a computer-readable medium; querying this information to determine at least one of whether the patient has a
  • Some embodiments further comprise displaying PTEN, clinical parameters (or their values) and/or the CCGs and their status (including, e.g., expression levels), optionally together with an indication of whether the PTEN or CCG status and/or clinical parameter indicates increased likelihood of risk.
  • Computer software products of the invention typically include computer readable media having computer-executable instructions for performing the logic steps of the method of the invention.
  • Suitable computer readable medium include floppy disk, CD- ROM/DVD/DVD-ROM, hard-disk drive, flash memory, ROM/RAM, magnetic tapes and etc.
  • Basic computational biology methods are described in, for example, Setubal et ah, INTRODUCTION TO COMPUTATIONAL BIOLOGY METHODS (PWS Publishing Company, Boston, 1997); Salzberg et al.
  • BIOINFORMATICS A PRACTICAL GUIDE FOR COMPUTATIONAL METHODS IN MOLECULAR BIOLOGY, (Elsevier, Amsterdam, 1998); Rashidi & Buehler, BIOINFORMATICS BASICS : APPLICATION IN BIOLOGICAL SCIENCE AND MEDICINE (CRC Press, London, 2000); and Ouelette & Bzevanis, BIOINFORMATICS: A PRACTICAL GUIDE FOR
  • the present invention may also make use of various computer program products and software for a variety of purposes, such as probe design, management of data, analysis, and instrument operation. See U.S. Pat. Nos. 5,593,839; 5,795,716; 5,733,729; 5,974, 164;
  • the present invention may have embodiments that include methods for providing genetic information over networks such as the Internet as shown in U.S. Ser. Nos. 10/197,621 (U.S. Pub. No.
  • the invention provides a system for determining gene expression in a tumor sample, comprising:
  • sample analyzer for determining the expression levels in a sample of a panel of genes including at least 4 CCGs, wherein the sample analyzer contains the sample, RNA from the sample and expressed from the panel of genes, or DNA synthesized from said RNA;
  • At least 20%>, 50%>, 75%, or 90%> of said plurality of test genes are CCGs.
  • the sample analyzer contains reagents for determining the expression levels in the sample of said panel of genes including at least 4 CCGs.
  • the sample analyzer contains CCG-specific reagents as described below.
  • the invention provides a system for determining gene expression in a tumor sample, comprising: (1) a sample analyzer for determining the expression levels of a panel of genes in a tumor sample including at least 4 CCGs, wherein the sample analyzer contains the tumor sample which is from a patient identified as having prostate cancer, breast cancer, brain cancer, bladder cancer, or lung cancer, R A from the sample and expressed from the panel of genes, or DNA synthesized from said RNA; (2) a first computer program for (a) receiving gene expression data on at least 4 test genes selected from the panel of genes, (b) weighting the determined expression of each of the test genes with a predefined coefficient, and (c) combining the weighted expression to provide a test value, wherein the combined weight given to said at least 4 or 5 or 6 CCGs is at least 40% (or 50%, 60%, 70%, 80%, 90%, 95% or 100%) of the total weight given to the expression of all of said plurality of test genes; and optionally (3) a second computer program for comparing the test value
  • the system comprises a computer program for determining the patient's prognosis and/or determining (including quantifying) the patient's degree of risk of cancer recurrence or progression based at least in part on the comparison of the test value with said one or more reference values.
  • the system further comprises a display module displaying the comparison between the test value and the one or more reference values, or displaying a result of the comparing step, or displaying the patient's prognosis and/or degree of risk of cancer recurrence or progression.
  • the amount of RNA transcribed from the panel of genes including test genes (and/or DNA reverse transcribed therefrom) is measured in the sample.
  • the amount of RNA of one or more housekeeping genes in the sample (and/or DNA reverse transcribed therefrom) is also measured, and used to normalize or calibrate the expression of the test genes, as described above.
  • the plurality of test genes includes at least 2, 3 or 4
  • the plurality of test genes includes at least 5, 6 or 7, or at least 8 CCGs, which constitute at least 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 of the following genes: ASPM, BIRC5, BUB IB, CCNB2, CDC2, CDC20, CDCA8, CDKN3, CENPF, DLGAP5, FOXM1, KIAA0101, KIF11, KIF2C, KIF4A, MCM10, NUSAP1, PRC1, RACGAP1, and TPX2.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • ASPM BIRC5, BUB IB, CCNB2, CDC2, CDC20, CDCA8, CDKN3, CENPF, DLGAP5, FOXM1, KIAA0101, KIF11, KIF2C
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, or nine or all of gene numbers 2 & 3, 2 to
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4,
  • this plurality of CCGs comprises any one, two, three, four, five, six, seven, or eight or all of gene numbers 3 & 4, 3 to 5, 3 to 6, 3 to 7, 3 to 8, 3 to 9, or 3 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • CCGs e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs
  • this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25.
  • the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, 10, 11, 12, 13, 14, or 15 or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, 1 to 10, 1 to 11, 1 to 12, 1 to 13, 1 to 14, or 1 to 15 of any of Tables 9, 11, 23, 24, or 25. [00133] In some other embodiments, the plurality of test genes includes at least 8, 10,
  • CCGs which constitute at least 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%) or 90%) of the plurality of test genes, and preferably 100% of the plurality of test genes.
  • the sample analyzer can be any instrument useful in determining gene expression, including, e.g., a sequencing machine (e.g., Illumina HiSeqTM, Ion Torrent PGM, ABI SOLiDTM sequencer, PacBio RS, Helicos HeliscopeTM, etc.), a real-time PCR machine (e.g., ABI 7900, Fluidigm BioMarkTM, etc.), a microarray instrument, etc.
  • a sequencing machine e.g., Illumina HiSeqTM, Ion Torrent PGM, ABI SOLiDTM sequencer, PacBio RS, Helicos HeliscopeTM, etc.
  • a real-time PCR machine e.g., ABI 7900, Fluidigm BioMarkTM, etc.
  • microarray instrument e.g., a microarray instrument, etc.
  • the present invention provides methods of treating a cancer patient comprising obtaining CCG status information (e.g., the CCGs in Table 1 or Panels A through G), and recommending, prescribing or administering a treatment for the cancer patient based on the CCG status.
  • the method further includes obtaining clinical parameter information, and/or obtaining PTEN status information from a sample from the patient and treating the patient with a particular treatment based on the CCG status, clinical parameter and/or PTEN status information.
  • the invention provides a method of treating a cancer patient comprising:
  • Whether a treatment is aggressive or not will generally depend on the cancer- type, the age of the patient, etc.
  • adjuvant chemotherapy is a common aggressive treatment given to complement the less aggressive standards of surgery and hormonal therapy.
  • Those skilled in the art are familiar with various other aggressive and less aggressive treatments for each type of cancer.
  • Active treatment in prostate cancer is well-understood by those skilled in the art and, as used herein, has the conventional meaning in the art. Generally speaking, active treatment in prostate cancer is anything other than "watchful waiting.” Active treatment currently applied in the art of prostate cancer treatment includes, e.g.
  • Watchful-waiting is sometimes used, e.g. , when an early stage, slow-growing prostate cancer is found in an older man. Watchful-waiting may also be suggested when the risks of surgery, radiation therapy, or hormonal therapy outweigh the possible benefits. Other treatments can be started if symptoms develop, or if there are signs that the cancer growth is accelerating (e.g., rapidly rising PSA, increase in Gleason score on repeat biopsy, etc.).
  • watchful-waiting carries its own risks, e.g., increased risk of metastasis.
  • a trial of active surveillance may not mean avoiding treatment altogether, but may reasonably allow a delay of a few years or more, during which time the quality of life impact of active treatment can be avoided.
  • Published data to date suggest that carefully selected men will not miss a window for cure with this approach. Additional health problems that develop with advancing age during the observation period can also make it harder to undergo surgery and radiation therapy. Thus it is clinically important to carefully determine which prostate cancer patients are good candidates for watchful-waiting and which patients should receive active treatment.
  • the invention provides a method of treating a prostate cancer patient or providing guidance to the treatment of a patient.
  • the status of at least one CCG e.g., those in Table 1 or Panels A through G
  • at least one recurrence-associated clinical parameter, and/or the status of PTEN is determined, and (a) active treatment is recommended, initiated or continued if a sample from the patient has an elevated status for at least one CCG, the patient has at least one recurrence-associated clinical parameter, and/or low/negative PTEN status, or (b) watchful-waiting is recommended/initiated/ continued if the patient has neither an elevated status for at least one CCG, a recurrence-associated clinical parameter, nor low/negative PTEN status.
  • CCG e.g., those in Table 1 or Panels A through G
  • active treatment is recommended, initiated or continued if a sample from the patient has an elevated status for at least one CCG, the patient has at least one recurrence-associated clinical parameter, and/
  • CCG status, the clinical parameter(s) and PTEN status may indicate not just that active treatment is recommended, but that a particular active treatment is preferable for the patient (including relatively aggressive treatments such as, e.g., RP and/or adjuvant therapy).
  • the invention provides a method of treating a patient ⁇ e.g., a prostate cancer patient) comprising determining the status of at least one CCG ⁇ e.g.
  • the status of at least one recurrence-associated clinical parameter, and/or the status of PTEN and initiating adjuvant therapy after prostatectomy or radiotherapy if a sample from the patient has an elevated status for at least one CCG, the patient has at least one recurrence- associated clinical parameter and/or the patient has low/negative PTEN status.
  • the invention provides compositions for use in the above methods.
  • Such compositions include, but are not limited to, nucleic acid probes hybridizing to PTEN or a CCG (or to any nucleic acids encoded thereby or complementary thereto); nucleic acid primers and primer pairs suitable for amplifying all or a portion of PTEN or a CCG or any nucleic acids encoded thereby; antibodies binding immunologically to a polypeptide encoded by PTEN or a CCG; probe sets comprising a plurality of said nucleic acid probes, nucleic acid primers, antibodies, and/or polypeptides; microarrays comprising any of these; kits comprising any of these; etc.
  • the invention provides computer methods, systems, software and/or modules for use in the above methods.
  • the invention provides a probe comprising an isolated oligonucleotide capable of selectively hybridizing to PTEN or at least one of the genes in Table 1 or Panels A through G.
  • probe and “oligonucleotide” (also “oligo"), when used in the context of nucleic acids, interchangeably refer to a relatively short nucleic acid fragment or sequence.
  • primers useful in the methods of the invention. “Primers” are probes capable, under the right conditions and with the right companion reagents, of selectively amplifying a target nucleic acid ⁇ e.g., a target gene).
  • probe is used herein to encompass “primer” since primers can generally also serve as probes.
  • the probe can generally be of any suitable size/length. In some embodiments the probe has a length from about 8 to 200, 15 to 150, 15 to 100, 15 to 75, 15 to 60, or 20 to 55 bases in length. They can be labeled with detectable markers with any suitable detection marker including but not limited to, radioactive isotopes, fluorophores, biotin, enzymes (e.g., alkaline phosphatase), enzyme substrates, ligands and antibodies, etc. See Jablonski et al., NUCLEIC ACIDS RES.
  • probes may be modified in any conventional manner for various molecular biological applications. Techniques for producing and using such oligonucleotide probes are conventional in the art.
  • Probes according to the invention can be used in the
  • some embodiments of the invention comprise probe sets suitable for use in a microarray in detecting, amplifying and/or quantitating PTEN and/or a plurality of CCGs.
  • the probe sets have a certain proportion of their probes directed to CCGs— e.g., a probe set consisting of 10%, 20%, 30%>, 40%>, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% probes specific for CCGs.
  • the probe set comprises probes directed to at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 250, 300, 350, 400, 450, 500, 600, 700, or 800 or more, or all, of the genes in Table 1 or Panels A through G.
  • probe sets can be incorporated into high-density arrays comprising 5,000, 10,000, 20,000, 50,000, 100,000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, or 1 ,000,000 or more different probes.
  • the probe sets comprise primers (e.g., primer pairs) for amplifying nucleic acids comprising at least a portion of PTEN ox of one or more of the CCGs in Table 1 or Panels A through G.
  • kits for practicing the prognosis of the present invention.
  • the kit may include a carrier for the various components of the kit.
  • the carrier can be a container or support, in the form of, e.g. , bag, box, tube, rack, and is optionally compartmentalized.
  • the carrier may define an enclosed confinement for safety purposes during shipment and storage.
  • the kit includes various components useful in determining the status of one or more CCGs and one or more housekeeping gene markers, using the above-discussed detection techniques.
  • the kit many include oligonucleotides specifically hybridizing under high stringency to mR A or cDNA of the genes in Table 1 or Panels A through G.
  • kits comprises reagents (e.g., probes, primers, and or antibodies) for determining the expression level of a panel of genes, where said panel comprises at least 25%, 30%>, 40%>, 50%>, 60%, 75%, 80%, 90%, 95%, 99%, or 100% CCGs (e.g., CCGs in Table 1 or any of Panels A through G).
  • reagents e.g., probes, primers, and or antibodies
  • CCGs e.g., CCGs in Table 1 or any of Panels A through G.
  • the kit consists of reagents (e.g., probes, primers, and or antibodies) for determining the expression level of no more than 2500 genes, wherein at least 5, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 150, 200, 250, or more of these genes are CCGs (e.g., CCGs in Table 1 or any of Panels A through G).
  • reagents e.g., probes, primers, and or antibodies
  • the oligonucleotides in the detection kit can be labeled with any suitable detection marker including but not limited to, radioactive isotopes, fluorephores, biotin, enzymes (e.g., alkaline phosphatase), enzyme substrates, ligands and antibodies, etc. See Jablonski et al., Nucleic Acids Res., 14:6115-6128 (1986); Nguyen et al, Biotechniques, 13: 116-123 (1992); Rigby et al., J. Mol. Biol, 113:237-251 (1977).
  • the oligonucleotides included in the kit are not labeled, and instead, one or more markers are provided in the kit so that users may label the oligonucleotides at the time of use.
  • the detection kit contains one or more antibodies selectively immunoreactive with one or more proteins encoded by PTEN or one or more CCGs or optionally any additional markers. Examples include antibodies that bind
  • the detection kit of this invention may also be included in the detection kit of this invention.
  • examples of such components include, but are not limited to, Taq polymerase, deoxyribonucleotides, dideoxyribonucleotides, other primers suitable for the amplification of a target DNA sequence, RNase A, and the like.
  • the detection kit preferably includes instructions on using the kit for practice the prognosis method of the present invention using human samples.
  • EXAMPLE 1 [00150] The following cell cycle gene (CCG) signature was tested for predicting time to chemical recurrence after radical prostatectomy.
  • the mean of CCG expression is robust to measurement error and individual variation between genes.
  • the predictive power of the mean was tested for randomly selected sets of from 1 to 30 of the CCGs listed above. This simulation showed that there is a threshold number of CCGs in a panel that provides significantly improved predictive power.
  • Predictive power of the CCG signature after accounting for clinical variables typically included in a post-surgical nomogram was also evaluated.
  • the nomogram was a highly significant predictor of recurrence (p-value 1.6xl0 "10 ).
  • the CCG signature was a significant predictor of biochemical recurrence (FIG.3) in the discovery cohort (p-value 0.03) and in the clinical validation cohort (p- value 4.8 x 10 ⁇ 5 ).
  • Somatic mutations in PTEN were found to be significantly associated with recurrence, and importantly, it added prognostic information beyond both the established clinical nomogram for prostate cancer recurrence (the Kattan-Stephenson nomogram) and the CCG signature score (described in Examples 1 & 2, supra).
  • FFPE slides were first stained with hematoxylin and eosin and examined by a pathologist to identify the tumor region. After deparaffinization, tumor tissue was cut out from the slides by a razor blade. For a few samples dissection was aided by laser capture microscopy (LCM), owing to the dispersion of the tumor cells
  • PTEN genomic sequence The primers contained Ml 3 forward and reverse tails to facilitate sequencing.
  • DNA sequence was determined on a Mega BASE 4500 (GE healthcare) using dye -primer chemistry as described in Frank et al., J. CLIN. ONCOL. (2002) 20: 1480- 1490. Due to the technical difficulties associated with sequencing DNA derived from FFPE material, each mutation was detected by at least two independent amplification and sequencing reactions.
  • Table 12 List of 15 housekeeping (HK) genes
  • RNA Isolated total RNA was treated with DNase I (Sigma) prior to cDNA synthesis. Subsequently, we employed the High- capacity cDNA Archive Kit (Applied Biosystems) to convert total RNA into single strand cDNA as described by the manufacturer. A minimum of 200ng RNA was required for the RT reaction.
  • the cDNA Prior to measuring expression levels, the cDNA was pre-amplified with a pooled reaction containing TaqManTM assays. Pre-amplification reaction conditions were: 14 cycles of 95°C for 15sec and 60°C for 4 minutes. The first cycle was modified to include a 10 minute incubation at 95°C. The amplification reaction was diluted 1 :20 using the IX TE buffer prior to loading on TaqManTM Low Density Arrays (TLDA, Applied Biosystems) to measure gene expression.
  • TLDA TaqManTM Low Density Arrays
  • the CCG score is calculated from RNA expression of 31 CCGs (Panel F) normalized by 15 housekeeper genes (HK).
  • the relative numbers of CCGs (31) and HK genes (15) were optimized in order to minimize the variance of the CCG score.
  • the CCG score is the unweighted mean of CT values for CCG expression, normalized by the unweighted mean of the HK genes so that higher values indicate higher expression.
  • One unit is equivalent to a two-fold change in expression. Missing values were imputed using the mean expression for each gene determined in the training set using only good quality samples.
  • the CCG scores were centered by the mean value, again determined in the training set.
  • the CCG score threshold for determining low-risk was based on the lowest
  • a Cox proportional hazards model was used to summarize the available clinical parameter data and estimate the prior clinical risk of biochemical recurrence for each patient.
  • the data set consisted of 195 cases from the training set and 248 other cases with clinical parameter information but insufficient sample to measure R A expression. Univariate tests were performed on clinical parameters known to be associated with outcome (see Table 13 below). Non-significant parameters were excluded from the model.
  • a composite variable was created for organ-confined disease, with invasion defined as surgical margins, extracapsular extension, or involvement of any of seminal vesicles, bladder neck/urethral margins, or lymph nodes. The composite variable for organ- confined disease proved more significant in the model than any of its five components, some of which were inter-correlated or not prevalent. Model fitting was performed using the AIC criteria for post-operative covariates.
  • the final model (i.e., nomogram) has binary variables for organ-confined disease and Gleason score less than or equal to 6, and a continuous variable for logarithmic PSA (Table 14).
  • This model includes all of the clinical parameters incorporated in the post-RP nomogram (i.e., Kattan- Stephenson nomogram) except for Year of RP and the two components of the Gleason score.
  • the distribution of prior clinical risk shows three distinct nodes ( Figure 8). K- means clustering with 3 centers was used to set the threshold for the low-risk cluster, which comprises approximately 50% of the sample.
  • Kaplan-Meier plots are used to show estimated survival probabilities for subsets of patients; however, p-values are from the Cox likelihood ratio test for the continuous values of the variable. All statistical analyses were performed in S+ Version 8.1.1 for Linux (TIBCO Spotfire) or R 2.9.0 (http://www.r-project.org).
  • RNA from FFPE tumor sections derived from 442 prostate cancer patients treated with RP was split into 195 patients for initial characterization of the signature ("training set") and 247 patients for validation.
  • the clinical parameters of the training and validation cohort are listed in Table 15. There were no significant differences after adjusting for multiple comparisons.
  • Table 15 Clinical parameters of training and validation patient cohorts
  • the distribution of the scores from the clinical model contained several modes ( Figure 8), separating high- and low-risk patient groups. Therefore, the score was used subsequently as a binary variable (high or low risk).
  • CCGs cell cycle genes
  • the signature is immediately useful for defining the risk of patients who present with low-risk clinical parameters.
  • low-risk we essentially defined low-risk as Gleason ⁇ 7, PSA ⁇ 10 and organ-confined disease.
  • the CCG signature score effectively subdivides the low-risk group into patients with very low recurrence rates (5%), and a higher risk of recurrence (22%) (Figure 9 & Table 18). This is the most dramatic effect of the molecular signature - accurately redefining the risk of patients previously defined as low-risk based on clinical parameters. It is noteworthy that within this patient subpopulation (i.e., patients defined as low-risk based on clinical parameters) clinical parameters are not particularly prognostic (see Table 17).
  • the signature could be useful for a large number of patients.
  • nearly 60% of the cohort was characterized as low-risk and 40% of those are expected to have low CCG scores. Therefore, the CCG signature can predict indolent disease in a quarter of the patients who have previously been identified as high-risk (and therefore identified as candidates for radical prostatectomy).
  • the validation data in particular suggests that the CCG signature may be useful for defining risk in all patients. Specifically, it helped to divide patients defined as high-risk according to clinical parameters into those with 30% and 70% recurrence rates (Table 18).
  • GSE8894 (Son et al; no publication); Shedden (Shedden et al, NATURE MED. (2008) 14:822; PMID 18641660); GSE4412 (Freije et al, CANCER RES. (2004) 64:6503-10; PMID 15374961); GSE4271 (Phillips et al, CANCER CELL (2006) 9: 157-73; PMID 16530701); GSE5287 (Als et al, CLIN. CANCER RES. (2007) 13:4407-14; PMID 17671123).
  • Each of these datasets has an associated detailed description of the experimental procedures used in gathering expression and patient data.
  • the expression microarrays used to generate each dataset are summarized below in Table 20.
  • CCG score is an average expression of the genes in a panel. If a gene is represented by more than one probe set on the array, the gene expression is an average expression of all the probe sets representing the gene. The association between CCG score and survival or disease recurrence was tested using univariate and multivariate Cox proportional hazard model.
  • Multivariate analysis was performed when relevant clinical parameters (grade in brain cancer, stage in lung cancer, NPI in breast cancer) were available.
  • Brain cancer GSE4412 3.2xl0 ⁇ 5 2.2xl0 ⁇ 5 9.0xl0 ⁇ 5
  • Brain cancer GSE4271 1.3xl0 ⁇ 3 l .OxlO "3 2.8xl0 ⁇ 4
  • each Panel was also prognostic in multivariate analysis when combined with at least one clinical parameter (or nomogram).
  • Tables 23 & 24 below provide rankings of select CCGs according to their correlation with the mean CCG expression.
  • Table 25 provides a ranking of the CCGs in Panel F according to their relative predictive value in Example 5 (analogous to Table 9).
  • Table 1 below provides a large, but not exhaustive, list of CCGs.
  • TOP2A topoisomerase (DNA) II alpha (170kD) Hs.156346 AA504348
  • CENPE centromere protein E (312kD) Hs.75573 AA402431
  • CENP-E putative kinetochore motor that accumulates just befo
  • TOP2A topoisomerase (DNA) II alpha (170kD) Hs.156346 AA026682
  • KPNA2 karyopherin alpha 2 (RAG cohort 1, importin alpha 1) Hs.159557 AA676460
  • FLJ10468 hypothetical protein FLJ10468 Hs.48855 N63744
  • CCNF cyclin F Hs.1973 AA676797
  • DKFZp762E1312 hypothetical protein DKFZp762E1312 Hs.104859 T66935
  • BUB1 budding uninhibited by benzimidazoles 1 (yeast homolog) Hs.98658
  • TOP2A **topoisomerase (DNA) II alpha (170kD) Hs.156346 AI734240
  • CKS2 CDC2-Associated Protein
  • ARL6IP ADP-ribosylation factor-like 6 interacting protein Hs.75249 H20558
  • L2DTL L2DTL protein Hs.126774 R06900
  • E2-EPF ubiquitin carrier protein Hs.174070 AA464019
  • UBCH10 ubiquitin carrier protein E2-C Hs.93002 R80790
  • KNSL5 kinesin-like 5 (mitotic kinesin-like protein 1) Hs.270845 AA452513 Mitotic kinesin-like protein- 1
  • CENPF centromere protein F (350/400kD, mitosin) Hs.77204 AA701455
  • CCNA2 cyclin A2 Hs.85137 AA608568 Cyclin A
  • HMMR **hyaluronan-mediated motility receptor (RHAMM) Hs.72550 AA171715
  • KIAA0008 gene product Hs.77695 AA262211
  • HSPC145 HSPC145 protein Hs.18349 R22949
  • FLJ20510 hypothetical protein FLJ20510 Hs.6844 N53214
  • HSPC216 hypothetical protein Hs.13525 T87341
  • P37NB 37 kDa leucine-rich repeat (LRR) protein Hs.155545 AA423870
  • CCNE1 cyclin El Hs.9700 T54121
  • FLJ11252 hypothetical protein FLJ11252 Hs.23495 N30185 LOC51203: clone HQ0310 PRO0310pl Hs.279905 AA620485
  • FLJ10491 hypothetical protein FL J 10491 Hs.274283 AA425404
  • K SL1 kinesin-like 1 Hs.8878 AA504625
  • CENPA centromere protein A (17kD) Hs.1594 AI369629
  • CDC6 cell division cycle 6, S. cerevisiae homolog Hs.69563 H59203
  • TSN translin Hs.75066 AA460927
  • KRNA2 karyopherin alpha 2 (RAG cohort 1, importin alpha 1) Hs.159557 AA489087
  • RRM2 ribonucleotide reductase M2 polypeptide Hs.75319 AA187351
  • CCNB1 cyclin Bl Hs.23960 R25788
  • C20ORF1 chromosome 20 open reading frame 1 Hs.9329 AA936183
  • TACC3 transforming, acidic coiled-coil containing protein 3 Hs.l 04019 AA279990 JkRl mRNA downregulated upon T-cell activation
  • E2F1 E2F transcription factor 1 Hs.96055 H61303
  • BUB IB budding uninhibited by benzimidazoles 1 (yeast homolog), beta Hs.36708 AA488324
  • KIAA0074 KIAA0074 protein Hs. l 192 N54344
  • MPHOSPH1 M-phase phosphoprotein 1 Hs.240 AA282935
  • ANLN anillin (Drosophila Scraps homolog), actin binding protein Hs.62180 R12261
  • BIRC5 baculoviral IAP repeat-containing 5 (survivin) Hs.1578 AA460685
  • PTTG1 pituitary tumor-transforming 1 Hs.252587 AA430032
  • KIAA0159 chromosome condensation-related SMC-associated protein 1 Hs.5719 AA668256
  • HMMR hyaluronan-mediated motility receptor
  • DKFZp762E1312 hypothetical protein DKFZp762E1312 Hs.104859 AA936181
  • CKAP2 cytoskeleton associated protein 2 Hs.24641 T52152
  • RAMP RA-regulated nuclear matrix-associated protein
  • FLJ22624 hypothetical protein FLJ22624 Hs.166425 AA488791
  • NEK2 NIMA (never in mitosis gene a)-related kinase 2 Hs.153704 W93379 72
  • MKI67 antigen identified by monoclonal antibody Ki-67
  • TTK TTK protein kinase Hs.169840 AI337292
  • VEGFC vascular endothelial growth factor C Hs.79141 H07899 vascular endothelial
  • VRP growth factor related protein
  • CDKN3 cyclin-dependent kinase inhibitor 3 (CDK2-associated dual specificity
  • FLJ23311 hypothetical protein FLJ23311 Hs.94292 N73916
  • ADH4 alcohol dehydrogenase 4 (class II), pi polypeptide Hs.1219 AA007395
  • E2F1 E2F transcription factor 1 Hs.96055 AA424949
  • CDKN2C cyclin-dependent kinase inhibitor 2C (pl8, inhibits CDK4) Hs.4854
  • N72115 pl8-INK6 Cyclin-dependent kinase 6 inhibitor
  • MCM4 minichromosome maintenance deficient (S. cerevisiae) 4 Hs.154443
  • PMSCL1 ⁇ polymyositis/scleroderma autoantigen 1 (75kD) Hs.91728 AA458994
  • Ki-67 antigen identified by monoclonal antibody Ki-67 Hs.80976 AA425973 Ki67
  • H2AFX H2A histone family, member X Hs.147097 H95392
  • FLJ20333 hypothetical protein FLJ20333 Hs.79828 AA147792
  • MCM5 minichromosome maintenance deficient (S. cerevisiae) 5 (cell division cycle
  • CDKN1B cyclin-dependent kinase inhibitor IB (p27, Kipl) Hs.238990 AA630082
  • NEK2 NIMA (never in mitosis gene a)-related kinase 2 Hs.153704 AA682321
  • RAD51 S. cerevisiae homolog (E coli RecA homolog) Hs.23044 N70010
  • HEC highly expressed in cancer, rich in leucine heptad repeats Hs.58169 W72679
  • TROAP trophinin associated protein (tastin) Hs.171955 H94949
  • FEN1 flap structure-specific endonuclease 1 Hs.4756 AA620553
  • CASP8AP2 CASP8 associated protein 2 Hs.122843 H50582
  • CKAP2 cytoskeleton associated protein 2 Hs.24641 AA504130
  • HLA-DRA major histocompatibility complex, class II, DR alpha Hs.76807 R47979
  • HBP Hairpin binding protein, histone Hs.75257 AA629558
  • CASP3 caspase 3, apoptosis-related cysteine protease Hs.74552 R14760 CASPASE-
  • HMG2 high-mobility group (nonhistone chromosomal) protein 2 Hs.80684
  • PRO2000 PRO2000 protein Hs.46677 H58234
  • TIMP1 tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity
  • FLJ10858 hypothetical protein FLJ10858 Hs.134403 AA677552
  • RFC4 replication factor C (activator 1) 4 (37kD) Hs.35120 N93924 replication factor
  • PRO2000 PRO2000 protein Hs.46677 N47113
  • ECT2 epithelial cell transforming sequence 2 oncogene Hs.132808 AI031571
  • PCF11 PCF1 lp homolog Hs.123654 AA053411
  • DKFZP566C134 DKFZP566C134 protein Hs.20237 N39306
  • ORCIL origin recognition complex, subunit 1 (yeast homolog)-like Hs.17908 H51719
  • HMG4 high-mobility group (nonhistone chromosomal) protein 4 Hs.19114
  • TSC22 transforming growth factor beta-stimulated protein TSC-22 Hs. l 14360
  • FLJ23293 ** hypothetical protein FLJ23293 similar to ARL-6 interacting protein-2
  • H 11 protein kinase H 11 ; small stress protein- like protein HSP22 Hs.111676
  • SERPINB3 serine (or cysteine) proteinase inhibitor, clade B (ovalbumin), member 3
  • DNAJB1 DnaJ (Hsp40) homolog, subfamily B, member 1 Hs.82646 AA435948
  • BUB3 BUB3 (budding uninhibited by benzimidazoles 3, yeast) homolog Hs.40323
  • SNC73 Homo sapiens SNC73 protein (SNC73) mRNA, complete cds Hs.293441 H28469:
  • BUB3 BUB3 (budding uninhibited by benzimidazoles 3, yeast) homolog Hs.40323
  • FLJ11186 hypothetical protein FLJ11186 Hs.89278 AA394225
  • TOPK PDZ-binding kinase
  • ZNF265 zinc finger protein 265 Hs.194718 AA452256
  • NS1-BP NSl-binding protein Hs.197298 AA486796
  • BIRC3 baculoviral IAP repeat-containing 3 Hs.127799 AA002125 c-
  • C TNFR2-TRAF signalling complex prot
  • MCM6 minichromosome maintenance deficient (mis5, S. pombe) 6 Hs.155462
  • NPAT nuclear protein, ataxia-telangiectasia locus Hs.89385 AA284172
  • 225 KNSL6 kinesin-like 6 (mitotic centromere-associated kinesin) Hs.69360 AA400450
  • CTNNA1 **catenin (cadherin-associated protein), alpha 1 (102kD) Hs.178452
  • SMC4L1 structural maintenance of chromosomes 4, yeast)-like 1 Hs.50758
  • ICBP90 transcription factor Hs.108106 AA026356 235
  • EXOl exonuclease 1 Hs.47504 AA703000
  • FLJ11269 hypothetical protein FLJ11269 Hs.25245 R37817
  • SFPQ splicing factor proline/glutamine rich (polypyrimidine tract-binding protein-
  • BTEB1 basic transcription element binding protein 1 Hs.150557 N80235
  • D21S2056E DNA segment on chromosome 21 (unique) 2056 expressed sequence
  • HDAC3 histone deacetylase 3 Hs.279789 H88540
  • RAD51C RAD51 (S. cerevisiae) homolog C Hs.l 1393 R37145
  • RAD5 lC Recombination/repair Rad51 -related protein
  • MGC5528 hypothetical protein MGC5528 Hs.315167 AA934904
  • CXCR4 chemokine (C-X-C motif), receptor 4 (fusin) Hs.89414 T62491 CXC
  • MSH2 **mutS (E. coli) homolog 2 (colon cancer, nonpolyposis type 1) Hs.78934
  • DDXl 1 DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 11 (S.cerevisiae CHL1-
  • ARGBP2 Arg/Abl-interacting protein ArgBP2 Hs.278626 N89738
  • DKFZP564A122 DKFZP564A122 protein Hs.187991 AA025807

Abstract

Biomarkers and methods using the biomarkers for the prediction of the recurrence risk of cancer in a patient are provided.

Description

GENE SIGNATURES FOR CANCER PROGNOSIS
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the priority benefit of U.S. Provisional Application
Serial No. 61/362,209 (filed on July 7, 2010), which is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
[0002] The invention generally relates to a molecular classification of disease and particularly to molecular markers for cancer prognosis and methods of use thereof.
BACKGROUND OF THE INVENTION
[0003] Cancer is a major public health problem, accounting for roughly 25% of all deaths in the United States. Though many treatments have been devised for various cancers, these treatments often vary in severity of side effects. It is useful for clinicians to know how aggressive a patient's cancer is in order to determine how aggressively to treat the cancer.
[0004] For example, most patients with early-stage asymptomatic prostate cancer are treated with radical prostatectomy or radiotherapy and optionally adjuvant therapy (e.g. , hormone or chemotherapy), all of which have severe side effects. For many of these patients, however, these treatments and their associated side effects and costs are unnecessary because the cancer in these patients is not aggressive (i.e., grows slowly and is unlikely to cause mortality or significant morbidity during the patient's lifetime). In other patients the cancer is virulent (i.e., more likely to recur) and aggressive treatment is necessary to save the patient's life.
[0005] Some tools have been devised to help physicians in deciding which patients need aggressive treatment and which do not. In fact, several clinical parameters are currently in use for this purpose in various different cancers. In prostate cancer, for example, such clinical parameters include serum prostate-specific antigen (PSA), Gleason grade, pathologic stage, and surgical margins. In recent years clinical parameters have been made more helpful through their incorporation into continuous multivariable postoperative nomograms that calculate a patient's probability of having cancer progression/recurrence. See, e.g., Kattan et ah, J. CLIN. ONCOL. (1999) 17: 1499-1507; Stephenson et al, J. CLIN. ONCOL. (2005) 23:7005-7012. Despite these advances, however, many patients are given improper cancer treatments and there is still a serious need for novel and improved tools for predicting cancer recurrence.
SUMMARY OF THE INVENTION
[0006] The present invention is based in part on the surprising discovery that the expression of those genes whose expression closely tracks the cell cycle ("cell-cycle genes" or "CCGs" as further defined below) is particularly useful in classifying selected types of cancer and determining the prognosis of these cancers.
[0007] Accordingly, in a first aspect of the present invention, a method is provided for determining gene expression in a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer. Generally, the method includes at least the following steps: (1) obtaining a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer; (2) determining the expression of a panel of genes in said tumor sample including at least 4 cell-cycle genes; and (3) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide said test value, wherein at least 50%, at least 75% or at least 90% of said plurality of test genes are cell-cycle genes.
[0008] In preferred embodiments, the plurality of test genes includes at least 8 cell- cycle genes, or at least 10, 15, 20, 25 or 30 cell-cycle genes. Preferably, all of the test genes are cell- cycle genes.
[0009] Also in preferred embodiments, the step of determining the expression of the panel of genes in the tumor sample comprises measuring the amount of mRNA in the tumor sample transcribed from each of from 4 to about 200 cell-cycle genes; and measuring the amount of mRNA of one or more housekeeping genes in the tumor sample. [0010] In another aspect of the present invention, a method is provided for determining the prognosis of prostate cancer, lung cancer, bladder cancer or brain cancer, which comprises determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of at least 6, 8 or 10 cell-cycle genes, wherein overexpression of said at least 6, 8 or 10 cell-cycle genes indicates a poor prognosis or an increased likelihood of recurrence of cancer in the patient.
[0011] In one embodiment, the prognosis method comprises (1) determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of a panel of genes in said tumor sample including at least 4 or at least 8 cell- cycle genes; and (2) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from the panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide the test value, wherein at least 50%, at least 75% or at least 85% of the plurality of test genes are cell-cycle genes, and wherein an increased level of overall expression of the plurality of test genes indicates a poor prognosis, whereas if there is no increase in the overall expression of the test genes, it would indicate a good prognosis or a low likelihood of recurrence of cancer in the patient.
[0012] In preferred embodiments, the prognosis method further includes a step of comparing the test value provided in step (2) above to one or more reference values, and correlating the test value to a risk of cancer progression or risk of cancer recurrence. Optionally an increased likelihood of poor prognosis is indicated if the test value is greater than the reference value.
[0013] In yet another aspect, the present invention also provide a method of treating cancer in a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer, comprising: (1) determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of a panel of genes in the tumor sample including at least 4 or at least 8 cell-cycle genes; (2) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide said test value, wherein at least 50% or 75% or 85% of the plurality of test genes are cell-cycle genes, wherein an increased level of expression of the plurality of test genes indicates a poor prognosis, and an un- increased level of expression of the plurality of test genes indicates a good prognosis; and recommending, prescribing or administering a treatment regimen or watchful waiting based on the prognosis provided in step (2).
[0014] The present invention further provides a diagnostic kit for prognosing cancer in a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, comprising, in a compartmentalized container, a plurality of oligonucleotides hybridizing to at least 8 test genes, wherein less than 10%, 30% or less than 40% of all of the at least 8 test genes are non-cell-cycle genes; and one or more oligonucleotide hybridizing to at least one housekeeping gene. The oligonucleotides can be hybridizing probes for hybridization with the test genes under stringent conditions or primers suitable for PCR amplification of the test genes. In one embodiment, the kit consists essentially of, in a compartmentalized container, a first plurality of PCR reaction mixtures for PCR amplification of from 5 or 10 to about 300 test genes, wherein at least 50%, at least 60% or at least 80% of such test genes are cell-cycle genes, and wherein each reaction mixture comprises a PCR primer pair for PCR amplifying one of the test genes; and a second plurality of PCR reaction mixtures for PCR amplification of at least one housekeeping gene.
[0015] The present invention also provides the use of (1) a plurality of
oligonucleotides hybridizing to at least 4 or at least 8 cell-cycle genes; and (2) one or more oligonucleotides hybridizing to at least one housekeeping gene, for the manufacture of a diagnostic product for determining the expression of the test genes in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, to predict the prognosis of cancer, wherein an increased level of the overall expression of the test genes indicates a poor prognosis or an increased likelihood of recurrence of cancer in the patient, whereas if there is no increase in the overall expression of the test genes, it would indicate a good prognosis or a low likelihood of recurrence of cancer in the patient. In some embodiments, the oligonucleotides are PCR primers suitable for PCR amplification of the test genes. In other embodiments, the oligonucleotides are probes hybridizing to the test genes under stringent conditions. In some embodiments, the plurality of oligonucleotides are probes for hybridization under stringent conditions to, or are suitable for PCR amplification of, from 4 to about 300 test genes, at least 50%, 70% or 80% or 90% of the test genes being cell-cycle genes. In some other embodiments, the plurality of oligonucleotides are hybridization probes for, or are suitable for PCR amplification of, from 20 to about 300 test genes, at least 30%, 40%, 50%, 70% or 80% or 90% of the test genes being cell-cycle genes. [0016] The present invention further provides a system for determining gene expression in a tumor sample, comprising: (1) a sample analyzer for determining the expression levels of a panel of genes in a tumor sample including at least 4 cell-cycle genes, wherein the sample analyzer contains the tumor sample which is from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer, or cDNA molecules from mRNA expressed from the panel of genes; (2) a first computer program means for (a) receiving gene expression data on at least 4 test genes selected from the panel of genes, (b) weighting the determined expression of each of the test genes with a predefined coefficient, and (c) combining the weighted expression to provide a test value, wherein at least 50%, at least at least 75% of at least 4 test genes are cell-cycle genes; and optionally (3) a second computer program means for comparing the test value to one or more reference values each associated with a predetermined degree of risk of cancer recurrence or progression of the prostate cancer, lung cancer, bladder cancer or brain cancer. In some
embodiments, the system further comprises a display module displaying the comparison between the test value to the one or more reference values, or displaying a result of the comparing step.
[0017] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
[0018] Other features and advantages of the invention will be apparent from the following Detailed Description, and from the Claims.
BRIEF DESCRIPTION OF THE DRAWINGS
[0019] Figure 1 is an illustration of the predictive power over nomogram for
CCG panels of different sizes.
[0020] Figure 2 is an illustration of CCGs predicting time to recurrence.
[0021] Figure 3 is an illustration of nomogram predicting time to recurrence. [0022] Figure 4 is an illustration of the non-overlapping recurrence predicted by nomogram and a CCG signature.
[0023] Figure 5 is an illustration of time to recurrence for several patient populations defined by nomogram and/or CCG status.
[0024] Figure 6 is an illustration of an example of a system useful in certain aspects and embodiments of the invention.
[0025] Figure 7 is a flowchart illustrating an example of a computer-implemented method of the invention.
[0026] Figure 8 a scatter plot comparing clinical parameters and CCG score as predictors of recurrence from Example 5.
[0027] Figure 9 illustrates, from Example 5, the CCG threshold derived from analysis of the training cohort to the validation data set, with the CCG signature score effectively subdividing patients identified as low-risk using clinical parameters into patients with very low recurrence rates and a higher risk of recurrence.
[0028] Figure 10 illustrates the predicted recurrence rate versus CCG score for patients in the validation cohort of Example 5.
[0029] Figure 11 illustrates the predicted recurrence rate versus CCG score for patients in the validation cohort of Example 5.
[0030] Figure 12 illustrates the distribution of clinical risk score in 443 patients studied in Example 5. The dark vertical line represents the threshold chosen by KM means to divide low- and high-risk patients and used throughout this study.
[0031] Figure 13 illustrates the correlation between CCP score and survival in brain cancer.
[0032] Figure 14 illustrates illustrates the correlation between CCP score and survival in bladder cancer.
[0033] Figure 15 illustrates illustrates the correlation between CCP score and survival in breast cancer. [0034] Figure 16 illustrates the correlation between CCP score and survival in lung cancer.
[0035] Figure 17 is an illustration of the predictive power over nomogram for CCG panels of different sizes.
DETAILED DESCRIPTION OF THE INVENTION
I. Determining Cell-Cycle Gene Expression
[0036] The present invention is based in part on the discovery that genes whose expression closely tracks the cell cycle ("cell-cycle genes" or "CCGs") are particularly powerful genes for classifying selected cancers including prostate cancer, lung cancer, bladder cancer, brain cancer and breast cancer, but not other types of cancer such as colorectal cancer.
[0037] "Cell-cycle gene" and "CCG" herein refer to a gene whose expression level closely tracks the progression of the cell through the cell-cycle. See, e.g., Whitfield et al, MOL. BIOL. CELL (2002) 13: 1977-2000. The term "cell-cycle progression" or "CCP" will also be used in this application and will generally be interchangeable with CCG (i.e., a CCP gene is a CCG; a CCP score is a CCG score). More specifically, CCGs show periodic increases and decreases in expression that coincide with certain phases of the cell cycle— e.g., STK15 and PLK show peak expression at G2/M. Id. Often CCGs have clear, recognized cell-cycle related function— e.g., in DNA synthesis or repair, in chromosome condensation, in cell-division, etc. However, some CCGs have expression levels that track the cell-cycle without having an obvious, direct role in the cell- cycle— e.g., UBE2S encodes a ubiquitin-conjugating enzyme, yet its expression closely tracks the cell-cycle. Thus a CCG according to the present invention need not have a recognized role in the cell-cycle. Exemplary CCGs are listed in Tables 1, 2, 3, and 4.
[0038] Whether a particular gene is a CCG may be determined by any technique known in the art, including that taught in Whitfield et al, MOL. BIOL. CELL (2002) 13: 1977-2000. For example, a sample of cells, e.g., HeLa cells, can be synchronized such that they all progress through the different phases of the cell cycle at the same time. Generally this is done by arresting the cells in each phase— e.g., cells may be arrested in S phase by using a double thymidine block or in mitosis with a thymidine-nocodazole block. See, e.g., Whitfield et al., MOL. CELL. BIOL. (2000) 20:4188-4198. RNA is extracted from the cells after arrest in each phase and gene expression is quantitated using any suitable technique— e.g. , expression microarray (genome-wide or specific genes of interest), real-time quantitative PCR™ (RTQ-PCR). Finally, statistical analysis (e.g., Fourier Transform) is applied to determine which genes show peak expression during particular cell- cycle phases. Genes may be ranked according to a periodicity score describing how closely the gene's expression tracks the cell-cycle— e.g., a high score indicates a gene very closely tracks the cell cycle. Finally, those genes whose periodicity score exceeds a defined threshold level (see Whitfield et al, MOL. BIOL. CELL (2002) 13: 1977-2000) may be designated CCGs. A large, but not exhaustive, list of nucleic acids associated with CCGs (e.g., genes, ESTs, cDNA clones, etc.) is given in Table 1. See Whitfield et al, MOL. BIOL. CELL (2002) 13:1977-2000. All of the CCGs in Table 2 below form a panel of CCGs ("Panel A") useful in the methods of the invention.
Table 2
Figure imgf000009_0001
CDC20* 991 Hs03004916_gl NM_001255.2
CDC45L* 8318 Hs00185895_ml NM_003504.3
CDC6* 990 Hs00154374_ml NM_001254.3
CDCA3* 83461 Hs00229905_ml NM_031299.4
CDCA8* 55143 Hs00983655_ml NM_018101.2
NM 001130851.1;
CDKN3* 1033 Hs00193192_ml
NM 005192.3
CDT1 * 81620 Hs00368864_ml NM_030928.3
NM 001042426.1;
CENPA 1058 Hs00156455_ml
NM 001809.3
CENPE* 1062 Hs00156507_ml NM_001813.2
CENPF* 1063 Hs00193201_ml NM_016343.3
CENPI* 2491 Hs00198791_ml NM_006733.2
CENPM* 79019 Hs00608780_ml NM_024053.3
NM 018455.4;
CENPN* 55839 Hs00218401_ml NM 001100624.1;
NM 001100625.1
NM 018131.4;
CEP 55* 55165 Hs00216688_ml
NM_001127182.1
NM 001114121.1;
CHEK1 * 1111 Hs00967506_ml NM 001114122.1;
NM 001274.4
NM 018204.3;
CKAP2* 26586 Hs00217068_ml
NM_001098525.1
CKS1B* 1163 Hs01029137_gl NM_001826.2
CKS2* 1164 Hs01048812_gl NM_001827.1
CTPS* 1503 Hs01041851_ml NM 001905.2
CTSL2* 1515 Hs00952036_ml NM_001333.2
DBF '4* 10926 Hs00272696_ml NM_006716.3
DDX39* 10212 Hs00271794_ml NM_005804.2
DLGAP5/DLG7* 9787 Hs00207323_ml NM_014750.3
DONSON* 29980 Hs00375083_ml NM_017613.2
DSN1 * 79980 Hs00227760_ml NM_024918.2
DTL* 51514 Hs00978565_ml NM_016448.2
E2F8* 79733 Hs00226635_ml NM_024680.2 ECT2* 1894 Hs00216455_ml NM_018098.4
ESPL1 * 9700 Hs00202246_ml NM_012291.4
NM 130398.2;
EXOl * 9156 Hs00243513_ml NM 003686.3;
NM 006027.3
NM 152998.1;
EZH2* 2146 Hs00544830_ml
NM_004456.3
NM 018193.2;
FANCI* 55215 Hs00289551_ml
NM_001113378.1
NM 001142522.1;
FBX05* 26271 Hs03070834_ml
NM_012177.3
NM 202003.1;
FOXM1 * 2305 Hs01073586_ml NM 202002.1;
NM 021953.2
GINS1 * 9837 Hs00221421_ml NM_021067.3
GMPS* 8833 Hs00269500_ml NM_003875.2
GPSM2* 29899 Hs00203271_ml NM_013296.4
GTSE1 * 51512 Hs00212681_ml NM_016426.5
H2AFX* 3014 Hs00266783_sl NM_002105.2
NM 001142556.1; NM 001142557.1;
HMMR* 3161 Hs00234864_ml
NM 012484.2; NM 012485.2
NM 001002033.1;
HN1 * 51155 Hs00602957_ml NM 001002032.1;
NM_016185.2
KIAA0101 * 9768 Hs00207134_ml NM_014736.4
KIF11 * 3832 Hs00189698_ml NM_004523.3
KIF15* 56992 Hs00173349_ml NM_020242.2
KIF18A * 81930 Hs01015428_ml NM_031217.3
KIF20A * 10112 Hs00993573_ml NM_005733.2
KIF20B /MPHOSPHl * 9585 Hs01027505_ml NM_016195.2
NM 138555.1;
KIF23* 9493 Hs00370852_ml
NM_004856.4
KIF2C* 11004 Hs00199232_ml NM_006845.3 KIF4A * 24137 Hs01020169_ml NM_012310.3
KIFC1 * 3833 Hs00954801_ml NM_002263.3
KPNA2 3838 Hs00818252_gl NM_002266.2
LMNB2* 84823 Hs00383326_ml NM_032737.2
MAD2L1 4085 Hs01554513_gl NM_002358.3
MCAM* 4162 Hs00174838_ml NM 006500.2
NM 018518.3;
MCM10* 55388 Hs00960349_ml
NM 182751.1
MCM2* 4171 Hs00170472_ml NM_004526.2
NM 005914.2;
MCM4* 4173 Hs00381539_ml
NM 182746.1
MCM6* 4175 Hs00195504_ml NM_005915.4
NM 005916.3;
MCM7* 4176 Hs01097212_ml
NM 182776.1
MELK 9833 Hs00207681_ml NM_014791.2
MKI67* 4288 Hs00606991_ml NM_002417.3
MYBL2* 4605 Hs00231158_ml NM_002466.2
NCAPD2* 9918 Hs00274505_ml NM_014865.3
NCAPG* 64151 Hs00254617_ml NM_022346.3
NCAPG2* 54892 Hs00375141_ml NM_017760.5
NCAPH* 23397 Hs01010752_ml NM_015341.3
NDC80* 10403 Hs00196101_ml NM 006101.2
NEK2* 4751 Hs00601227_mH NM_002497.2
NM 018454.6;
NUSAP1 * 51203 Hs01006195_ml NM 001129897.1;
NM 016359.3
OIP5* 11339 Hs00299079_ml NM_007280.1
ORC6L* 23594 Hs00204876_ml NM_014321.2
NM 001079524.1;
PAICS* 10606 Hs00272390_ml NM 001079525.1;
NM 006452.3
PBK* 55872 Hs00218544_ml NM_018492.2
NM 182649.1;
PCNA * 5111 Hs00427214_gl
NM 002592.2 PDSS1 * 23590 Hs00372008_ml NM_014317.3
PLK1 * 5347 Hs00153444_ml NM 005030.3
PLK4* 10733 Hs00179514_ml NM_014264.3
POLE 2* 5427 Hs00160277_ml NM_002692.2
NM 199413.1;
PRC1 * 9055 Hs00187740_ml NM 199414.1;
NM 003981.2
PSMA7* 5688 Hs00895424_ml NM_002792.2
NM 032636.6; NM 001005290.2;
PSRC1 * 84722 Hs00364137_ml
NM 001032290.1; NM 001032291.1
PTTG1 * 9232 Hs00851754_ul NM_004219.2
RACGAP1 * 29127 Hs00374747_ml NM_013277.3
NM 133487.2;
RAD51 * 5888 Hs00153418_ml
NM 002875.3
NM 001130862.1;
RAD51AP1 * 10635 HsOl 54889 l ml
NM_006479.4
RAD54B* 25788 Hs00610716_ml NM_012415.2
NM 001142548.1;
RAD54L * 8438 Hs00269177_ml
NM 003579.3
NM 181471.1;
RFC2* 5982 Hs00945948_ml
NM_002914.3
NM 181573.2;
RFC4* 5984 Hs00427469_ml
NM_002916.3
NM 181578.2; NM 001130112.1;
RFC 5* 5985 Hs00738859_ml
NM 001130113.1; NM 007370.4
RNASEH2A * 10535 Hs00197370_ml NM_006397.2
RRM2* 6241 Hs00357247_gl NM_001034.2
SHCBP1 * 79801 Hs00226915_ml NM_024745.4 NM 001042550.1;
SMC 2* 10592 Hs00197593_ml NM 001042551.1;
NM 006444.2
SPAG5* 10615 Hs00197708_ml NM 006461.3
SPC25* 57405 Hs00221100_ml NM_020675.3
NM 001048166.1;
STIL* 6491 Hs00161700_ml
NM 003035.2
Hs00606370 ml; NM 005563.3;
STMN1 * 3925
Hs01033129_ml NM_203399.1
TACC3* 10460 Hs00170751_ml NM_006342.1
TIMELESS* 8914 Hs01086966_ml NM_003920.2
TK1 * 7083 Hs01062125_ml NM_003258.4
TOP 2 A * 7153 Hs00172214_ml NM_001067.2
TPX2* 22974 Hs00201616_ml NM_012112.4
TRIP 13* 9319 Hs01020073_ml NM_004237.2
TTK* 7272 Hs00177412_ml NM_003318.3
TUBA1C* 84790 Hs00733770_ml NM_032704.3
TYMS* 7298 Hs00426591_ml NM 001071.2
NM 181799.1;
NM 181800.1;
NM 181801.1;
UBE2C 11065 Hs00964100_gl
NM 181802.1;
NM 181803.1;
NM 007019.2
UBE2S 27338 Hs00819350_ml NM 014501.2
VRKl * 7443 Hs00177470_ml NM_003384.2
NM 017975.3;
ZWILCH* 55055 Hs01555249_ml
NR 003105.1
NM 032997.2;
ZWINT* 11130 Hs00199952_ml NM 001005413.1;
NM 007057.3
* 124-gene subset of CCGs useful in the invention ("Panel B"). ABI Assay ID means the catalogue ID number for the gene expression assay commercially available from Applied Biosystems Inc. (Foster City, CA) for the particular gene. [0039] Accordingly, in a first aspect of the present invention, a method is provided for determining gene expression in a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer. Generally, the method includes at least the following steps: (1) obtaining a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer; (2) determining the expression of a panel of genes in the tumor sample including at least 2, 4, 6, 8 or 10 cell-cycle genes; and (3) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide said test value, wherein at least 20%, 50%>, at least 75% or at least 90%> of said plurality of test genes are cell- cycle genes.
[0040] Gene expression can be determined either at the R A level (i.e., m NA or noncoding RNA (ncRNA)) (e.g., miRNA, tRNA, rRNA, snoRNA, siRNA and piRNA) or at the protein level. Levels of proteins in a tumor sample can be determined by any known techniques in the art, e.g., HPLC, mass spectrometry, or using antibodies specific to selected proteins (e.g., IHC, ELISA, etc.).
[0041] In preferred embodiment, the amount of RNA transcribed from the panel of genes including test genes is measured in the tumor sample. In addition, the amount of RNA of one or more housekeeping genes in the tumor sample is also measured, and used to normalize or calibrate the expression of the test genes. The terms "normalizing genes" and "housekeeping genes" are defined herein below.
[0042] In some embodiments, the plurality of test genes includes at least 2, 3 or 4 cell-cycle genes, which constitute at least 50%>, 75% or 80%> of the plurality of test genes, and preferably 100% of the plurality of test genes. In some embodiments, the plurality of test genes includes at least 5, 6, 7, or at least 8 cell-cycle genes, which constitute at least 20%>, 25%, 30%>, 40%>, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes. As will be clear from the context of this document, a panel of genes is a plurality of genes. Typically these genes are assayed together in one or more samples from a patient.
[0043] In some other embodiments, the plurality of test genes includes at least 8, 10,
12, 15, 20, 25 or 30 cell-cycle genes, which constitute at least 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes. [0044] As will be apparent to a skilled artisan apprised of the present invention and the disclosure herein, "tumor sample" means any biological sample containing one or more tumor cells, or one or more tumor derived R A or protein, and obtained from a cancer patient. For example, a tissue sample obtained from a tumor tissue of a cancer patient is a useful tumor sample in the present invention. The tissue sample can be an FFPE sample, or fresh frozen sample, and preferably contain largely tumor cells. A single malignant cell from a cancer patient's tumor is also a useful tumor sample. Such a malignant cell can be obtained directly from the patient's tumor, or purified from the patient's bodily fluid such as blood and urine. In addition, a bodily fluid such as blood, urine, sputum and saliva containing one or tumor cells, or tumor-derived RNA or proteins, can also be useful as a tumor sample for purposes of practicing the present invention.
[0045] Those skilled in the art are familiar with various techniques for determining the status of a gene or protein in a tissue or cell sample including, but not limited to, microarray analysis (e.g., for assaying mRNA or microRNA expression, copy number, etc.), quantitative realtime PCR™ ("qRT-PCR™", e.g., TaqMan™), immunoanalysis (e.g., ELISA,
immunohistochemistry), etc. The activity level of a polypeptide encoded by a gene may be used in much the same way as the expression level of the gene or polypeptide. Often higher activity levels indicate higher expression levels and while lower activity levels indicate lower expression levels. Thus, in some embodiments, the invention provides any of the methods discussed above, wherein the activity level of a polypeptide encoded by the CCG is determined rather than or in adition to the expression level of the CCG. Those skilled in the art are familiar with techniques for measuring the activity of various such proteins, including those encoded by the genes listed in Tables 1, 2, 3, and 4. The methods of the invention may be practiced independent of the particular technique used.
[0046] In preferred embodiments, the expression of one or more normalizing genes is also obtained for use in normalizing the expression of test genes. As used herein, "normalizing genes" referred to the genes whose expression is used to calibrate or normalize the measured expression of the gene of interest (e.g., test genes). Importantly, the expression of normalizing genes should be independent of cancer outcome/prognosis, and the expression of the normalizing genes is very similar among all the tumor samples. The normalization ensures accurate comparison of expression of a test gene between different samples. For this purpose, housekeeping genes known in the art can be used. Housekeeping genes are well known in the art, with examples including, but are not limited to, GUSB (glucuronidase, beta), HMBS (hydroxymethylbilane synthase), SDHA (succinate dehydrogenase complex, subunit A, flavoprotein), UBC (ubiquitin C) and YWHAZ (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide). One or more housekeeping genes can be used. Preferably, at least 2, 5, 10 or 15 housekeeping genes are used to provide a combined normalizing gene set. The amount of gene expression of such normalizing genes can be averaged, combined together by straight additions or by a defined algorithm. Some examples of particularly useful housekeeper genes for use in the methods and compositions of the invention include those listed in Table A below.
Table A
Figure imgf000017_0001
* Subset of housekeeping genes used in, e.g., Example 5.
[0047] In the case of measuring RNA levels for the genes, one convenient and sensitive approach is real-time quantitative PCR (qPCR) assay, following a reverse transcription reaction. Typically, a cycle threshold (Ct) is determined for each test gene and each normalizing gene, i.e., the number of cycle at which the fluoescence from a qPCR reaction above background is detectable.
[0048] The overall expression of the one or more normalizing genes can be represented by a "normalizing value" which can be generated by combining the expression of all normalizing genes, either weighted eaqually (straight addition or averaging) or by different predefined coefficients. For example, in a simplest manner, the normalizing value Cm can be the cycle threshold (Ct) of one single normalizing gene, or an average of the Ct values of 2 or more, preferably 10 or more, or 15 or more normalizing genes, in which case, the predefined coefficient is 1/N, where N is the total number of normalizing genes used. Thus, QH = (Qm + Qm + " CtHn) N. As will be apparent to skilled artisans, depending on the normalizing genes used, and the weight desired to be given to each normalizing gene, any coefficients (from 0/N to N/N) can be given to the normalizing genes in weighting the expression of such normalizing genes. That is, H = xCtm + yCtH2 + " zCtHn, wherein x + y + " ' + z = 1.
[0049] As discussed above, the methods of the invention generally involve determining the level of expression of a panel of CCGs. With modern high-throughput techniques, it is often possible to determine the expression level of tens, hundreds or thousands of genes. Indeed, it is possible to determine the level of expression of the entire transcriptome (i.e., each transcribed sequence in the genome). Once such a global assay has been performed, one may then
informatically analyze one or more subsets of transcripts (i.e., panels or, as often used herein, pluralities of test genes). After measuring the expression of hundreds or thousands of transcripts in a sample, for example, one may analyze (e.g., informatically) the expression of a panel or plurality of test genes comprising primarily CCGs according to the present invention by combining the expression level values of the individual test genes to obtain a test value.
[0050] As will be apparent to a skilled artisan, the test value provided in the present inveniton represents the overall expression level of the plurality of test genes composed substantially of cell-cycle genes. In one embodiment, to provide a test value in the methods of the invention, the normalized expression for a test gene can be obtained by normalizing the measured Ct for the test gene against the H, i.e., AQi = (Qi - Cm)- Thus, the test value representing the overall expression of the plurality of test genes can be provided by combining the normalized expression of all test genes, either by straight addition or averaging (i.e., weighted eaqually) or by a different predefined coefficient. For example, the simplest approach is averaging the normalized expression of all test genes: test value = (ACtl + ACt2 + ' " + ACtn)/n. As will be apparent to skilled artisans, depending on the test genes used, different weight can also be given to different test genes in the present invention.
[0051] It has been determined that, once the CCP phenomenon reported herein is appreciated, the choice of individual CCGs for a test panel can often be somewhat arbitrary. In other words, many CCGs have been found to be very good surrogates for each other. One way of assessing whether particular CCGs will serve well in the methods and compositions of the invention is by assessing their correlation with the mean expression of CCGs (e.g., all known CCGs , a specific set of CCGs , etc.). Those CCGs that correlate particularly well with the mean are expected to perform well in assays of the invention, e.g., because these will reduce noise in the assay.
Rankings of select CCGs according to their correlation with the mean CCG expression as well as their ranking according to predictive value are given in Tables 9, 11 & 23 to 25.
[0052] Thus, in some embodiments of each of the various aspects of the invention the plurality of test genes comprises the top 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40 or more CCGs listed in Tables 9, 11, 23, 24 or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 of the following genes: ASPM, BIRC5, BUB1B, CCNB2, CDC2, CDC20, CDCA8, CDKN3, CENPF, DLGAP5, FOXM1, KIAA0101, KIF11, KIF2C, KIF4A, MCM10, NUSAP1, PRC1,
RACGAP1, and TPX2. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, or nine or all of gene numbers 2 & 3, 2 to 4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, or 2 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, or eight or all of gene numbers 3 & 4, 3 to 5, 3 to 6, 3 to 7, 3 to 8, 3 to 9, or 3 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, 10, 11, 12, 13, 14, or 15 or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to
5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, 1 to 10, 1 to 11, 1 to 12, 1 to 13, 1 to 14, or 1 to 15 of any of Tables 9, 11, 23, 24, or 25.
II. Cancer Prognosis
[0053] It has been surprisingly discovered that in selected cancers such as prostate cancer, lung cancer, bladder cancer and brain cancer, but not other cancers including certain colon cancer, the expression of cell-cycle genes in tumor cells can accurately predict the degree of aggression of the cancer and risk of recurrence after treatment (e.g., surgical removal of cancer tissue, chemotherapy and radiation therapy, etc.). Thus, the above-described method of determining cell-cycle gene expression can be applied in the prognosis and treatment of such cancers.
[0054] Generally, a method is provided for prognosing cancer selected from prostate cancer, lung cancer, bladder cancer or brain cancer, which comprises determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of at least 2, 4, 5, 6, 7 or at least 8, 9, 10 or 12 cell-cycle genes, wherein overexpression of the at least 4 cell-cycle genes indicates a poor prognosis or an increased likelihood of recurrence of cancer in the patient. The expression can be determined in accordance with the method described above.
[0055] In some embodiments, the prognosis method includes (1) obtaining a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer; (2) determining the expression of a panel of genes in the tumor sample including at least 2, 4,
6, 8 or 10 cell-cycle genes; and (3) providing a test value by (a) weighting the determined expression of each of a plurality of test genes selected from the panel of genes with a predefined coefficient, and (b) combining the weighted expression to provide said test value, wherein at least 20%, 50%>, at least 75%) or at least 90%> of said plurality of test genes are cell-cycle genes, and wherein an increased level of expression of the plurality of test genes indicates a poor prognosis or an increased likelihood of cancer recurrence.
[0056] In preferred embodiments, the test value representing the overall expression of the plurality of test genes is compared to one or more reference values (or index values), and optionally correlated to a risk of cancer progression or risk of cancer recurrence. Optionally an increased likelihood of poor prognosis is indicated if the test value is greater than the reference value.
[0057] For example, the index value may represent the gene expression levels found in a normal sample obtained from the patient of interest, in which case an expression level in the tumor sample significantly higher than this index value would indicate, e.g., a poor prognosis or increased likelihood of cancer recurrence or a need for aggressive treatment.
[0058] Alternatively, the index value may represent the average expression level of for a set of individuals from a diverse cancer population or a subset of the population. For example, one may determine the average expression level of a gene or gene panel in a random sampling of patients with cancer (e.g., prostate, bladder, brain, breast, or lung cancer). This average expression level may be termed the "threshold index value," with patients having CCG expression higher than this value expected to have a poorer prognosis than those having expression lower than this value.
[0059] Alternatively the index value may represent the average expression level of a particular gene marker in a plurality of training patients {e.g., prostate cancer patients) with similar outcomes whose clinical and follow-up data are available and sufficient to define and categorize the patients by disease outcome, e.g., recurrence or prognosis. See, e.g., Examples, infra. For example, a "good prognosis index value" can be generated from a plurality of training cancer patients characterized as having "good outcome", e.g., those who have not had cancer recurrence five years (or ten years or more) after initial treatment, or who have not had progression in their cancer five years (or ten years or more) after initial diagnosis. A "poor prognosis index value" can be generated from a plurality of training cancer patients defined as having "poor outcome", e.g., those who have had cancer recurrence within five years (or ten years, etc.) after initial treatment, or who have had progression in their cancer within five years (or ten years, etc.) after initial diagnosis. Thus, a good prognosis index value of a particular gene may represent the average level of expression of the particular gene in patients having a "good outcome," whereas a poor prognosis index value of a particular gene represents the average level of expression of the particular gene in patients having a "poor outcome."
[0060] Thus one aspect of the invention provides a method of classifying cancer comprising determining the status of a panel of genes comprising at least two CCGs, in tissue or cell sample, particularly a tumor sample, from a patient, wherein an abnormal status indicates a negative cancer classification. As used herein, "determining the status" of a gene refers to determining the presence, absence, or extent/level of some physical, chemical, or genetic characteristic of the gene or its expression product(s). Such characteristics include, but are not limited to, expression levels, activity levels, mutations, copy number, methylation status, etc.
[0061] In the context of CCGs as used to determine risk of cancer recurrence or progression or need for aggressive treatment, particularly useful characteristics include expression levels (e.g., mR A or protein levels) and activity levels. Characteristics may be assayed directly (e.g., by assaying a CCG's expression level) or determined indirectly (e.g., assaying the level of a gene or genes whose expression level is correlated to the expression level of the CCG). Thus some embodiments of the invention provide a method of classifying cancer comprising determining the expression level, particularly mRNA level of a panel of genes comprising at least two CCGs, in a tumor sample, wherein elevated expression indicates a negative cancer classification, or an increased risk of cancer recurrence or progression, or a need for aggressive treatment.
[0062] "Abnormal status" means a marker's status in a particular sample differs from the status generally found in average samples (e.g., healthy samples or average diseased samples). Examples include mutated, elevated, decreased, present, absent, etc. An "elevated status" means that one or more of the above characteristics (e.g., expression or mRNA level) is higher than normal levels. Generally this means an increase in the characteristic (e.g., expression or mRNA level) as compared to an index value. Conversely a "low status" means that one or more of the above characteristics (e.g., gene expression or mRNA level) is lower than normal levels. Generally this means a decrease in the characteristic (e.g., expression) as compared to an index value. In this context, a "negative status" generally means the characteristic is absent or undetectable. For example, PTEN status is negative if PTEN nucleic acid and/or protein is absent or undetectable in a sample. However, negative PTEN status also includes a mutation or copy number reduction in PTEN. [0063] In some embodiments of the invention the methods comprise determining the expression of one or more CCGs and, if this expression is "increased," the patient has a poor prognosis. In the context of the invention, "increased" expression of a CCG means the patient's expression level is either elevated over a normal index value or a threshold index (e.g. , by at least some threshold amount) or closer to the "poor prognosis index value" than to the "good prognosis index value."
[0064] Thus, when the determined level of expression of a relevant gene marker is closer to the good prognosis index value of the gene than to the poor prognosis index value of the gene, then it can be concluded that the patient is more likely to have a good prognosis, i.e., a low (or no increased) likelihood of cancer recurrence. On the other hand, if the determined level of expression of a relevant gene marker is closer to the poor prognosis index value of the gene than to the good prognosis index value of the gene, then it can be concluded that the patient is more likely to have a poor prognosis, i.e., an increased likelihood of cancer recurrence.
[0065] Alternatively index values may be determined thusly: In order to assign patients to risk groups, a threshold value will be set for the cell cycle mean. The optimal threshold value is selected based on the receiver operating characteristic (ROC) curve, which plots sensitivity vs (1 - specificity). For each increment of the cell cycle mean, the sensitivity and specificity of the test is calculated using that value as a threshold. The actual threshold will be the value that optimizes these metrics according to the artisans requirements (e.g., what degree of sensitivity or specificity is desired, etc.). Example 5 demonstrates determination of a threshold value determined and validated experimentally.
[0066] Panels of CCGs (e.g., 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more CCGs) can accurately predict prognosis, as shown in Example 3. Those skilled in the art are familiar with various ways of determining the expression of a panel of genes (i.e., a plurality of genes). One may determine the expression of a panel of genes by determining the average expression level (normalized or absolute) of all panel genes in a sample obtained from a particular patient (either throughout the sample or in a subset of cells from the sample or in a single cell). Increased expression in this context will mean the average expression is higher than the average expression level of these genes in normal patients (or higher than some index value that has been determined to represent the average expression level in a reference population such as patients with the same cancer). Alternatively, one may determine the expression of a panel of genes by determining the average expression level (normalized or absolute) of at least a certain number (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30 or more) or at least a certain proportion (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100%) of the genes in the panel. Alternatively, one may determine the expression of a panel of genes by determining the absolute copy number of the mRNA (or protein) of all the genes in the panel and either total or average these across the genes.
[0067] As used herein, "classifying a cancer" and "cancer classification" refer to determining one or more clinically-relevant features of a cancer and/or determining a particular prognosis of a patient having said cancer. Thus "classifying a cancer" includes, but is not limited to: (i) evaluating metastatic potential, potential to metastasize to specific organs, risk of recurrence, and/or course of the tumor; (ii) evaluating tumor stage; (iii) determining patient prognosis in the absence of treatment of the cancer; (iv) determining prognosis of patient response (e.g. , tumor shrinkage or progression- free survival) to treatment (e.g. , chemotherapy, radiation therapy, surgery to excise tumor, etc.); (v) diagnosis of actual patient response to current and/or past treatment; (vi) determining a preferred course of treatment for the patient; (vii) prognosis for patient relapse after treatment (either treatment in general or some particular treatment); (viii) prognosis of patient life expectancy (e.g., prognosis for overall survival), etc.
[0068] Thus, a "negative classification" means an unfavorable clinical feature of the cancer (e.g., a poor prognosis). Examples include (i) an increased metastatic potential, potential to metastasize to specific organs, and/or risk of recurrence; (ii) an advanced tumor stage; (iii) a poor patient prognosis in the absence of treatment of the cancer; (iv) a poor prognosis of patient response (e.g. , tumor shrinkage or progression- free survival) to a particular treatment (e.g. , chemotherapy, radiation therapy, surgery to excise tumor, etc.); (v) a poor prognosis for patient relapse after treatment (either treatment in general or some particular treatment); (vi) a poor prognosis of patient life expectancy (e.g., prognosis for overall survival), etc. In some embodiments a recurrence- associated clinical parameter (or a high nomogram score) and increased expression of a CCG indicate a negative classification in cancer (e.g., increased likelihood of recurrence or progression).
[0069] As discussed above, it is thought that elevated CCG expression accompanies rapidly proliferating (and thus more aggressive) cancer cells. Such a cancer in a patient will often mean the patient has an increased likelihood of recurrence after treatment (e.g., the cancer cells not killed or removed by the treatment will quickly grow back). Such a cancer can also mean the patient has an increased likelihood of cancer progression for more rapid progression (e.g., the rapidly proliferating cells will cause any tumor to grow quickly, gain in virulence, and/or metastasize). Such a cancer can also mean the patient may require a relatively more aggressive treatment. Thus, in some embodiments the invention provides a method of classifying cancer comprising determining the status of a panel of genes comprising at least two CCGs, wherein an abnormal status indicates an increased likelihood of recurrence or progression. As discussed above, in some embodiments the status to be determined is gene expression levels. Thus in some embodiments the invention provides a method of determining the prognosis of a patient's cancer comprising determining the expression level of a panel of genes comprising at least two CCGs, wherein elevated expression indicates an increased likelihood of recurrence or progression of the cancer.
[0070] "Recurrence" and "progression" are terms well-known in the art and are used herein according to their known meanings. As an example, the meaning of "progression" may be cancer-type dependent, with progression in lung cancer meaning something different from
progression in prostate cancer. However, within each cancer-type and subtype "progression" is clearly understood to those skilled in the art. As used herein, a patient has an "increased likelihood" of some clinical feature or outcome (e.g., recurrence or progression) if the probability of the patient having the feature or outcome exceeds some reference probability or value. The reference probability may be the probability of the feature or outcome across the general relevant patient population. For example, if the probability of recurrence in the general prostate cancer population is X% and a particular patient has been determined by the methods of the present invention to have a probability of recurrence of Y%, and if Y > X, then the patient has an "increased likelihood" of recurrence. Alternatively, as discussed above, a threshold or reference value may be determined and a particular patient's probability of recurrence may be compared to that threshold or reference.
Because predicting recurrence and predicting progression are prognostic endeavors, "predicting prognosis" will often be used herein to refer to either or both. In these cases, a "poor prognosis" will generally refer to an increased likelihood of recurrence, progression, or both.
[0071] As shown in Example 3, individual CCGs can predict prognosis quite well.
Thus the invention provides a method of predicting prognosis comprising determining the expression of at least one CCG listed in Table 1 or Panels A through G.
[0072] Example 3 also shows that panels of CCGs (e.g., 2, 3, 4, 5, or 6 CCGs) can accurately predict prognosis. Thus in some aspects the invention provides a method of classifying a cancer comprising determining the status of a panel of genes (e.g., a plurality of test genes) comprising a plurality of CCGs. For example, increased expression in a panel of genes (or plurality of test genes) may refer to the average expression level of all panel or test genes in a particular patient being higher than the average expression level of these genes in normal patients (or higher than some index value that has been determined to represent the normal average expression level). Alternatively, increased expression in a panel of genes may refer to increased expression in at least a certain number (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30 or more) or at least a certain proportion (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100%) of the genes in the panel as compared to the average normal expression level.
[0073] In some embodiments the panel comprises at least 3, 4, 5, 6, 7, 8, 9, 10, 15,
20, 25, 30, 35, 40, 45, 50, 70, 80, 90, 100, 200, or more CCGs. In some embodiments the panel comprises at least 10, 15, 20, or more CCGs. In some embodiments the panel comprises between 5 and 100 CCGs, between 7 and 40 CCGs, between 5 and 25 CCGs, between 10 and 20 CCGs, or between 10 and 15 CCGs. In some embodiments CCGs comprise at least a certain proportion of the panel. Thus in some embodiments the panel comprises at least 25%, 30%>, 40%>, 50%>, 60%>, 70%>, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% CCGs. In some preferred embodiments the panel comprises at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 70, 80, 90, 100, 200, or more CCGs, and such CCGs constitute of at least 50%, 60%, 70%, preferably at least 75%, 80%, 85%, more preferably at least 90%, 95%, 96%, 97%, 98%, or 99% or more of the total number of genes in the panel. In some embodiments the CCGs are chosen from the group consisting of the genes in Table 1 and Panels A through G. In some embodiments the panel comprises at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 20, 25, 30, or more of the genes in any of Table 1 and Panels A through G. In some embodiments the invention provides a method of predicting prognosis comprising determining the status of the CCGs in Panels A through G, wherein abnormal status indicates a poor prognosis.
[0074] In some of these embodiments elevated expression indicates an increased likelihood of recurrence or progression. Thus in a preferred embodiment the invention provides a method of predicting risk of cancer recurrence or progression in a patient comprising determining the status of a panel of genes, wherein the panel comprises between about 10 and about 15 CCGs, the CCGs constitute at least 90% of the panel, and an elevated status for the CCGs indicates an increased likelihood or recurrence or progression. [0075] Several panels of CCGs (Table 2, supra, and Tables 3 & 4, infra) have been evaluated for their ability to predict prognosis in several different cancers. The results of these studies are described in Examples 1 through 6 below.
Table 3: "Panel C" Evaluated in Examples 1 through 4
Figure imgf000027_0001
These genes were used as a 26-gene subset panel ("Panel D") in the validation arm of the experiment described in Example 2.
Table 4: "Panel E
Figure imgf000027_0002
These genes were used as a 31 -gene subset panel ("Panel F") in the experiment described in Example 5. [0076] It has been determined that the choice of individual CCGs for a panel can often be relatively arbitrary. In other words, most CCGs have been found to be very good surrogates for each other. One way of assessing whether particular CCGs will serve well in the methods and compositions of the invention is by assessing their correlation with the mean expression of CCGs (e.g., all known CCGs, a specific set of CCGs, etc.). Those CCGs that correlate particularly well with the mean are expected to perform well in assays of the invention, e.g., because these will reduce noise in the assay. A ranking of select CCGs according to their correlation with the mean CCG expression is given in Table 23.
[0077] In CCG signatures the particular CCGs assayed is often not as important as the total number of CCGs. The number of CCGs assayed can vary depending on many factors, e.g., technical constraints, cost considerations, the classification being made, the cancer being tested, the desired level of predictive power, etc. Increasing the number of CCGs assayed in a panel according to the invention is, as a general matter, advantageous because, e.g., a larger pool of mR As to be assayed means less "noise" caused by outliers and less chance of an assay error throwing off the overall predictive power of the test. However, cost and other considerations will generally limit this number and finding the optimal number of CCGs for a signature is desirable.
[0078] It has been discovered that the predictive power of a CCG signature often ceases to increase significantly beyond a certain number of CCGs (see FIG. l; Example 1). More specifically, the optimal number of CCGs in a signature (no) can be found wherever the following is true
(P«+i - P«) < Co,
wherein P is the predictive power (i.e., P„ is the predictive power of a signature with n genes and P„+i is the predictive power of a signature with n genes plus one) and Co is some optimization constant. Predictive power can be defined in many ways known to those skilled in the art including, but not limited to, the signature's p-value. Co can be chosen by the artisan based on his or her specific constraints. For example, if cost is not a critical factor and extremely high levels of sensitivity and specificity are desired, Co can be set very low such that only trivial increases in predictive power are disregarded. On the other hand, if cost is decisive and moderate levels of sensitivity and specificity are acceptable, Co can be set higher such that only significant increases in predictive power warrant increasing the number of genes in the signature. [0079] Alternatively, a graph of predictive power as a function of gene number may be plotted (as in FIG.l) and the second derivative of this plot taken. The point at which the second derivative decreases to some predetermined value (Co') may be the optimal number of genes in the signature.
[0080] Examples 1 & 3 and FIGs 1 & 17 illustrate the empirical determination of optimal numbers of CCGs in CCG panels of the invention. Randomly selected subsets of the 31 CCGs listed in Table 3 were tested as distinct CCG signatures and predictive power (i.e., p-value) was determined for each. As FIG. 1 shows, p-values ceased to improve significantly between about 10 and about 15 CCGs, thus indicating that an optimal number of CCGs in a prognostic panel is from about 10 to about 15. Thus some embodiments of the invention provide a method of predicting prognosis in a patient having prostate cancer comprising determining the status of a panel of genes, wherein the panel comprises between about 10 and about 15 CCGs and an elevated status for the CCGs indicates a poor prognosis. In some embodiments the panel comprises between about 10 and about 15 CCGs and the CCGs constitute at least 90% of the panel. In other embodiments the panel comprises CCGs plus one or more additional markers that significantly increase the predictive power of the panel (i.e., make the predictive power significantly better than if the panel consisted of only the CCGs). Any other combination of CCGs (including any of those listed in Table 1 or Panels A through G) can be used to practice the invention.
[0081] It has been discovered that CCGs are particularly predictive in certain cancers.
For example, panels of CCGs have been determined to be accurate in predicting recurrence in prostate cancer (Examples 1 through 5). Further, CCGs can determine prognosis in bladder, brain, breast and lung cancers, as summarized in Example 6 and Tables 21 and 22 below.
[0082] Thus the invention provides a method comprising determining the status of a panel of genes comprising at least two CCGs, wherein an abnormal status indicates a poor prognosis. In some embodiments the panel comprises at least 2 genes chosen from the group of genes in at least one of Panels A through G. In some embodiments the panel comprises at least 10 genes chosen from the group of genes in at least one of Panels A through G. In some embodiments the panel comprises at least 15 genes chosen from the group of genes in at least one of Panels A through G. In some embodiments the panel comprises all of the genes in at least one of Panels A through G. The invention also provides a method of determining the prognosis of bladder cancer, comprising determining the status of a panel of genes comprising at least two CCGs (e.g., at least two of the genes in any of Panels B, C, & F), wherein an abnormal status indicates a poor prognosis. The invention also provides a method of determining the prognosis of brain cancer, comprising determining the status of a panel of genes comprising at least two CCGs (e.g., at least two of the genes in any of Panels B, C, & F), wherein an abnormal status indicates a poor prognosis. The invention further provides a method of determining the prognosis of breast cancer, comprising determining the status of a panel of genes comprising at least two CCGs (e.g., at least two of the genes in any of Panels B, C, & F), wherein an abnormal status indicates a poor prognosis. The invention also provides a method of determining the prognosis of lung cancer, comprising determining the status of a panel of genes comprising at least two CCGs (e.g., at least two of the genes in any of Panels B, C, & F), wherein an abnormal status indicates a poor prognosis.
[0083] In some embodiments the panel comprises at least 3, 4, 5, 6, 7, 8, 9, 10, 15,
20, 25, 30, 35, 40, 45, 50 or more CCGs. In some embodiments the panel comprises between 5 and 100 CCGs, between 7 and 40 CCGs, between 5 and 25 CCGs, between 10 and 20 CCGs, or between 10 and 15 CCGs. In some embodiments CCGs comprise at least a certain proportion of the panel. Thus in some embodiments the panel comprises at least 25%, 30%>, 40%>, 50%>, 60%>, 70%>, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% CCGs. In some embodiments the CCGs are chosen from the group consisting of the genes listed in Tables 1, 9 & 11 and Panels A through G. In some embodiments the panel comprises at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more genes chosen from the group of genes in any of Tables 1, 9 or 11 or Panels A through G. In some embodiments the panel comprises all of the genes in any of Tables 1, 9, or 11 or Panels A through G.
[0084] As mentioned above, many of the CCGs of the invention have been analyzed to determine their correlation to the CCG mean and also, for the genes, to determine their relative predictive value within a panel (see Tables 9, 11, & 23 to 25). Thus in some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises the top 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40 or more CCGs listed in Table 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 of the following genes: ASPM, BIRC5, BUBIB, CCNB2, CDC2, CDC20, CDCA8, CDKN3, CENPF, DLGAP5, FOXMl, KIAAOlOl, KIFll, KIF2C, KIF4A, MCM10, NUSAP1, PRC1, RACGAP1, and TPX2. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, or nine or all of gene numbers 2 & 3, 2 to 4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, or 2 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, or eight or all of gene numbers 3 & 4, 3 to 5, 3 to 6, 3 to 7, 3 to 8, 3 to 9, or 3 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, 10, 11, 12, 13, 14, or 15 or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, 1 to 10, 1 to 11, 1 to 12, 1 to 13, 1 to 14, or 1 to 15 of any of Tables 9, 11, 23, 24, or 25.
[0085] It has further been discovered that CCG status synergistically adds to clinical parameters in prognosing cancer. In the case of prostate cancer, for example, it has been discovered that a high level of gene expression of any one of the genes in Panels C through F is associated with an increased risk of prostate cancer recurrence or progression in patients whose clinical nomogram score indicates a relatively low risk of recurrence or progression. Because evaluating CCG expression levels can thus detect increased risk not detected using clinical parameters alone, the invention generally provides methods combining evaluating at least one clinical parameter with evaluating the status of at least one CCG.
[0086] As Example 3 shows, even individual CCGs add to clinical parameters in predicting cancer recurrence. Thus one aspect of the invention provides an in vitro diagnostic method comprising determining at least one clinical parameter for a cancer patient and determining the status of at least one CCG in a sample obtained from the patient. However, assessing the status of multiple CCGs improves predictive power even more (also shown in Example 1). Thus in some embodiments the status of a plurality of CCGs (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 50 or more) is determined. In some embodiments abnormal status indicates an increased likelihood of recurrence or progression. In some embodiments the patient has prostate cancer. In some embodiments the patient has lung cancer. Often the clinical parameter is at least somewhat independently predictive of recurrence or progression and the addition of CCG status improves the predictive power. As used herein, "clinical parameter" and "clinical measure" refer to disease or patient characteristics that are typically applied to assess disease course and/or predict outcome. Examples in cancer generally include tumor stage, tumor grade, lymph node status, histology, performance status, type of surgery, surgical margins, type of treatment, and age of onset. In prostate cancer clinicians often use pre-sugery blood PSA levels, stage (defined by size of tumor and evidence of metastasis), and Gleason score (similar to concept of grade). After surgical intervention, important clinical parameters in prostate cancer include margin and lymph node status. In breast cancer clinicians often use size of index lesion in cm, invasion, number of nodes involved, and grade.
[0087] Often certain clinical parameters are correlated with a particular disease character. For example, in cancer generally as well as in specific cancers, certain clinical parameters are correlated with, e.g., likelihood of recurrence or metastasis, prognosis for survival for a certain amount of time, likelihood of response to treatment generally or to a specific treatment, etc. In prostate cancer some clinical parameters are such that their status (presence, absence, level, etc.) is associated with increased likelihood of recurrence. Examples of such recurrence-associated parameters (some but not all of which are specific to prostate cancer) include high PSA levels (e.g., greater than 4ng/ml), high Gleason score, large tumor size, evidence of metastasis, advanced tumor stage, nuclear grade, lymph node involvement, early age of onset. Other types of cancer may have different parameters correlated to likelihood of recurrence or progression, and CCG status, as a measure of proliferative activity, adds to these parameters in predicting prognosis in these cancers. As used herein, "recurrence-associated clinical parameter" has its conventional meaning for each specific cancer, with which those skilled in the art are quite familiar. In fact, those skilled in the art are familiar with various recurrence-associated clinical parameters beyond those listed here. [0088] Often a physician will assess more than one clinical parameter in a patient and make a more comprehensive evaluation for the disease characters of interest. Example 5 shows how CCG status can add to one particular grouping of clinical parameters used to determine risk of recurrence in prostate cancer. Clinical parameters in Example 5 include binary variables for organ- confined disease and Gleason score less than or equal to 6, and a continuous variable for logarithmic PSA (Table 14). This model includes all of the clinical parameters incorporated in the post-RP nomogram (i.e., Kattan- Stephenson nomogram) except for Year of RP and the two components of the Gleason score. Thus in some embodiments at least two clinical parameters (e.g., two of the above listed parameters) are assessed along with the expression level of at least one CCG.
[0089] One way in which single, but more often multiple, clinical parameters are utilized by physicians is with the help of nomograms. In the clinical setting, nomograms are representations (often visual) of a correlation between one or more parameters and one or more patient or disease characters. An example of a prevalent clinical nomogram used in determining a prostate cancer patient's likelihood of recurrence is described in Kattan et al., J. CLIN. ONCOL.
(1999) 17: 1499-1507, and updated in Stephenson et al, J. CLIN. ONCOL. (2005) 23 :7005-7012 ("Kattan- Stephenson nomogram"). This nomogram evaluates a patient by assigning a point value to each of several clinical parameters (year of RP, surgical margins, extracapsular extension, seminal vesicle invasion, lymph node involvement, primary Gleason score, secondary Gleason score, and preoperative PSA level), totalling the points for a patient into a nomogram score, and then predicting the patient's likelihood of being recurrence-free at varying time intervals (up to 10 years) based on this nomogram score. An example of a prevalent clinical nomogram used in determining a breast cancer patient's prognosis for survival is the Nottingham Prognostic Index (NPI). See, e.g., Galea et al, BREAST CANCER RES. & TREAT. (1992) 22:207- 19.
[0090] It has been discovered that determining the status of a CCG in a sample obtained from a prostate cancer patient, along with the patient's Kattan- Stephenson nomogram score, is a better predictor of 10-year recurrence-free survival than the nomogram score alone. See, e.g., Examples 2 & 5, infra. Specifically, adding CCG status to the Kattan-Stephenson nomogram detects patients at significantly increased risk of recurrence that the nomogram alone does not.
Table 3 above provides an exemplary panel of 31 CCGs (Panel C) and a subset panel of 26 CCGs (Panel D, shown with ) determined in Example 2 to show predictive synergy with the Kattan- Stephenson nomogram in prostate cancer prognosis. It has also been discovered that determining the status of a CCG in a sample obtained from a breast cancer patient, along with the patient's NPI score, is a better prognostic predictor than NPI score alone. See, e.g., Example 6, infra.
Specifically, adding CCG status to the NPI nomogram detects patients at significantly increased risk of recurrence that the nomogram alone does not. Panels B, C and D were determined in Example 2 to show predictive synergy with the NPI nomogram in breast cancer prognosis.
[0091] Thus another aspect of the invention provides an in vitro method comprising determining a clinical nomogram score {e.g., Kattan- Stephenson or NPI nomogram score) for a cancer patient and determining the status of at least one CCG in a sample obtained from the patient. Example 3 illustrates the empirical determination of the predictive power of individual CCGs and of several CCG panels of varying size over the Kattan-Stephenson nomogram. Randomly selected subsets of the 31 CCGs listed in Table 3 were tested as distinct CCG signatures and predictive power (i.e., p-value) was determined for each. As FIG. l shows, CCG signatures of 2, 3, 4, 5, 6, 10, 15, 20,
25, and 26 genes each add predictive power to the nomogram. Thus the invention provides a method of determining whether a prostate cancer patient has an increased likelihood of recurrence comprising determining the status of a panel of genes comprising at least 2, 3, 4, 5, 6, 10, 15, 20, 25,
26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 40, 45, 50, 60, 70, 80, 90, or 100 or more CCGs, wherein an elevated status (e.g., increased expression) for the CCGs indicates an increased likelihood of recurrence. In some embodiments the method further comprises determining a clinical nomogram score of the patient. The invention further provides a method of determining whether a breast cancer patient has an increased likelihood of recurrence comprising determining the status of a panel of genes comprising at least 2, 3, 4, 5, 6, 10, 15, 20, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 40, 45, 50, 60, 70, 80, 90, or 100 or more CCGs, wherein an elevated status (e.g., increased expression) for the CCGs indicates an increased likelihood of recurrence. In some embodiments the method further comprises determining a clinical nomogram score of the patient.
[0092] Often clinical nomograms for cancer are designed such that a particular value
{e.g., high score) correlates with an increased risk of recurrence. Elevated CCG status {e.g., increased expression or activity) is also correlated with increased risk. Thus, in some embodiments the invention provides a method of determining whether a cancer patient has an increased likelihood of recurrence or progression comprising determining a clinical nomogram score for the patient and determining the status of at least one CCG in a sample obtained from the patient, wherein a high nomogram score and/or an elevated CCG status indicate the patient has an increased likelihood of recurrence or progression. In some embodiments the cancer is prostate cancer. In some embodiments the cancer is lung cancer.
[0093] In some embodiments this assessment is made before radical prostatectomy
(e.g., using a prostate biopsy sample) while in some embodiments it is made after (e.g., using the resected prostate sample). In some embodiments, a sample of one or more cells are obtained from a prostate cancer patient before or after treatment for analysis according to the present invention. Prostate cancer treatment currently applied in the art includes, e.g., prostatectomy, radiotherapy, hormonal therapy (e.g., using GnRH antagonists, GnRH agonists, antiandrogens), chemotherapy, and high intensity focused ultrasound. In some embodiments, one or more prostate tumor cells from prostate cancer tissue are obtained from a prostate cancer patient during biopsy or prostatectomy and are used for analysis in the method of the present invention.
[0094] The present invention is also based on the discovery that PTEN status predicts aggressive prostate cancer. PTEN status adds to both clinical parameters (e.g., Kattan- Stephenson nomogram) and CCGs (e.g., the genes in Table 1 or Panels A through G). As described in more detail in Example 4 below, PTEN status was determined in 191 prostate cancer patient samples with accompanying clinical history data and CCG signature data. Negative PTEN status was found to be a significant predictor for risk of recurrence (p-value 0.031). PTEN remained a significant predictor of recurrence after adjusting for post-surgery clinical parameters and the CCG signature shown in Table 3 (p-value 0.026). In addition, and importantly, the combination of PTEN and the CCG signature seems to be a better predictor of recurrence than post-surgery clinical parameters (p-value 0.0002).
[0095] Because PTEN is an independent predictor of prostate cancer recurrence, one aspect of the invention provides a method of predicting a patient's likelihood of prostate cancer recurrence comprising determining PTEN status in a sample from the patient, wherein a low or negative PTEN status indicates the patient has a high likelihood of recurrence. PTEN status can be determined by any technique known in the art, including but not limited to those discussed herein.
[0096] Because PTEN adds to CCG status in predicting prostate cancer recurrence, another aspect of the invention provides an in vitro method comprising determining PTEN status and determining the status of a plurality of CCGs in a sample obtained from a patient. Different combinations of techniques can be used to determine the status the various markers. For example, in one embodiment PTEN status is determined by immunohistochemistry (IHC) while the status of the plurality of CCGs is determined by quantitative polymerase chain reaction (qPCR™), e.g.,
TaqMan™. Some embodiments of the invention provide a method of determining a prostate cancer patient's likelihood of recurrence comprising determining PTEN status in a sample obtained from the patient, determining the status of a plurality of CCGs in a sample obtained from the patient, wherein low or negative PTEN status and/or elevated CCG status indicate the patient has an increased likelihood of recurrence.
[0097] Because PTEN status adds predictive value to clinical parameters in predicting prostate recurrence, yet another aspect of the invention provides an in vitro method comprising determining PTEN status and determining at least one clinical parameter for a cancer patient. Often the clinical parameter is at least somewhat independently predictive of recurrence and the addition of PTEN status improves the predictive power. In some embodiments the invention provides a method of determining whether a cancer patient has an increased likelihood of recurrence comprising determining the status of PTEN in a sample obtained from the patient and determining a clinical nomogram score for the patient, wherein low or negative PTEN status and/or a high nomogram score indicate the patient has an increased likelihood of recurrence.
[0098] Because all three of the above markers are additive, some embodiments of the invention provide a method of determining whether a cancer patient has an increased likelihood of recurrence comprising determining the status of PTEN in a sample obtained from the patient, determining a clinical nomogram score for the patient and determining the status of at least one CCG in a sample obtained from the patient, wherein low or negative PTEN status, a high nomogram score and an elevated CCG status indicate the patient has an increased likelihood of recurrence.
[0099] The results of any analyses according to the invention will often be communicated to physicians, genetic counselors and/or patients (or other interested parties such as researchers) in a transmittable form that can be communicated or transmitted to any of the above parties. Such a form can vary and can be tangible or intangible. The results can be embodied in descriptive statements, diagrams, photographs, charts, images or any other visual forms. For example, graphs showing expression or activity level or sequence variation information for various genes can be used in explaining the results. Diagrams showing such information for additional target gene(s) are also useful in indicating some testing results. The statements and visual forms can be recorded on a tangible medium such as papers, computer readable media such as floppy disks, compact disks, etc., or on an intangible medium, e.g., an electronic medium in the form of email or website on internet or intranet. In addition, results can also be recorded in a sound form and transmitted through any suitable medium, e.g., analog or digital cable lines, fiber optic cables, etc., via telephone, facsimile, wireless mobile phone, internet phone and the like.
[00100] Thus, the information and data on a test result can be produced anywhere in the world and transmitted to a different location. As an illustrative example, when an expression level, activity level, or sequencing (or genotyping) assay is conducted outside the United States, the information and data on a test result may be generated, cast in a transmittable form as described above, and then imported into the United States. Accordingly, the present invention also encompasses a method for producing a transmittable form of information on at least one of (a) expression level or (b) activity level for at least one patient sample. The method comprises the steps of (1) determining at least one of (a) or (b) above according to methods of the present invention; and (2) embodying the result of the determining step in a transmittable form. The transmittable form is the product of such a method.
[00101] Techniques for analyzing such expression, activity, and/or sequence data
(indeed any data obtained according to the invention) will often be implemented using hardware, software or a combination thereof in one or more computer systems or other processing systems capable of effectuating such analysis.
[00102] Thus, the present invention further provides a system for determining gene expression in a tumor sample, comprising: (1) a sample analyzer for determining the expression levels of a panel of genes in a tumor sample including at least 2, 4, 6, 8 or 10 cell-cycle genes, wherein the sample analyzer contains the tumor sample which is from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer, or cDNA molecules from mR A expressed from the panel of genes; (2) a first computer program means for (a) receiving gene expression data on at least 4 test genes selected from the panel of genes, (b) weighting the determined expression of each of the test genes, and (c) combining the weighted expression to provide a test value, wherein at least 20%, 50%, at least 75% or at least 90% of the test genes are cell-cycle genes; and optionally (3) a second computer program means for comparing the test value to one or more reference values each associated with a predetermined degree of risk of cancer recurrence or progression of the prostate cancer, lung cancer, bladder cancer or brain cancer. In some embodiments, the system further comprises a display module displaying the comparison between the test value to the one or more reference values, or displaying a result of the comparing step.
[00103] In preferred embodiment, the amount of R A transcribed from the panel of genes including test genes is measured in the tumor sample. In addition, the amount of RNA of one or more housekeeping genes in the tumor sample is also measured, and used to normalize or calibrate the expression of the test genes, as described above.
[00104] In some embodiments, the plurality of test genes includes at least 2, 3 or 4 cell-cycle genes, which constitute at least 50%, 75% or 80%> of the plurality of test genes, and preferably 100% of the plurality of test genes. In some embodiments, the plurality of test genes includes at least 5, 6 or 7, or at least 8 cell-cycle genes, which constitute at least 20%>, 25%, 30%>, 40%, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes.
[00105] In some other embodiments, the plurality of test genes includes at least 8, 10,
12, 15, 20, 25 or 30 cell-cycle genes, which constitute at least 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes.
[00106] The sample analyzer can be any instruments useful in determining gene expression, including, e.g., a sequencing machine, a real-time PCR machine, and a microarray instrument.
[00107] The computer-based analysis function can be implemented in any suitable language and/or browsers. For example, it may be implemented with C language and preferably using object-oriented high-level programming languages such as Visual Basic, SmallTalk, C++, and the like. The application can be written to suit environments such as the Microsoft Windows™ environment including Windows™ 98, Windows™ 2000, Windows™ NT, and the like. In addition, the application can also be written for the Macintosh™, SUN™, UNIX or LINUX environment. In addition, the functional steps can also be implemented using a universal or platform-independent programming language. Examples of such multi-platform programming languages include, but are not limited to, hypertext markup language (HTML), JAVA™, JavaScript™, Flash programming language, common gateway interface/structured query language (CGI/SQL), practical extraction report language (PERL), AppleScript™ and other system script languages, programming language/structured query language (PL/SQL), and the like. Java - or JavaScript -enabled browsers such as HotJava™, Microsoft™ Explorer™, or Netscape™ can be used. When active content web pages are used, they may include Java™ applets or ActiveX™ controls or other active content technologies.
[00108] The analysis function can also be embodied in computer program products and used in the systems described above or other computer- or internet-based systems. Accordingly, another aspect of the present invention relates to a computer program product comprising a computer-usable medium having computer-readable program codes or instructions embodied thereon for enabling a processor to carry out gene status analysis. These computer program instructions may be loaded onto a computer or other programmable apparatus to produce a machine, such that the instructions which execute on the computer or other programmable apparatus create means for implementing the functions or steps described above. These computer program
instructions may also be stored in a computer-readable memory or medium that can direct a computer or other programmable apparatus to function in a particular manner, such that the instructions stored in the computer-readable memory or medium produce an article of manufacture including instruction means which implement the analysis. The computer program instructions may also be loaded onto a computer or other programmable apparatus to cause a series of operational steps to be performed on the computer or other programmable apparatus to produce a computer implemented process such that the instructions which execute on the computer or other
programmable apparatus provide steps for implementing the functions or steps described above.
[00109] Thus one aspect of the present invention provides a system for determining whether a patient has increased likelihood of recurrence. Generally speaking, the system comprises (1) computer program means for receiving, storing, and/or retrieving a patient's gene status data (e.g. , expression level, activity level, variants) and optionally clinical parameter data (e.g. , Gleason score, nomogram score); (2) computer program means for querying this patient data; (3) computer program means for concluding whether there is an increased likelihood of recurrence based on this patient data; and optionally (4) computer program means for outputting/displaying this conclusion. In some embodiments this means for outputting the conclusion may comprise a computer program means for informing a health care professional of the conclusion.
[00110] One example of such a computer system is the computer system [600] illustrated in FIG.6. Computer system [600] may include at least one input module [630] for entering patient data into the computer system [600] . The computer system [600] may include at least one output module [624] for indicating whether a patient has an increased or decreased likelihood of response and/or indicating suggested treatments determined by the computer system
[600] . Computer system [600] may include at least one memory module [606] in communication with the at least one input module [630] and the at least one output module [624] .
[00111] The at least one memory module [606] may include, e.g., a removable storage drive [608], which can be in various forms, including but not limited to, a magnetic tape drive, a floppy disk drive, a VCD drive, a DVD drive, an optical disk drive, etc. The removable storage drive [608] may be compatible with a removable storage unit [610] such that it can read from and/or write to the removable storage unit [610]. Removable storage unit [610] may include a computer usable storage medium having stored therein computer-readable program codes or instructions and/or computer readable data. For example, removable storage unit [610] may store patient data. Example of removable storage unit [610] are well known in the art, including, but not limited to, floppy disks, magnetic tapes, optical disks, and the like. The at least one memory module [606] may also include a hard disk drive [612], which can be used to store computer readable program codes or instructions, and/or computer readable data.
[00112] In addition, as shown in Fig.l, the at least one memory module [606] may further include an interface [614] and a removable storage unit [616] that is compatible with interface [614] such that software, computer readable codes or instructions can be transferred from the removable storage unit [616] into computer system [600] . Examples of interface [614] and removable storage unit [616] pairs include, e.g., removable memory chips (e.g., EPROMs or PROMs) and sockets associated therewith, program cartridges and cartridge interface, and the like. Computer system [600] may also include a secondary memory module [618], such as random access memory (RAM).
[00113] Computer system [600] may include at least one processor module [602] . It should be understood that the at least one processor module [602] may consist of any number of devices. The at least one processor module [602] may include a data processing device, such as a microprocessor or microcontroller or a central processing unit. The at least one processor module
[602] may include another logic device such as a DMA (Direct Memory Access) processor, an integrated communication processor device, a custom VLSI (Very Large Scale Integration) device or an ASIC (Application Specific Integrated Circuit) device. In addition, the at least one processor module [602] may include any other type of analog or digital circuitry that is designed to perform the processing functions described herein.
[00114] As shown in FIG.6, in computer system [600], the at least one memory module [606], the at least one processor module [602], and secondary memory module [618] are all operably linked together through communication infrastructure [620] , which may be a
communications bus, system board, cross-bar, etc.). Through the communication infrastructure [620] , computer program codes or instructions or computer readable data can be transferred and exchanged. Input interface [626] may operably connect the at least one input module [626] to the communication infrastructure [620] . Likewise, output interface [622] may operably connect the at least one output module [624] to the communication infrastructure [620] .
[00115] The at least one input module [630] may include, for example, a keyboard, mouse, touch screen, scanner, and other input devices known in the art. The at least one output module [624] may include, for example, a display screen, such as a computer monitor, TV monitor, or the touch screen of the at least one input module [630]; a printer; and audio speakers. Computer system [600] may also include, modems, communication ports, network cards such as Ethernet cards, and newly developed devices for accessing intranets or the internet.
[00116] The at least one memory module [606] may be configured for storing patient data entered via the at least one input module [630] and processed via the at least one processor module [602] . Patient data relevant to the present invention may include expression level, activity level, copy number and/or sequence information for PTEN and/or a CCG. Patient data relevant to the present invention may also include clinical parameters relevant to the patient's disease. Any other patient data a physician might find useful in making treatment decisions/recommendations may also be entered into the system, including but not limited to age, gender, and race/ethnicity and lifestyle data such as diet information. Other possible types of patient data include symptoms currently or previously experienced, patient's history of illnesses, medications, and medical procedures.
[00117] The at least one memory module [606] may include a computer-implemented method stored therein. The at least one processor module [602] may be used to execute software or computer-readable instruction codes of the computer-implemented method. The computer- implemented method may be configured to, based upon the patient data, indicate whether the patient has an increased likelihood of recurrence, progression or response to any particular treatment, generate a list of possible treatments, etc.
[00118] In certain embodiments, the computer-implemented method may be configured to identify a patient as having or not having an increased likelihood of recurrence or progression. For example, the computer-implemented method may be configured to inform a physician that a particular patient has an increased likelihood of recurrence. Alternatively or additionally, the computer-implemented method may be configured to actually suggest a particular course of treatment based on the answers to/results for various queries.
[00119] FIG.7 illustrates one embodiment of a computer-implemented method [700] of the invention that may be implemented with the computer system [600] of the invention. The method [700] begins with one of three queries ([710], [711], [712]), either sequentially or substantially simultaneously. If the answer to/result for any of these queries is "Yes" [720], the method concludes [730] that the patient has an increased likelihood of recurrence. If the answer to/result for all of these queries is "No" [721], the method concludes [731] that the patient does not have an increased likelihood of recurrence. The method [700] may then proceed with more queries, make a particular treatment recommendation ([740], [741]), or simply end.
[00120] When the queries are performed sequentially, they may be made in the order suggested by FIG.7 or in any other order. Whether subsequent queries are made can also be dependent on the results/answers for preceding queries. In some embodiments of the method illustrated in FIG.7, for example, the method asks about clinical parameters [712] first and, if the patient has one or more clinical parameters identifying the patient as at increased risk for recurrence then the method concludes such [730] or optionally confirms by querying CCG status, while if the patient has no such clinical parameters then the method proceeds to ask about CCG status [711] . Optionally, if CCG status is not elevated, then the method may continue to ask about PTEN status
[710] . As mentioned above, the preceding order of queries may be modified. In some embodiments an answer of "yes" to one query {e.g., [712]) prompts one or more of the remaining queries to confirm that the patient has increased risk of recurrence.
[00121] In some embodiments, the computer-implemented method of the invention
[700] is open-ended. In other words, the apparent first step [710, 711, and/or 712] in FIG.7 may actually form part of a larger process and, within this larger process, need not be the first step/query. Additional steps may also be added onto the core methods discussed above. These additional steps include, but are not limited to, informing a health care professional (or the patient itself) of the conclusion reached; combining the conclusion reached by the illustrated method [700] with other facts or conclusions to reach some additional or refined conclusion regarding the patient's diagnosis, prognosis, treatment, etc.; making a recommendation for treatment (e.g., "patient should/should not undergo radical prostatectomy"); additional queries about additional biomarkers, clinical parameters, or other useful patient information (e.g., age at diagnosis, general patient health, etc.).
[00122] Regarding the above computer-implemented method [700], the answers to the queries may be determined by the method instituting a search of patient data for the answer. For example, to answer the respective queries [710, 711, 712], patient data may be searched for PTEN status (e.g., PTEN IHC or mutation screening), CCG status (e.g., CCG expression level data), or clinical parameters (e.g., Gleason score, nomogram score, etc.). If such a comparison has not already been performed, the method may compare these data to some reference in order to determine if the patient has an abnormal (e.g., elevated, low, negative) status. Additionally or alternatively, the method may present one or more of the queries [710, 711, 712] to a user (e.g., a physician) of the computer system [100] . For example, the questions [710, 711, 712] may be presented via an output module [624] . The user may then answer "Yes" or "No" via an input module [630] . The method may then proceed based upon the answer received. Likewise, the conclusions [730, 731] may be presented to a user of the computer-implemented method via an output module [624] .
[00123] Thus in some embodiments the invention provides a method comprising: accessing information on a patient's CCG status, clinical parameters and/or PTEN status stored in a computer-readable medium; querying this information to determine at least one of whether a sample obtained from the patient shows increased expression of at least one CCG, whether the patient has a recurrence-associated clinical parameter, and/or whether the patient has a low/negative PTEN status,; outputting [or displaying] the sample's CCG expression status, the patient's recurrence-associated clinical parameter status, and/or the sample's PTEN status. As used herein in the context of computer-implemented embodiments of the invention, "displaying" means communicating any information by any sensory means. Examples include, but are not limited to, visual displays, e.g., on a computer screen or on a sheet of paper printed at the command of the computer, and auditory displays, e.g., computer generated or recorded auditory expression of a patient's genotype. [00124] As discussed at length above, recurrence-associated clinical parameters or
PTEN status combined with elevated CCG status indicate a significantly increased likelihood of recurrence. Thus some embodiments provide a computer-implemented method of determining whether a patient has an increased likelihood of recurrence comprising accessing information on a patient's PTEN status {e.g., from a tumor sample obtained from the patient) or clinical parameters and CCG status {e.g., from a tumor sample obtained from the patient) stored in a computer-readable medium; querying this information to determine at least one of whether the patient has a
low/negative PTEN status or whether the patient has a recurrence-associated clinical parameter; querying this information to determine whether a sample obtained from the patient shows increased expression of at least one CCG; outputting (or displaying) an indication that the patient has an increased likelihood of recurrence if the patient has a low/negative PTEN status or a recurrence- associated clinical parameter and the sample shows increased expression of at least one CCG. Some embodiments further comprise displaying PTEN, clinical parameters (or their values) and/or the CCGs and their status (including, e.g., expression levels), optionally together with an indication of whether the PTEN or CCG status and/or clinical parameter indicates increased likelihood of risk.
[00125] The practice of the present invention may also employ conventional biology methods, software and systems. Computer software products of the invention typically include computer readable media having computer-executable instructions for performing the logic steps of the method of the invention. Suitable computer readable medium include floppy disk, CD- ROM/DVD/DVD-ROM, hard-disk drive, flash memory, ROM/RAM, magnetic tapes and etc. Basic computational biology methods are described in, for example, Setubal et ah, INTRODUCTION TO COMPUTATIONAL BIOLOGY METHODS (PWS Publishing Company, Boston, 1997); Salzberg et al. (Ed.), COMPUTATIONAL METHODS IN MOLECULAR BIOLOGY, (Elsevier, Amsterdam, 1998); Rashidi & Buehler, BIOINFORMATICS BASICS : APPLICATION IN BIOLOGICAL SCIENCE AND MEDICINE (CRC Press, London, 2000); and Ouelette & Bzevanis, BIOINFORMATICS: A PRACTICAL GUIDE FOR
ANALYSIS OF GENE AND PROTEINS (Wiley & Sons, Inc., 2ND ed., 2001); see also, U.S. Pat. No.
6,420, 108.
[00126] The present invention may also make use of various computer program products and software for a variety of purposes, such as probe design, management of data, analysis, and instrument operation. See U.S. Pat. Nos. 5,593,839; 5,795,716; 5,733,729; 5,974, 164;
6,066,454; 6,090,555; 6, 185,561 ; 6, 188,783; 6,223, 127; 6,229,91 1 and 6,308, 170. Additionally, the present invention may have embodiments that include methods for providing genetic information over networks such as the Internet as shown in U.S. Ser. Nos. 10/197,621 (U.S. Pub. No.
20030097222); 10/063,559 (U.S. Pub. No. 20020183936), 10/065,856 (U.S. Pub. No.
20030100995); 10/065,868 (U.S. Pub. No. 20030120432); 10/423,403 (U.S. Pub. No.
20040049354).
[00127] Techniques for analyzing such expression, activity, and/or sequence data
(indeed any data obtained according to the invention) will often be implemented using hardware, software or a combination thereof in one or more computer systems or other processing systems capable of effectuating such analysis.
[00128] Thus one aspect of the present invention provides systems related to the above methods of the invention. In one embodiment the invention provides a system for determining gene expression in a tumor sample, comprising:
(1) a sample analyzer for determining the expression levels in a sample of a panel of genes including at least 4 CCGs, wherein the sample analyzer contains the sample, RNA from the sample and expressed from the panel of genes, or DNA synthesized from said RNA;
(2) a first computer program for
(a) receiving gene expression data on at least 4 test genes selected from the panel of genes,
(b) weighting the determined expression of each of the test genes with a predefined coefficient, and
(c) combining the weighted expression to provide a test value, wherein the combined weight given to said at least 4 or 5 or 6 CCGs is at least 40% (or 50%>, 60%, 70%, 80%, 90%, 95% or 100%) of the total weight given to the expression of all of said plurality of test genes; and optionally
(3) a second computer program for comparing the test value to one or more reference values each associated with a predetermined degree of risk of cancer.
In some embodiments at least 20%>, 50%>, 75%, or 90%> of said plurality of test genes are CCGs. In some embodiments the sample analyzer contains reagents for determining the expression levels in the sample of said panel of genes including at least 4 CCGs. In some embodiments the sample analyzer contains CCG-specific reagents as described below. [00129] In another embodiment the invention provides a system for determining gene expression in a tumor sample, comprising: (1) a sample analyzer for determining the expression levels of a panel of genes in a tumor sample including at least 4 CCGs, wherein the sample analyzer contains the tumor sample which is from a patient identified as having prostate cancer, breast cancer, brain cancer, bladder cancer, or lung cancer, R A from the sample and expressed from the panel of genes, or DNA synthesized from said RNA; (2) a first computer program for (a) receiving gene expression data on at least 4 test genes selected from the panel of genes, (b) weighting the determined expression of each of the test genes with a predefined coefficient, and (c) combining the weighted expression to provide a test value, wherein the combined weight given to said at least 4 or 5 or 6 CCGs is at least 40% (or 50%, 60%, 70%, 80%, 90%, 95% or 100%) of the total weight given to the expression of all of said plurality of test genes; and optionally (3) a second computer program for comparing the test value to one or more reference values each associated with a predetermined degree of risk of cancer recurrence or progression of the prostate cancer, breast cancer, brain cancer, bladder cancer, or lung cancer. In some embodiments at least 20%>, 50%>, 75%, or 90%> of said plurality of test genes are CCGs. In some embodiments the system comprises a computer program for determining the patient's prognosis and/or determining (including quantifying) the patient's degree of risk of cancer recurrence or progression based at least in part on the comparison of the test value with said one or more reference values.
[00130] In some embodiments, the system further comprises a display module displaying the comparison between the test value and the one or more reference values, or displaying a result of the comparing step, or displaying the patient's prognosis and/or degree of risk of cancer recurrence or progression.
[00131] In a preferred embodiment, the amount of RNA transcribed from the panel of genes including test genes (and/or DNA reverse transcribed therefrom) is measured in the sample. In addition, the amount of RNA of one or more housekeeping genes in the sample (and/or DNA reverse transcribed therefrom) is also measured, and used to normalize or calibrate the expression of the test genes, as described above.
[00132] In some embodiments, the plurality of test genes includes at least 2, 3 or 4
CCGs, which constitute at least 50%>, 75% or 80%> of the plurality of test genes, and preferably 100% of the plurality of test genes. In some embodiments, the plurality of test genes includes at least 5, 6 or 7, or at least 8 CCGs, which constitute at least 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80% or 90% of the plurality of test genes, and preferably 100% of the plurality of test genes. Thus in some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least
3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises the top 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40 or more CCGs listed in Table 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 of the following genes: ASPM, BIRC5, BUB IB, CCNB2, CDC2, CDC20, CDCA8, CDKN3, CENPF, DLGAP5, FOXM1, KIAA0101, KIF11, KIF2C, KIF4A, MCM10, NUSAP1, PRC1, RACGAP1, and TPX2. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, or ten or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, or 1 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, or nine or all of gene numbers 2 & 3, 2 to
4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, or 2 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4,
5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, or eight or all of gene numbers 3 & 4, 3 to 5, 3 to 6, 3 to 7, 3 to 8, 3 to 9, or 3 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, or seven or all of gene numbers 4 & 5, 4 to 6, 4 to 7, 4 to 8, 4 to 9, or 4 to 10 of any of Tables 9, 11, 23, 24, or 25. In some embodiments the plurality of test genes comprises at least some number of CCGs (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more CCGs) and this plurality of CCGs comprises any one, two, three, four, five, six, seven, eight, nine, 10, 11, 12, 13, 14, or 15 or all of gene numbers 1 & 2, 1 to 3, 1 to 4, 1 to 5, 1 to 6, 1 to 7, 1 to 8, 1 to 9, 1 to 10, 1 to 11, 1 to 12, 1 to 13, 1 to 14, or 1 to 15 of any of Tables 9, 11, 23, 24, or 25. [00133] In some other embodiments, the plurality of test genes includes at least 8, 10,
12, 15, 20, 25 or 30 CCGs, which constitute at least 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%) or 90%) of the plurality of test genes, and preferably 100% of the plurality of test genes.
[00134] The sample analyzer can be any instrument useful in determining gene expression, including, e.g., a sequencing machine (e.g., Illumina HiSeq™, Ion Torrent PGM, ABI SOLiD™ sequencer, PacBio RS, Helicos Heliscope™, etc.), a real-time PCR machine (e.g., ABI 7900, Fluidigm BioMark™, etc.), a microarray instrument, etc.
[00135] In one aspect, the present invention provides methods of treating a cancer patient comprising obtaining CCG status information (e.g., the CCGs in Table 1 or Panels A through G), and recommending, prescribing or administering a treatment for the cancer patient based on the CCG status. In some embodiments, the method further includes obtaining clinical parameter information, and/or obtaining PTEN status information from a sample from the patient and treating the patient with a particular treatment based on the CCG status, clinical parameter and/or PTEN status information. For example, the invention provides a method of treating a cancer patient comprising:
(1) determining the status of at least one CCG;
(2) determining the status of at least on clinical parameter;
(3) determining the status of PTEN in a sample obtained from the patient; and
(4) recommending, prescribing or administering either
(a) an active (including aggressive) treatment if the patient has at least one of increased expression of the CCG, recurrence-associated clinical parameter, or low/negative PTEN status, or
(b) a passive (or less aggressive) treatment if the patient has none of increased expression of the CCG, recurrence-associated clinical parameter, or low/negative PTEN status.
[00136] Whether a treatment is aggressive or not will generally depend on the cancer- type, the age of the patient, etc. For example, in breast cancer adjuvant chemotherapy is a common aggressive treatment given to complement the less aggressive standards of surgery and hormonal therapy. Those skilled in the art are familiar with various other aggressive and less aggressive treatments for each type of cancer. "Active treatment" in prostate cancer is well-understood by those skilled in the art and, as used herein, has the conventional meaning in the art. Generally speaking, active treatment in prostate cancer is anything other than "watchful waiting." Active treatment currently applied in the art of prostate cancer treatment includes, e.g. , prostatectomy, radiotherapy, hormonal therapy (e.g., GnRH antagonists, GnRH agonists, antiandrogens), chemotherapy, high intensity focused ultrasound ("HIFU"), etc. Each treatment option carries with it certain risks as well as side-effects of varying severity, e.g., impotence, urinary incontinence, etc. Thus it is common for doctors, depending on the age and general health of the man diagnosed with prostate cancer, to recommend a regime of "watchful-waiting."
[00137] "Watchful-waiting," also called "active surveillance," also has its
conventional meaning in the art. This generally means observation and regular monitoring without invasive treatment. Watchful-waiting is sometimes used, e.g. , when an early stage, slow-growing prostate cancer is found in an older man. Watchful-waiting may also be suggested when the risks of surgery, radiation therapy, or hormonal therapy outweigh the possible benefits. Other treatments can be started if symptoms develop, or if there are signs that the cancer growth is accelerating (e.g., rapidly rising PSA, increase in Gleason score on repeat biopsy, etc.).
[00138] Although men who choose watchful-waiting avoid the risks of surgery and radiation, watchful-waiting carries its own risks, e.g., increased risk of metastasis. For younger men, a trial of active surveillance may not mean avoiding treatment altogether, but may reasonably allow a delay of a few years or more, during which time the quality of life impact of active treatment can be avoided. Published data to date suggest that carefully selected men will not miss a window for cure with this approach. Additional health problems that develop with advancing age during the observation period can also make it harder to undergo surgery and radiation therapy. Thus it is clinically important to carefully determine which prostate cancer patients are good candidates for watchful-waiting and which patients should receive active treatment.
[00139] Thus, the invention provides a method of treating a prostate cancer patient or providing guidance to the treatment of a patient. In this method, the status of at least one CCG (e.g., those in Table 1 or Panels A through G), at least one recurrence-associated clinical parameter, and/or the status of PTEN is determined, and (a) active treatment is recommended, initiated or continued if a sample from the patient has an elevated status for at least one CCG, the patient has at least one recurrence-associated clinical parameter, and/or low/negative PTEN status, or (b) watchful-waiting is recommended/initiated/ continued if the patient has neither an elevated status for at least one CCG, a recurrence-associated clinical parameter, nor low/negative PTEN status. In certain embodiments, CCG status, the clinical parameter(s) and PTEN status may indicate not just that active treatment is recommended, but that a particular active treatment is preferable for the patient (including relatively aggressive treatments such as, e.g., RP and/or adjuvant therapy).
[00140] In general, adjuvant therapy {e.g., chemotherapy, radiotherapy, HIFU, hormonal therapy, etc. after prostatectomy or radiotherapy) is not the standard of care in prostate cancer. According to the present invention, however, physicians may be able to determine which prostate cancer patients have particularly aggressive disease and thus should receive adjuvant therapy. Thus in one embodiment, the invention provides a method of treating a patient {e.g., a prostate cancer patient) comprising determining the status of at least one CCG {e.g. , those in Table 1 or Panels A through G), the status of at least one recurrence-associated clinical parameter, and/or the status of PTEN and initiating adjuvant therapy after prostatectomy or radiotherapy if a sample from the patient has an elevated status for at least one CCG, the patient has at least one recurrence- associated clinical parameter and/or the patient has low/negative PTEN status.
[00141] In one aspect, the invention provides compositions for use in the above methods. Such compositions include, but are not limited to, nucleic acid probes hybridizing to PTEN or a CCG (or to any nucleic acids encoded thereby or complementary thereto); nucleic acid primers and primer pairs suitable for amplifying all or a portion of PTEN or a CCG or any nucleic acids encoded thereby; antibodies binding immunologically to a polypeptide encoded by PTEN or a CCG; probe sets comprising a plurality of said nucleic acid probes, nucleic acid primers, antibodies, and/or polypeptides; microarrays comprising any of these; kits comprising any of these; etc. In some aspects, the invention provides computer methods, systems, software and/or modules for use in the above methods.
[00142] In some embodiments the invention provides a probe comprising an isolated oligonucleotide capable of selectively hybridizing to PTEN or at least one of the genes in Table 1 or Panels A through G. The terms "probe" and "oligonucleotide" (also "oligo"), when used in the context of nucleic acids, interchangeably refer to a relatively short nucleic acid fragment or sequence. The invention also provides primers useful in the methods of the invention. "Primers" are probes capable, under the right conditions and with the right companion reagents, of selectively amplifying a target nucleic acid {e.g., a target gene). In the context of nucleic acids, "probe" is used herein to encompass "primer" since primers can generally also serve as probes. [00143] The probe can generally be of any suitable size/length. In some embodiments the probe has a length from about 8 to 200, 15 to 150, 15 to 100, 15 to 75, 15 to 60, or 20 to 55 bases in length. They can be labeled with detectable markers with any suitable detection marker including but not limited to, radioactive isotopes, fluorophores, biotin, enzymes (e.g., alkaline phosphatase), enzyme substrates, ligands and antibodies, etc. See Jablonski et al., NUCLEIC ACIDS RES. (1986) 14:61 15-6128; Nguyen et al, BIOTECHNIQUES (1992) 13 : 1 16-123; Rigby et al, J. MOL. BIOL. (1977) 1 13 :237-251. Indeed, probes may be modified in any conventional manner for various molecular biological applications. Techniques for producing and using such oligonucleotide probes are conventional in the art.
[00144] Probes according to the invention can be used in the
hybridization/amplification/detection techniques discussed above. Thus, some embodiments of the invention comprise probe sets suitable for use in a microarray in detecting, amplifying and/or quantitating PTEN and/or a plurality of CCGs. In some embodiments the probe sets have a certain proportion of their probes directed to CCGs— e.g., a probe set consisting of 10%, 20%, 30%>, 40%>, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% probes specific for CCGs. In some embodiments the probe set comprises probes directed to at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 250, 300, 350, 400, 450, 500, 600, 700, or 800 or more, or all, of the genes in Table 1 or Panels A through G. Such probe sets can be incorporated into high-density arrays comprising 5,000, 10,000, 20,000, 50,000, 100,000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, or 1 ,000,000 or more different probes. In other embodiments the probe sets comprise primers (e.g., primer pairs) for amplifying nucleic acids comprising at least a portion of PTEN ox of one or more of the CCGs in Table 1 or Panels A through G.
[00145] In another aspect of the present invention, a kit is provided for practicing the prognosis of the present invention. The kit may include a carrier for the various components of the kit. The carrier can be a container or support, in the form of, e.g. , bag, box, tube, rack, and is optionally compartmentalized. The carrier may define an enclosed confinement for safety purposes during shipment and storage. The kit includes various components useful in determining the status of one or more CCGs and one or more housekeeping gene markers, using the above-discussed detection techniques. For example, the kit many include oligonucleotides specifically hybridizing under high stringency to mR A or cDNA of the genes in Table 1 or Panels A through G. Such oligonucleotides can be used as PCR primers in RT-PCR reactions, or hybridization probes. In some embodiments the kit comprises reagents (e.g., probes, primers, and or antibodies) for determining the expression level of a panel of genes, where said panel comprises at least 25%, 30%>, 40%>, 50%>, 60%, 75%, 80%, 90%, 95%, 99%, or 100% CCGs (e.g., CCGs in Table 1 or any of Panels A through G). In some embodiments the kit consists of reagents (e.g., probes, primers, and or antibodies) for determining the expression level of no more than 2500 genes, wherein at least 5, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 150, 200, 250, or more of these genes are CCGs (e.g., CCGs in Table 1 or any of Panels A through G).
[00146] The oligonucleotides in the detection kit can be labeled with any suitable detection marker including but not limited to, radioactive isotopes, fluorephores, biotin, enzymes (e.g., alkaline phosphatase), enzyme substrates, ligands and antibodies, etc. See Jablonski et al., Nucleic Acids Res., 14:6115-6128 (1986); Nguyen et al, Biotechniques, 13: 116-123 (1992); Rigby et al., J. Mol. Biol, 113:237-251 (1977). Alternatively, the oligonucleotides included in the kit are not labeled, and instead, one or more markers are provided in the kit so that users may label the oligonucleotides at the time of use.
[00147] In another embodiment of the invention, the detection kit contains one or more antibodies selectively immunoreactive with one or more proteins encoded by PTEN or one or more CCGs or optionally any additional markers. Examples include antibodies that bind
immunologically to PTEN or a protein encoded by a gene in Table 1 or Panels A through G.
Methods for producing and using such antibodies have been described above in detail.
[00148] Various other components useful in the detection techniques may also be included in the detection kit of this invention. Examples of such components include, but are not limited to, Taq polymerase, deoxyribonucleotides, dideoxyribonucleotides, other primers suitable for the amplification of a target DNA sequence, RNase A, and the like. In addition, the detection kit preferably includes instructions on using the kit for practice the prognosis method of the present invention using human samples.
[00149] In a specific embodiment,
EXAMPLE 1 [00150] The following cell cycle gene (CCG) signature was tested for predicting time to chemical recurrence after radical prostatectomy.
Figure imgf000053_0001
[00151] Mean mR A expression for the above 31 CCGs was tested on 440 prostate tumor FFPE samples using a Cox Proportional Hazard model in Splus 7.1 (Insightful, Inc., Seattle WA). The p-value for the likelihood ratio test was 3.98xl0"5.
[00152] The mean of CCG expression is robust to measurement error and individual variation between genes. In order to determine the optimal number of cell cycle genes for the signature, the predictive power of the mean was tested for randomly selected sets of from 1 to 30 of the CCGs listed above. This simulation showed that there is a threshold number of CCGs in a panel that provides significantly improved predictive power.
EXAMPLE 2
[00153] In a univariate analysis a set of 31 CCGs (Table 3) was found to be a significant predictor of biochemical recurrence (p-value = 1.8 x 10"9) after RP in prostate cancer patients. This signature was further evaluated to determine whether it added to an established clinical nomogram for prostate cancer recurrence (the Kattan- Stephenson nomogram). In summary, the nomogram was a highly significant predictor of recurrence (p-value 1.6 x 10"10) and, after adjusting for the nomogram, the CCG signature was a significant predictor of biochemical recurrence (p-value 4.8 x 10"5, Table 6).
Patients and Methods [00154] Eight hundred four consecutive RP patients were followed for a median of 9.5 years. The patient characteristics and the treatment outcomes of the entire cohort have been previously reported (Swanson et al., UROL ONCOL. (2007) 25: 110-114). Tissue blocks and/or slides from the final pathological evaluation with enough tissue for analysis were available for 430 patients. The cohort was divided randomly into 212 patients utilized for a training and 199 patient samples as a validation set.
[00155] Gene Expression (Statistical methods): Association between biochemical recurrence and CCG expression was evaluated using Cox PH models for time to recurrence. All of the p-values reported in this study were derived from a likelihood ratio test comparing the null model to the model containing the test variable. A set of 31 CCGs (Table 3, supra) was randomly selected. The assays were used to generate expression data from 212 patients in the training set. All of the expression data were generated in triplicate. The expression data were combined into a signature by calculating the mean expression level for 26 CCGs. Association between biochemical recurrence and CCG expression was evaluated using Cox PH models for time to recurrence.
[00156] Sample Preparation and Study Design: RNA was isolated from FFPE tumor sections derived from 411 prostate cancer patients treated with RP. Representative ΙΟμιη thick tumor sections were used to isolate RNA. When necessary, a pathologist guided macro- or microdissection of the sample was used to enrich for tumor tissue before RNA isolation. None of the samples in the validation cohort were micro-dissected. Prior to any analysis, the cohort was split into 212 patients for initial characterization of the signature ("training set") and 199 patients for validation. The clinical characteristics of the training and validation cohort are listed on Table 5.
Table 5
Figure imgf000054_0001
Lymph node 8/212 (3.8%) 10/199 (5.0%) 0.632 Fisher's exact
Capsular 100/212 (47.2%) 101/199 (50.8%) 0.49 Fisher's exact
Through capsule 59/212 (27.8%) 66/199 (33.2%) 0.283 Fisher's exact
Positive margins 43/212 (20.3%) 57/199 (28.6%) 0.051 Fisher's exact
Post-RP Gleason score > 6 80/212 (37.7%) 66/199 (33.2%) 0.354 Fisher's exact
Post-RP nomogram, mean (sd) 137 (19.5) 138 (23.0) 0.424 t-test
Results
[00157] The CCG expression signature (Table 3, supra) was predictive of disease recurrence in a univariate analysis (p-value = 1.8 x 10~9, Table 6). The distribution of the signature score was skewed toward higher values (lower expression). The median value of signature score was used to divide the training cohort into two groups containing samples with either high or low CCG expression. The survival versus time for both groups is shown in FIG.2.
[00158] Predictive power of the CCG signature after accounting for clinical variables typically included in a post-surgical nomogram (the Kattan-Stephenson nomogram) was also evaluated. The nomogram was a highly significant predictor of recurrence (p-value 1.6xl0"10). After adjusting for the nomogram, the CCG signature was a significant predictor of biochemical recurrence (FIG.3) in the discovery cohort (p-value 0.03) and in the clinical validation cohort (p- value 4.8 x 10~5).
Table 6
Figure imgf000055_0001
* Mean of cell cycle gene expression with imputation of missing values, likelihood ratio test for Cox proportional hazards model.
[00159] To help understand the interaction between the nomogram and the CCG expression signature, a scatter plot comparing these predictors (FIG.4) was generated (light gray stars represent patients whose cancer recurred while black stars represent patients whose cancer did not). Analysis of the scatter plot by KM means divided the samples into three clusters based on nomogram score only. Subsequently, it was discovered that the clusters were based on well- understood clinical parameters. The patients in the lowest scoring cluster (116/117) had organ- confined disease. Patients in the middle scoring cluster (48/60) had at least one post-surgical parameter known to be associated with poor outcome (i.e., disease through the capsule, disease positive lymph nodes, and/or disease positive seminal vesicles) and low pre-surgical PSA
(<10ng/ml). Patients in the highest scoring cluster had at least one unfavorable post- surgical parameter and high pre-surgery PSA. Next, the patients in the low and medium scoring clusters were divided by the mean of the CCG score. Outcomes for patients in the highest scoring cluster are adequately predicted by the nomogram and, therefore, were not divided further. As a result, the scatter plot defines five patient groups with disease recurrence rates of 2%, 40% (for two groups), 65%, and 80%> (Table 7). The recurrence rate of all five groups versus time is shown in FIG.5.
Table 7
Figure imgf000056_0001
[00160] The scatter plot shown in FIG.4 suggests that there is a non-linear interaction between the CCG signature and the post-surgical nomogram. That is, the CCG signature is a better predictor in patients with low nomogram scores. Therefore, the study tested for statistical evidence of an interaction between these variables in a multivariate model for predicting disease recurrence (Table 8). There was significant evidence for a favorable interaction in both training and validation studies. Including the interaction term in the model dramatically improved the prognostic significance of the CCG signature after adjusting for the nomogram (p-values of 0.0015 in training and 1.2xl0-8 in validation cohort).
Table 8
Figure imgf000056_0002
CCG
199 none NA 1.8xl0"9 2.68 signature
CCG
197 nomogram 0.0001 1.28xl0~8
signature
EXAMPLE 3
[00161] The following study aimed at determining the optimal number of CCGs to include in the signature. As mentioned above, CCG expression levels are correlated to each other so it was possible that measuring a small number of genes would be sufficient to predict disease outcome. In fact, single CCGs from the 31 -gene set in Table 3 (Panel C) add significantly to the Kattan- Stephenson nomogram, as shown in Table 9 below (after adjustment for the nomogram and an interaction term between the nomogram and CCG expression):
Table 9
Figure imgf000057_0001
* p-value for likelihood ratio test of full (post-RP nomogram score + cell cycle expression + nomogramxell cycle) vs reduced (post-RP nomogram score only) CoxPH model of time-to- recurrence.
[00162] To evaluate how smaller subsets of the larger CCG set (i.e., smaller CCG panels) performed, the study also compared how well the signature predicted outcome as a function of the number of CCGs included in the signature (FIG.1). Time to chemical recurrence after prostate surgery was regressed on the CCG mean adjusted by the post-RP nomogram score. Data consist of TLDA assays expressed as deltaCT for 199 FFPE prostate tumor samples and 26 CCGs and were analyzed by a CoxPH multivariate model. P-values are for the likelihood ratio test of the full model (nomogram + cell cycle mean including interaction) vs the reduced model (nomogram only). As shown in Table 10 below and FIG.l, small CCG signatures (e.g., 2, 3, 4, 5, 6 CCGS, etc.) add significantly to the Kattan- Stephenson nomogram:
Table 10
Figure imgf000058_0001
* For 1000 randomly drawn subsets, size 1 through 6, of cell cycle genes.
EXAMPLE 4
[00163] The aim of this experiment was to evaluate the association between PTEN mutations and biochemical recurrence in prostate cancer patients after radical prostatectomy.
Somatic mutations in PTEN were found to be significantly associated with recurrence, and importantly, it added prognostic information beyond both the established clinical nomogram for prostate cancer recurrence (the Kattan-Stephenson nomogram) and the CCG signature score (described in Examples 1 & 2, supra).
Patients and Methods
[00164] Eight hundred four consecutive RP patients were followed for a median of 9.5 years. The patient characteristics and the treatment outcomes of the entire cohort have been previously reported (Swanson et al., UROL. ONCOL. (2007) 25 : 1 10-1 14). Tissue blocks and/or slides from the final pathological evaluation with enough tissue for analysis were available for 430 patients. Of these, 191 were selected for PTEN mutation screening based on the amount of available tumor. [00165] Genomic DNA was isolated from the FFPE tumor samples for mutation screening of PTEN using the QIAamp DNA FFPE Tissue kit (Qiagen, Valencia, CA) according to the kit protocol. The FFPE slides were first stained with hematoxylin and eosin and examined by a pathologist to identify the tumor region. After deparaffinization, tumor tissue was cut out from the slides by a razor blade. For a few samples dissection was aided by laser capture microscopy (LCM), owing to the dispersion of the tumor cells
[00166] Mutations were detected by designing sequencing primers to interrogate the
PTEN genomic sequence. The primers contained Ml 3 forward and reverse tails to facilitate sequencing. After amplification, DNA sequence was determined on a Mega BASE 4500 (GE healthcare) using dye -primer chemistry as described in Frank et al., J. CLIN. ONCOL. (2002) 20: 1480- 1490. Due to the technical difficulties associated with sequencing DNA derived from FFPE material, each mutation was detected by at least two independent amplification and sequencing reactions.
[00167] Statistical methods: Unless otherwise specified, the association between biochemical recurrence and PTEN mutations was evaluated using Cox PH models for time to recurrence. The resultant p-values were derived from a likelihood ratio test comparing the null model to the model containing the test variable. In this example (Example 4), the CCG signature was derived from 26 CCGs (Panel D in Table 2, supra). All of the expression data were generated in triplicate. The expression data were combined into a signature by calculating the mean expression level for 26 CCGs. The clinical data were the variables included in the Kattan- Stephenson nomogram.
Results
[00168] PTEN mutations were found in 13 individuals (13/191). In this subset of 191 patients, PTEN was a significant predictor of biochemical recurrence (p-value = 0.031). The recurrence rate in mutation carriers was 69% (9/13) compared to 36% (64/178) in non-mutant patients. The difference in recurrence rate is also significant using a Fisher's exact test (p-value = 0.034). In the subset of patients with clinical parameter data, CCG signature score, and PTEN mutations, PTEN status was a significant predictor of biochemical recurrence after adjusting for both clinical parameters and CCG signature (p-value 0.024). Finally, the combination of PTEN mutation with CCG signature was a better predictor of outcome after adjusting for clinical parameters than using the CCG signature after adjusting for clinical parameters (p -value = 0.0002 for the combination compared to 0.0028 for CCG only). These results show that PTEN mutations provide information about the likelihood of recurrence that is uncorrected with either clinical parameters or CCG signature, and that using all three parameters to evaluate recurrence risk provides a more accurate estimate of recurrence probability than previously possible.
EXAMPLE 5
[00169] This Example describes further studies to validate and refine some
embodiments of the CCG signatures of the invention.
Patients and Methods
[00170] Eight hundred four consecutive radical prostatectomy patients were followed for a median of 9.5 years. The median age was 67 years. The clinical stage was Tl 34%, T2 66% and T3 <1%. The median preoperative PSA was 6.6 ng/ml with 72% < 10 ng/ml and 28% >10 ng/ml. The specimens were inked and clinical parameters were recorded as to positive bladder neck or urethral margin, invasion into the capsule, extension through the capsule, positive margins and the involvement of the seminal vesicles. Biochemical recurrence was defined as a PSA >0.3 ng/ml. For this study we had access to clinical data on 690 patients. Tissue blocks and/or slides from the final pathological evaluation with enough tissue for analysis were available for 442 patients. The cohort was divided into 195 patients for a training cohort, and 247 patients for validation.
Selection of genes
[00171] Assays of 126 CCGs and 47 HK (housekeeping) genes were run against 96 commercially obtained, anonymous prostate tumor FFPE samples without outcome or other clinical data. The working hypothesis was that the assays would measure with varying degrees of accuracy the same underlying phenomenon (cell cycle proliferation within the tumor for the CCGs, and sample concentration for the HK genes). Assays were ranked by the Pearson's correlation coefficient between the individual gene and the mean of all the candidate genes, that being the best available estimate of biological activity. Results for the correlation of each of the 126 CCGs to the mean are reported in Table 23. Not including CCGs with low average expression, or assays that produced sample failures, approximately half the CCGs had correlations less than 0.58, and a quarter of the HK genes had correlations less than 0.95. These assays were interpreted as not reflecting the underlying phenomenon and were eliminated, leaving a subset of 56 CCGs (Panel G) and 36 HK candidate genes (Tables 11 and 12). Correlation coefficients were recalculated on this subset, and the final selection was made from the ranked list.
Table 11: Complete list of evaluated CCGs ("Panel G")
Figure imgf000061_0001
Table 12: List of 15 housekeeping (HK) genes
Figure imgf000061_0002
CLTC 0.981
TXNL1 0.98
PSMA1 0.98
RPL8 0.98
MMADHC 0.979
RPL13A;
0.979
LOC728658
PPP2CA 0.978
MRFAP1 0.978
Gene Expression
[00172] Total RNA was extracted from representative 5 μΜ thick FFPE tumor sections. The samples were de-paraffmized using a xylene bath and subsequently hydrated in graded series of ethanol baths. Afterward, the tumor region was dissected from the slide using a razor blade according to the pathologist instructions. Alternatively, the tumor region was dissected directly into an eppendorf tube and the paraffin was removed using xylene and washed with ethanol. After, samples were treated overnight with proteinase K digestion at 55°C. Total RNA was extracted using either RNeasy FFPE or miRNeasy (Qiagen) as described by the manufacturer (with the only exception being the extended proteinase K digestion described above). Isolated total RNA was treated with DNase I (Sigma) prior to cDNA synthesis. Subsequently, we employed the High- capacity cDNA Archive Kit (Applied Biosystems) to convert total RNA into single strand cDNA as described by the manufacturer. A minimum of 200ng RNA was required for the RT reaction.
[00173] Prior to measuring expression levels, the cDNA was pre-amplified with a pooled reaction containing TaqMan™ assays. Pre-amplification reaction conditions were: 14 cycles of 95°C for 15sec and 60°C for 4 minutes. The first cycle was modified to include a 10 minute incubation at 95°C. The amplification reaction was diluted 1 :20 using the IX TE buffer prior to loading on TaqMan™ Low Density Arrays (TLDA, Applied Biosystems) to measure gene expression.
CCG score
[00174] The CCG score is calculated from RNA expression of 31 CCGs (Panel F) normalized by 15 housekeeper genes (HK). The relative numbers of CCGs (31) and HK genes (15) were optimized in order to minimize the variance of the CCG score. The CCG score is the unweighted mean of CT values for CCG expression, normalized by the unweighted mean of the HK genes so that higher values indicate higher expression. One unit is equivalent to a two-fold change in expression. Missing values were imputed using the mean expression for each gene determined in the training set using only good quality samples. The CCG scores were centered by the mean value, again determined in the training set.
[00175] A dilution experiment was performed on four of the commercial prostate samples to estimate the measurement error of the CCG score (se = 0.10) and the effect of missing values. It was found that the CCG score remained stable as concentration decreased to the point of 5 failures out of the total 31 CCGs. Based on this result, samples with more than 4 missing values were not assigned a CCG score.
[00176] The CCG score threshold for determining low-risk was based on the lowest
CCG score of recurrences in the training set. The threshold was then adjusted downward by 1 standard deviation in order to optimize the negative predictive value of the test.
Model of clinical risk
[00177] A Cox proportional hazards model was used to summarize the available clinical parameter data and estimate the prior clinical risk of biochemical recurrence for each patient. The data set consisted of 195 cases from the training set and 248 other cases with clinical parameter information but insufficient sample to measure R A expression. Univariate tests were performed on clinical parameters known to be associated with outcome (see Table 13 below). Non-significant parameters were excluded from the model. A composite variable was created for organ-confined disease, with invasion defined as surgical margins, extracapsular extension, or involvement of any of seminal vesicles, bladder neck/urethral margins, or lymph nodes. The composite variable for organ- confined disease proved more significant in the model than any of its five components, some of which were inter-correlated or not prevalent. Model fitting was performed using the AIC criteria for post-operative covariates.
Table 13: Univariate analysis of clinical parameters and association with biochemical recurrence
Figure imgf000063_0001
CAPSULAR l . lxlO"9 194 443 0.438
ETHNICITY
0.6741 416 439 0.948
(WHITE)
LYMPHNOD 0.0009 33 443 0.074
MARG.POS 6.1xl0"u 83 443 0.187
PATHGLEA 6.7xl0"16 NA 443 NA
PATHGRAD 2.4xl0~u NA 443 NA
PATHSTAG 3.1xl0"15 NA 443 NA
PRE.PSA.LOG10 6.2xl0~12 NA 443 NA
SEM.VES 3.0xl0"8 56 443 0.126
SURGERY .YEAR 0.0803 NA 443 NA
THRU.CAP 1.3xl0"10 114 443 0.257
Cox PH p-value for likelihood ratio test
[00178] The final model (i.e., nomogram) has binary variables for organ-confined disease and Gleason score less than or equal to 6, and a continuous variable for logarithmic PSA (Table 14). This model includes all of the clinical parameters incorporated in the post-RP nomogram (i.e., Kattan- Stephenson nomogram) except for Year of RP and the two components of the Gleason score. The distribution of prior clinical risk shows three distinct nodes (Figure 8). K- means clustering with 3 centers was used to set the threshold for the low-risk cluster, which comprises approximately 50% of the sample.
Table 14: Clinical Model
Figure imgf000064_0001
Statistical Analysis [00179] Clinical parameters were compared between the training and validation sets using the Student's t-test for continuous parameters and Fisher's exact test for categorical parameters. The prior clinical risk of patients for biochemical recurrence after surgery was estimated by a post-RP nomogram score summarizing 7 covariates. K-means clustering of the nomogram score was used to categorize patients as low or high prior clinical risk. Expression data were expressed as the CT (the PCR cycle at which the fluorescence intensity exceeds a
predetermined threshold) of each CCG normalized by the mean of the 15 housekeeper genes (Table 12 above).
[00180] Poor quality samples were excluded from analysis to eliminate poor quality samples or dubious readings without compromising the integrity of the signature by inadvertently excluding samples with low CCG expression. Accordingly, the thresholds for cleaning or filtering the data were set conservatively. Mean expression levels of the HK genes for each sample, which were higher than those of the CCGs, were used to identify poor quality samples. Technical metrics for the amplification efficiency and excessively high standard deviations of replicates were used to identify unreliable CT measurements. No failures of HK genes, and no more than 1 failure out of 3 replicates for CCGs, were allowed.
[00181] The association between biochemical recurrence and CCG expression after adjusting for clinical risk predicted by clinical parameters was evaluated using a Cox proportional hazards model for time-to-recurrence. The proportional hazards assumption of no time-dependence was tested for the full model of the CCG signature plus the binary clinical parameter score with an interaction term, and for the CCG signature only in the clinical risk subsets. It was not significant in either training or validation, indicating that there is no evidence for time-dependence. All of the p- values reported are from a likelihood ratio test comparing the reduced or null model to the model containing the test variable. Kaplan-Meier plots are used to show estimated survival probabilities for subsets of patients; however, p-values are from the Cox likelihood ratio test for the continuous values of the variable. All statistical analyses were performed in S+ Version 8.1.1 for Linux (TIBCO Spotfire) or R 2.9.0 (http://www.r-project.org).
Results
[00182] We isolated RNA from FFPE tumor sections derived from 442 prostate cancer patients treated with RP. The cohort was split into 195 patients for initial characterization of the signature ("training set") and 247 patients for validation. The clinical parameters of the training and validation cohort are listed in Table 15. There were no significant differences after adjusting for multiple comparisons.
Table 15: Clinical parameters of training and validation patient cohorts
Figure imgf000066_0001
[00183] To analyze the CCG signature for this study, we tested 126 CCGs on RNA derived from 96 prostate tumors (Table 11). The tumor samples were anonymous and not associated with clinical data. From this set of genes, we selected 31 genes (Panel F) for inclusion in our signature (Table 16). The genes were selected based on their technical performance, and by how well each gene correlated with the mean expression level of the entire CCG set, in the 96 anonymous samples.
Table 16: CCG Signature from Training Set (Panel F)
Figure imgf000067_0001
[00184] To evaluate the prognostic utility of the CCG signature, we generated expression data on 195 patients in the training set. Since the individual gene expression levels were correlated, we combined them into a signature score by calculating the mean expression for the entire set of 31 genes (Panel F), normalized by 15 housekeepers (Table 12). The CCG score distribution was centered at zero, and each score unit corresponds to a 2-fold change in expression level. Poor quality samples were identified by observing either low expression of housekeeping genes or an unacceptable number of CCG failures, and excluded from the analysis. After applying our exclusion rules, there were 140 samples available for analysis. Association between biochemical recurrence and CCG expression was evaluated using Cox PH models for time to recurrence. A high CCG expression value was predictive of disease recurrence in a univariate analysis (p-value= 0.01, Table 17).
[00185] Next, we evaluated the prognostic utility of the CCG signature after accounting for clinical parameters known to be associated with recurrence after RP. To account for clinical measures in our analysis, we created a model/nomogram that included preoperative PSA, Gleason score, and evidence of disease outside the prostate {i.e., any of either extracapsular extension, or positive post-surgical pathology on lymph nodes, margins, bladder neck, urethral margin or seminal vesicles). The model was optimized in 443 patients (Tables 13 & 14), including all patients for whom we had clinical data but were not in the validation set, and was a highly significant predictor of recurrence in the training cohort (p-value = 2.5 xlO 11). The distribution of the scores from the clinical model contained several modes (Figure 8), separating high- and low-risk patient groups. Therefore, the score was used subsequently as a binary variable (high or low risk). The low-risk cluster correlated with a consistent set of clinical parameters. Specifically, the vast majority (215/218) had organ-confined disease and Gleason score < 7. In addition, 80% had low pre-surgical PSA (< 10 ng/ml). Patients in the high-risk cluster (N=225) were more heterogeneous, but tended to have clinical characteristics known to be associated with poor outcome (e.g., Gleason > 6 and/or disease through the capsule).
[00186] Multivariate analysis of the training set incorporating our binary clinical model, showed evidence for a non-linear interaction between the expression signature and clinical parameters (Table 17). To help us understand the nature of this interaction, we generated a scatter plot comparing these predictors (Figure 8). As evident from the figure, the CCG score proved useful for evaluating recurrence risk in patients defined as low risk by clinical parameters. In fact, even after adjusting for the clinical model within the low risk patients, the CCG signature was a strong predictor of biochemical recurrence (p-value = 0.0071).
Table 17: Statistical Summary
Figure imgf000068_0001
Figure imgf000069_0001
[00187] We used our training data in the scatter plot to establish an optimized threshold score of -0.16 for the CCG signature (the mean CCG score is zero). Figure 12 shows this threshold applied to the 443 patients studied in this example. Forty percent of low-risk patients fall below this threshold, and it was selected so that there were no recurrences 10-years after RP (i.e., negative predictive value (NPV) of 100%). As a result of establishing threshold values for both the clinical model and CCG score, the scatter plot was divided into four sections with recurrence rates of 0% (low CCG) and 26% (high CCG) for low-risk patients; and 60% (low CCG) and 50% for high- risk patients.
[00188] Next, we generated CCG expression data on 247 patients in our validation cohort. Thirty-two samples were eliminated from further analysis according to the exclusion rules developed on the training cohort. Panel F was a significant predictor of biochemical recurrence in a univariate analysis (p-value = 5.8 x 10"8, Table 17). After adjusting for the binary clinical model, the CCG signature was highly predictive of recurrence in the validation cohort (p-value 8.3 xlO"7), and as in the training set, there was significant evidence for a non-linear interaction between variables. The CCG signature was informative across the entire spectrum of clinically defined risk (Table 17). In terms of validating the training results, the p-value for association between recurrence and CCG signature in low-risk patients was 1.9 xlO"4.
[00189] We applied the CCG threshold derived from our analysis of the training cohort to our validation data set (Figure 9). Low risk patients with CCG scores below the threshold had a 10-year predicted recurrence rate of 5% (equivalent to validated NPV of 0.95). Overall, the combination of CCG score and clinical parameters divided the cohort into four groups with 10 year predicted recurrence rates of 5%, 22%, 36%> and 70%> (Table 18). The predicted recurrence rate versus CCG score for patients in the validation cohort is shown in Figures 10 & 11.
Table 18: Summary of recurrence rates in validation cohort
defined by clinical risk and CCG score
Figure imgf000070_0001
[00190] We tested our validated threshold versus various definitions of low-risk patients (Table 19). The signature score was a significant prognostic indicator in a variety of low- risk clinical definitions, and depending on definition, generated a 10-year predicted recurrence rate of 0.05 to 0.10.
Table 19: NPV of CCG signature in other definitions of low-risk patients
Figure imgf000070_0002
Comment [00191] We have developed and validated a prognostic molecular signature for prostate cancer. The signature is based on measuring mR A expression levels of cell cycle genes (CCGs). By definition, expression of CCGs is regulated as a function of cell cycle stage. That is, they are turned on at specific cell cycle stages, so that actively growing cells have higher expression levels of CCG than quiescent cells. Presumably this fact underlies the signature's ability to predict cancer progression. Without wishing to be bound by theory, it is thought that by measuring the expression levels of CCG we are indirectly measuring the growth rate and inherent aggressiveness of the tumor, which ultimately impacts on the likelihood of prostate cancer recurrence after
prostatectomy.
[00192] There is an important distinction between this study and many others that have attempted to generate prognostic molecular signatures. Often, similar studies begin with a very large number of candidate biomarkers (sometimes exceeding 1000's of genes) that are then evaluated for association with a clinical phenotype of interest. This approach may at times suffer from inherent multiple testing which can make the significance of the derived signature uncertain. Here we have tested a single hypothesis: CCG would be prognostic in prostate cancer (in fact we selected genes based on their correlation with CCG expression, not based on association with recurrence). And since CCG expression is correlated, we combined the expression data into a predictive signature by determining the mean expression value of all the genes in the signature. The simplicity of this approach, biologically and computationally, supports the view that the central claim of this study is likely to be highly robust, and replicated in subsequent studies.
[00193] The CCG signature (Panel F) is independently predictive and adds
significantly to the predictive power of the clinical parameters typically employed to predict disease recurrence after surgery. This is true in both our training and validation cohorts.
[00194] The signature is immediately useful for defining the risk of patients who present with low-risk clinical parameters. Here, we essentially defined low-risk as Gleason <7, PSA <10 and organ-confined disease. The CCG signature score effectively subdivides the low-risk group into patients with very low recurrence rates (5%), and a higher risk of recurrence (22%) (Figure 9 & Table 18). This is the most dramatic effect of the molecular signature - accurately redefining the risk of patients previously defined as low-risk based on clinical parameters. It is noteworthy that within this patient subpopulation (i.e., patients defined as low-risk based on clinical parameters) clinical parameters are not particularly prognostic (see Table 17). Therefore as a diagnostic test, the signature could be useful for a large number of patients. In this study, nearly 60% of the cohort was characterized as low-risk and 40% of those are expected to have low CCG scores. Therefore, the CCG signature can predict indolent disease in a quarter of the patients who have previously been identified as high-risk (and therefore identified as candidates for radical prostatectomy). Finally, the validation data in particular suggests that the CCG signature may be useful for defining risk in all patients. Specifically, it helped to divide patients defined as high-risk according to clinical parameters into those with 30% and 70% recurrence rates (Table 18).
[00195] The combination of clinical parameters and CCG signature enables physicians to more accurately predict risk of surgical failure, and therefore, identify the appropriate course of therapeutic intervention. As we have shown, the signature dramatically improves the recurrence prediction for patients who present with general clinical parameters of non-aggressive disease (Table 19). Within this clinical subgroup, patients with low CCG scores would benefit from the absolute reassurance that no further treatment is indicated. Conversely, the high CCG group may warrant immediate intervention. Patients with unfavorable post-surgical clinical parameters benefit from adjuvant radiation therapy. Therefore the CCG signature should predict the efficacy of adjuvant radiation for patients with low-risk clinical characteristics and high CCG scores. In the validation cohort, patients with high CCG scores and disease beyond the prostate have a recurrence rate of 70%), which should clearly identify patients who are good candidates for adjuvant radiation. Thus the combination of clinical parameters and CCG signature clearly leads to more accurately defined patient risk, which should enable a more intelligent assessment of the need for further treatment.
EXAMPLE 6
[00196] Some of the CCGs panels described herein were further evaluated for their ability to prognose additional cancers. Panels C, D, and F were found to be prognostic to varying degrees in bladder, brain, breast, and lung cancer.
Methods
[00197] Gene expression and patient data was obtained from the following publicly available datasets: GSE7390 (Desmedt et al, CLIN. CANCER RES. (2007) 13:3207-14; PMID
17545524); GSE11121 (Schmidt et al, CANCER RES. (2008) 68:5405-13 ; PMID 18593943);
GSE8894 (Son et al; no publication); Shedden (Shedden et al, NATURE MED. (2008) 14:822; PMID 18641660); GSE4412 (Freije et al, CANCER RES. (2004) 64:6503-10; PMID 15374961); GSE4271 (Phillips et al, CANCER CELL (2006) 9: 157-73; PMID 16530701); GSE5287 (Als et al, CLIN. CANCER RES. (2007) 13:4407-14; PMID 17671123). Each of these datasets has an associated detailed description of the experimental procedures used in gathering expression and patient data. The expression microarrays used to generate each dataset are summarized below in Table 20.
Table 20
Figure imgf000073_0001
[00198] Expression data for each of the genes in Panels C, D and F was gathered from these datasets and the mean expression level for each Panel was determined for each patient, whose clinical outcome was known (e.g., recurrence, progression, progression-free survival, overall survival, etc.). CCG score is an average expression of the genes in a panel. If a gene is represented by more than one probe set on the array, the gene expression is an average expression of all the probe sets representing the gene. The association between CCG score and survival or disease recurrence was tested using univariate and multivariate Cox proportional hazard model.
Multivariate analysis was performed when relevant clinical parameters (grade in brain cancer, stage in lung cancer, NPI in breast cancer) were available.
Results
[00199] As shown in Table 21 below, each Panel, in univariate analysis, was a prognostic factor in each of the cancers analyzed.
Table 21
Figure imgf000073_0002
ER positive breast cancer GSE11121 1.2xl0~5 8.7xl0~6 1.5xl0~5
Lung adenocarcinoma GSE8894 2.0xl0~3 2.5xl0~3 5.6xl0~3
Lung adenocarcinoma Shedden 1.3xl0~7 2.6xl0~7 2.2xl0~7
Brain cancer GSE4412 3.2xl0~5 2.2xl0~5 9.0xl0~5
Brain cancer GSE4271 1.3xl0~3 l .OxlO"3 2.8xl0~4
Bladder cancer GSE5287 6.4xl0~2 5.0xl0~2 8.6xl0~2
[00200] As shown in Table 22 below, each Panel was also prognostic in multivariate analysis when combined with at least one clinical parameter (or nomogram).
Table 22
Figure imgf000074_0001
[00201] Tables 23 & 24 below provide rankings of select CCGs according to their correlation with the mean CCG expression.
Table 23
Figure imgf000074_0002
Figure imgf000075_0001
Table 24 Correl. Correl. Correl. w/
Gene Gene Gene Gene Gene Gene
w/ CCG w/ CCG CCG
# Symbol # Symbol # Symbol
mean mean mean
1 DLGAP5 0.931 12 C18orf24 0.885 22 TOP2A 0.852
2 ASPM 0.931 13 PLK1 0.879 23 KIF20A 0.851
3 KIF11 0.926 14 CDK 3 0.874 24 KIAA0101 0.839
4 BIRC5 0.916 15 RRM2 0.871 25 CDCA3 0.835
5 CDCA8 0.902 16 RAD51 0.864 26 ASF IB 0.797
6 CDC20 0.9 17 CEP55 0.862 27 CENPM 0.786
7 MCM10 0.899 18 ORC6L 0.86 28 TK1 0.783
8 PRC1 0.895 19 RAD54L 0.86 29 PBK 0.775
9 BUB IB 0.892 20 CDC2 0.858 30 PTTG1 0.751
10 FOXM1 0.889 21 CENPF 0.855 31 DTL 0.737
11 NUSAP1 0.888
[00202] Table 25 below provides a ranking of the CCGs in Panel F according to their relative predictive value in Example 5 (analogous to Table 9).
Table 25
Figure imgf000076_0001
[00203] Table 1 below provides a large, but not exhaustive, list of CCGs.
Table 1
Figure imgf000076_0002
MAPK13: mitogen-activated protein kinase 13 Hs.178695 AA157499 p38delta mR A=stress-activated protein kinase 4
CDC2: cell division cycle 2, Gl to S and G2 to M Hs.184572 AA598974
TOP2A: topoisomerase (DNA) II alpha (170kD) Hs.156346 AA504348
CENPE: centromere protein E (312kD) Hs.75573 AA402431 CENP-E=putative kinetochore motor that accumulates just befo
TOP2A: topoisomerase (DNA) II alpha (170kD) Hs.156346 AA026682
KPNA2: karyopherin alpha 2 (RAG cohort 1, importin alpha 1) Hs.159557 AA676460
FLJ10468: hypothetical protein FLJ10468 Hs.48855 N63744
CCNF: cyclin F Hs.1973 AA676797
DKFZp762E1312: hypothetical protein DKFZp762E1312 Hs.104859 T66935
CKS2: CDC2-Associated Protein CKS2 Hs.83758 AA292964
C20ORF1 : chromosome 20 open reading frame 1 Hs.9329 H73329
BUB1 : budding uninhibited by benzimidazoles 1 (yeast homolog) Hs.98658
AA430092 BUBl=putative mitotic checkpoint protein ser/thr kinase
TOP2A: **topoisomerase (DNA) II alpha (170kD) Hs.156346 AI734240
CKS2: CDC2-Associated Protein CKSl Hs.83758 AA010065 ckshs2=homolog of Cksl=p34Cdc28/Cdc2-associated protein
ARL6IP: ADP-ribosylation factor-like 6 interacting protein Hs.75249 H20558
L2DTL: L2DTL protein Hs.126774 R06900
STK15: ** serine/threonine kinase 15 Hs.48915 H63492 aurora/IPLl -related kinase
E2-EPF: ubiquitin carrier protein Hs.174070 AA464019
UBCH10: ubiquitin carrier protein E2-C Hs.93002 R80790
KNSL5: kinesin-like 5 (mitotic kinesin-like protein 1) Hs.270845 AA452513 Mitotic kinesin-like protein- 1
CENPF: centromere protein F (350/400kD, mitosin) Hs.77204 AA701455
CCNA2: cyclin A2 Hs.85137 AA608568 Cyclin A
CDC2: cell division cycle 2, Gl to S and G2 to M Hs.184572 AA278152 CDC2=Cell division control protein 2 homolog=P34 protein kin
HMMR: **hyaluronan-mediated motility receptor (RHAMM) Hs.72550 AA171715
KIAA0008: KIAA0008 gene product Hs.77695 AA262211
HSPC145: HSPC145 protein Hs.18349 R22949
FLJ20510: hypothetical protein FLJ20510 Hs.6844 N53214
Homo sapiens NUF2R mRNA, complete cds Hs.234545 AA421171 :
HSPC216: hypothetical protein Hs.13525 T87341
P37NB: 37 kDa leucine-rich repeat (LRR) protein Hs.155545 AA423870
CDC20:
CCNE1 : cyclin El Hs.9700 T54121
ESTs: Hs.221754 R84407
FLJ11252: hypothetical protein FLJ11252 Hs.23495 N30185 LOC51203: clone HQ0310 PRO0310pl Hs.279905 AA620485
FLJ10491 : hypothetical protein FL J 10491 Hs.274283 AA425404
K SL1 : kinesin-like 1 Hs.8878 AA504625
CENPA: centromere protein A (17kD) Hs.1594 AI369629
Homo sapiens, clone IMAGE:2823731, mRNA, partial cds Hs.70704 R96941 :
CDC6: CDC6 (cell division cycle 6, S. cerevisiae) homolog Hs.69563 H59203
Homo sapiens DNA helicase homolog (PIF1) mRNA, partial cds Hs.l 12160
AA464521 :
ESTs: Hs.48480 AA135809
TSN: translin Hs.75066 AA460927
KRNA2: karyopherin alpha 2 (RAG cohort 1, importin alpha 1) Hs.159557 AA489087
RRM2: ribonucleotide reductase M2 polypeptide Hs.75319 AA187351
ESTs: Hs.14119 AA204830
CCNB1 : cyclin Bl Hs.23960 R25788
GTSE1 : G-2 and S-phase expressed 1 Hs.122552 AI369284
C20ORF1 : chromosome 20 open reading frame 1 Hs.9329 AA936183
TACC3: transforming, acidic coiled-coil containing protein 3 Hs.l 04019 AA279990 JkRl mRNA downregulated upon T-cell activation
E2F1 : E2F transcription factor 1 Hs.96055 H61303
BUB IB: budding uninhibited by benzimidazoles 1 (yeast homolog), beta Hs.36708 AA488324
ESTs,: Weakly similar to CGHU7L collagen alpha 1 (III) chain precursor [H. sapiens] Hs.19322 AA088457
KIAA0074: KIAA0074 protein Hs. l 192 N54344
MPHOSPH1 : M-phase phosphoprotein 1 Hs.240 AA282935
ANLN: anillin (Drosophila Scraps homolog), actin binding protein Hs.62180 R12261
BIRC5: baculoviral IAP repeat-containing 5 (survivin) Hs.1578 AA460685
Survivin=apoptosis inhibitor=effector cell protease EPR-1
PTTG1 : pituitary tumor-transforming 1 Hs.252587 AA430032
KIAA0159: chromosome condensation-related SMC-associated protein 1 Hs.5719 AA668256
ESTs,: Weakly similar to OS-4 protein [H.sapiens] Hs.18714 W93120
HMMR: hyaluronan-mediated motility receptor (RHAMM) Hs.72550 R10284
DKFZp762E1312: hypothetical protein DKFZp762E1312 Hs.104859 AA936181
CKAP2: cytoskeleton associated protein 2 Hs.24641 T52152
RAMP: RA-regulated nuclear matrix-associated protein
SMAP: thyroid hormone receptor coactivating protein Hs.5464 AA481555
FLJ22624: hypothetical protein FLJ22624 Hs.166425 AA488791
CKS1 : CDC2-Associated Protein CKS1 Hs.77550 N48162
NEK2: NIMA (never in mitosis gene a)-related kinase 2 Hs.153704 W93379 72 MKI67: antigen identified by monoclonal antibody Ki-67
73 TTK: TTK protein kinase Hs.169840 AI337292
VEGFC: vascular endothelial growth factor C Hs.79141 H07899 vascular endothelial
74
growth factor related protein VRP
CDKN3 : cyclin-dependent kinase inhibitor 3 (CDK2-associated dual specificity
75 phosphatase) Hs.84113 AA284072 CIP2=Cdil=KAPl phosphatase=Gl/S cell cycle gene
76 Homo sapiens NUF2R mRNA, complete cds Hs.234545 R92435:
77 Homo sapiens cDNA FLJ10325 fis, clone NT2RM2000569 Hs.245342 AA235662:
78 HSPC145: HSPC145 protein Hs.18349 AA628867
79 HSU54999: LGN protein Hs.278338 W92010
80 FLJ20333: hypothetical protein FLJ20333 Hs.79828 R27552
81 KNSL2: kinesin-like 2 Hs.20830 N69491
82 ESTs: Hs.133294 AI053446
83 **ESTs: Hs.41294 H95819
84 SMTN: smoothelin Hs.149098 AA449234
85 FLJ23311 : hypothetical protein FLJ23311 Hs.94292 N73916
86 USF1 : upstream transcription factor 1 Hs.247842 AA719022
87 LOC51203: clone HQ0310 PRO0310pl Hs.279905 AA779949
88 ADH4: alcohol dehydrogenase 4 (class II), pi polypeptide Hs.1219 AA007395
89 ESTs: Hs.186579 AA960844
90 CCNB2: cyclin B2 Hs.194698 AA774665
Homo sapiens, Similar to gene rich cluster, C8 gene, clone MGC:2577, mRNA,
91
complete cds Hs.30114 AA634371 :
92 ESTs: Hs.99480 AA485454
Homo sapiens IRE lb mRNA for protein kinase/ribonuclease IREl beta, complete cds
93
Hs.l 14905 AA088442:
PCNA: proliferating cell nuclear antigen Hs.78996 AA450264 PCNA=proliferating
94
cell nuclear antigen
95 AA075920:
96 GTSE1 : G-2 and S-phase expressed 1 Hs.122552 AA449474
97 CKS1 : CDC2-Associated Protein CKS1 Hs.77550 AA278629
CDC25B: cell division cycle 25B Hs.153752 AA448659 cdc25B=M-phase inducer
98
phosphatase 2
ESTs,: Weakly similar to unnamed protein product [H. sapiens] Hs.99807 AA489023
99
Unknown UG Hs.99807 ESTs sc_id384
100 PCNA: proliferating cell nuclear antigen Hs.78996 H05891
101 LTBP3: **latent transforming growth factor beta binding protein 3 Hs.289019 R60197
Homo sapiens mRNA; cDNA DKFZp434D0818 (from clone DKFZp434D0818)
102
Hs.5855 N95578: 103 ESTs: Hs.126714 AA919126
104 CIT: citron (rho-interacting, serine/threonine kinase 21) Hs.15767 H10788
105 LBR: lamin B receptor Hs.152931 AA099136
106 E2F1 : E2F transcription factor 1 Hs.96055 AA424949
107 AA699928:
CDKN2C: cyclin-dependent kinase inhibitor 2C (pl8, inhibits CDK4) Hs.4854
108
N72115 pl8-INK6=Cyclin-dependent kinase 6 inhibitor
109 STK12: serine/threonine kinase 12 Hs.180655 H81023 ARK2=aurora-related kinase 2
110 ESTs: Hs. l 11471 AA682533
111 ESTs: Hs.44269 AA465090
MCM4: minichromosome maintenance deficient (S. cerevisiae) 4 Hs.154443
112
AA485983
PMSCL1 : ^^polymyositis/scleroderma autoantigen 1 (75kD) Hs.91728 AA458994
113
Cyclin A
MKI67: antigen identified by monoclonal antibody Ki-67 Hs.80976 AA425973 Ki67
114
(long type)
115 ESTs: Hs.133294 AI144063
CDC25B: cell division cycle 25B Hs.153752 H14343 cdc25B=M-phase inducer
116
phosphatase 2
FOXM1 : forkhead box Ml Hs.239 AA129552 MPP2=putative M phase
117
phosphoprotein 2
118 FLJ11029: hypothetical protein FLJ11029 Hs.274448 AI124082
119 H2AFX: H2A histone family, member X Hs.147097 H95392
120 FLJ20333: hypothetical protein FLJ20333 Hs.79828 AA147792
121 SLC17A2: solute carrier family 17 (sodium phosphate), member 2 Hs.19710 H60423
Homo sapiens IRE lb mRNA for protein kinase/ribonuclease IREl beta, complete cds
122
Hs.l 14905 AA102368:
123 ESTs: Hs.163921 AA573689
MCM5: minichromosome maintenance deficient (S. cerevisiae) 5 (cell division cycle
124
46) Hs.77171 AA283961
125 CDKN1B: cyclin-dependent kinase inhibitor IB (p27, Kipl) Hs.238990 AA630082
126 AA779865:
127 PTTG1 : pituitary tumor-transforming 1 Hs.252587 AI362866
128 RAD21 : RAD21 (S. pombe) homolog Hs.81848 AA683102
129 Homo sapiens cDNA FLJ10325 fis, clone NT2RM2000569 Hs.245342 AA430511 :
130 NEK2: NIMA (never in mitosis gene a)-related kinase 2 Hs.153704 AA682321
131 FLJ20101 : LIS 1 -interacting protein NUDE1, rat homolog Hs.263925 N79612
132 FZR1 : Fzrl protein Hs.268384 AA621026
133 ESTs: Hs.120605 AI220472
134 KIAA0855: golgin-67 Hs.l 82982 AA098902 SRD5A1 : steroid-5-alpha-reductase, alpha polypeptide 1 (3-oxo-5 alpha-steroid delta
135
4-dehydrogenase alpha 1) Hs.552 H16833
136 RAD51 : RAD51 (S. cerevisiae) homolog (E coli RecA homolog) Hs.23044 N70010
137 K SL2: kinesin-like 2 Hs.20830 Rl 1542
138 KIAA0097: KIAA0097 gene product Hs.76989 AA598942
139 TUBB: tubulin, beta polypeptide Hs.179661 AA427899
140 HEC: highly expressed in cancer, rich in leucine heptad repeats Hs.58169 W72679
141 TROAP: trophinin associated protein (tastin) Hs.171955 H94949
142 ESTs: Hs.49047 N64737
143 ESTs: Hs.15091 AA678348
144 ESTs: Hs.133431 AI061169
145 KIAA0042: KIAA0042 gene product Hs.3104 AA477501
146 FZR1 : Fzrl protein Hs.268384 AA862886
147 FEN1 : flap structure-specific endonuclease 1 Hs.4756 AA620553
CKS1 : CDC2-Associated Protein CKS1 Hs.77550 AA459292 ckshsl=homolog of
148
Cksl=p34Cdc28/Cdc2-associated protein
149 ESTs: Hs.193379 N57936
150 CASP8AP2: CASP8 associated protein 2 Hs.122843 H50582
BIRC2: baculoviral IAP repeat-containing 2 Hs.289107 R19628 c-IAPl=MIHB=IAP
151
homolog B
152 CKAP2: cytoskeleton associated protein 2 Hs.24641 AA504130
153 HLA-DRA: major histocompatibility complex, class II, DR alpha Hs.76807 R47979
154 HBP: Hairpin binding protein, histone Hs.75257 AA629558
155 FLJ10483: hypothetical protein FLJ10483 Hs.6877 H12254
CASP3: caspase 3, apoptosis-related cysteine protease Hs.74552 R14760 CASPASE-
156
3=CPP32 isoform alpha=yama=cysteine protease
**ESTs,: Weakly similar to protein that is immuno-reactive with anti-PTH polyclonal
157
antibodies [H.sapiens] Hs.301486 AA088258
HMG2: high-mobility group (nonhistone chromosomal) protein 2 Hs.80684
158
AA019203
159 PRO2000: PRO2000 protein Hs.46677 H58234
160 FLJ20333: hypothetical protein FLJ20333 Hs.79828 T48760
161 T56726:
TIMP1 : tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity,
162
collagenase inhibitor) Hs.5831 H80214
163 ESTs: Hs.102004 R94281
164 FLJ10858: hypothetical protein FLJ10858 Hs.134403 AA677552
165 Homo sapiens cDNA FLJ11883 fis, clone HEMBA1007178 Hs.157148 N62451 :
RFC4: replication factor C (activator 1) 4 (37kD) Hs.35120 N93924 replication factor
166
C
167 PRO2000: PRO2000 protein Hs.46677 N47113 168 ECT2: epithelial cell transforming sequence 2 oncogene Hs.132808 AI031571
169 ESTs: Hs.165909 AA629538
170 PCF11 : PCF1 lp homolog Hs.123654 AA053411
BIRC3: baculoviral IAP repeat-containing 3 Hs.127799 H48533 c-IAP2=MIHC=IAP
171
homolog C=TNFR2-TRAF signalling complex prot
172 EST,: Weakly similar to dJ45P21.2 [H.sapiens] Hs.326451 AA931528
173 KIAA0952: KIAA0952 protein Hs.7935 AA454989
174 KIF5B: kinesin family member 5B Hs.149436 AA608707
175 DKFZP566C134: DKFZP566C134 protein Hs.20237 N39306
176 ANLN: anillin (Drosophila Scraps homolog), actin binding protein Hs.62180 R17092
177 ORCIL: origin recognition complex, subunit 1 (yeast homolog)-like Hs.17908 H51719
178 ESTs: Hs. l4139 T77757
179 IFITl : interferon-induced protein with tetratricopeptide repeats 1 Hs.20315 AA074989
180 MGC5338: hypothetical protein MGC5338 Hs.99598 AA463627
181 COPEB: core promoter element binding protein Hs.285313 AA013481
182 UK114: translational inhibitor protein pl4.5 Hs.18426 N72715
183 ESTs: Hs.265592 H67282
HMG4: high-mobility group (nonhistone chromosomal) protein 4 Hs.19114
184
AA670197
185 MDS025: hypothetical protein MDS025 Hs.154938 AI225067
186 DKFZP564A122: DKFZP564A122 protein Hs.187991 N53236
TSC22: transforming growth factor beta-stimulated protein TSC-22 Hs. l 14360
187
AA664389
188 AAAS: aladin Hs.125262 AA916726
189 PLAG1 : **pleiomorphic adenoma gene 1 Hs.14968 AA418251
FLJ23293: ** hypothetical protein FLJ23293 similar to ARL-6 interacting protein-2
190
Hs.31236 R91583
H 11 : protein kinase H 11 ; small stress protein- like protein HSP22 Hs.111676
191
AA010110
192 POLD3: polymerase (DNA directed), delta 3 Hs.82502 AA504204
SERPINB3: serine (or cysteine) proteinase inhibitor, clade B (ovalbumin), member 3
193
Hs.227948 AA292860
194 DNAJB1 : DnaJ (Hsp40) homolog, subfamily B, member 1 Hs.82646 AA435948
195 ESTs: Hs.99480 AA458886
BUB3: BUB3 (budding uninhibited by benzimidazoles 3, yeast) homolog Hs.40323
196
AA405690
197 TUBB2: tubulin, beta, 2 Hs.251653 AI000256
198 Homo sapiens SNC73 protein (SNC73) mRNA, complete cds Hs.293441 H28469:
BUB3: BUB3 (budding uninhibited by benzimidazoles 3, yeast) homolog Hs.40323
199
H38804
200 FLJ20699: hypothetical protein FLJ20699 Hs.15125 AA459420 201 KIAA0013: KIAA0013 gene product Hs.172652 N63575
202 ESTs: Hs.20575 N20305
CDC25C: cell division cycle 25C Hs.656 W95000 cdc25C=M-phase inducer
203
phosphatase 3
204 FLJ11186: hypothetical protein FLJ11186 Hs.89278 AA394225
205 TOPK: PDZ-binding kinase; T-cell originated protein kinase Hs.104741 AA448898
206 KIAA0165: extra spindle poles, S. cerevisiae, homolog of Hs.153479 AA948058
207 LOC51659: HSPC037 protein Hs.108196 AA961752
208 ESTs: Hs.10338 AA436456
209 SUCLG2: succinate-CoA ligase, GDP-forming, beta subunit Hs.247309 AA465233
210 ZNF265: zinc finger protein 265 Hs.194718 AA452256
211 SKP2: S-phase kinase-associated protein 2 (p45) Hs.23348 R22188
212 NS1-BP: NSl-binding protein Hs.197298 AA486796
213 C21ORF50: chromosome 21 open reading frame 50 Hs.4055 AA416628
214 BIRC2: baculoviral IAP repeat-containing 2 Hs.289107 AA702174
BIRC3: baculoviral IAP repeat-containing 3 Hs.127799 AA002125 c-
215
IAP2=MIHC=IAP homolog C=TNFR2-TRAF signalling complex prot
216 INDO: indoleamine-pyrrole 2,3 dioxygenase Hs.840 AA478279
217 DEEPEST: mitotic spindle coiled-coil related protein Hs.16244 T97349
218 ESTs: Hs.105826 AA534321
219 C20ORF1 : chromosome 20 open reading frame 1 Hs.9329 AI654707
220 Homo sapiens cDNA: FLJ21869 fis, clone HEP02442 Hs.28465 R63929:
221 RGS3: regulator of G-protein signalling 3 Hs.82294 AI369623
222 Homo sapiens DC29 mRNA, complete cds Hs.85573 AA186460:
MCM6: minichromosome maintenance deficient (mis5, S. pombe) 6 Hs.155462
223
AA663995
NPAT: nuclear protein, ataxia-telangiectasia locus Hs.89385 AA284172
224
NPAT=E14=gene in ATM locus
225 KNSL6: kinesin-like 6 (mitotic centromere-associated kinesin) Hs.69360 AA400450
226 HN1 : hematological and neurological expressed 1 Hs.109706 AA459865
227 TUB A3 : Tubulin, alpha, brain-specific Hs.272897 AA865469
228 ESTs: Hs.221197 N55457
229 KIAA0175: KIAA0175 gene product Hs.184339 AA903137
230 CLASPIN: homolog of Xenopus Claspin Hs.175613 AA857804
CTNNA1 : **catenin (cadherin-associated protein), alpha 1 (102kD) Hs.178452
231
AA026631
232 ESTs: Hs.221962 AA229644
SMC4L1 : SMC4 (structural maintenance of chromosomes 4, yeast)-like 1 Hs.50758
233
AA452095
234 ICBP90: transcription factor Hs.108106 AA026356 235 EXOl : exonuclease 1 Hs.47504 AA703000
236 Homo sapiens TRAF4 associated factor 1 mRNA, partial cds Hs.181466 T84975:
237 ESTs: Hs.186814 AA700879
238 FLJ11269: hypothetical protein FLJ11269 Hs.25245 R37817
SFPQ: splicing factor proline/glutamine rich (polypyrimidine tract-binding protein-
239
associated) Hs.180610 AA425258
240 ZF: HCF-binding transcription factor Zhangfei Hs.29417 AA 164474
241 TUBA2: tubulin, alpha 2 Hs.98102 AA626698
Homo sapiens mRNA; cDNA DKFZp434M0435 (from clone DKFZp434M0435)
242
Hs.25700 N94435:
243 FLJ20530: ** hypothetical protein FLJ20530 Hs.279521 AA425442
244 BTEB1 : basic transcription element binding protein 1 Hs.150557 N80235
245 LOC51053: geminin Hs.234896 H51100
D21S2056E: DNA segment on chromosome 21 (unique) 2056 expressed sequence
246
Hs.l 10757 AI362799
247 HDAC3: histone deacetylase 3 Hs.279789 H88540
248 USP1 : ubiquitin specific protease 1 Hs.35086 AA099033
249 C21ORF50: chromosome 21 open reading frame 50 Hs.4055 AA135912
250 FLJ13046: ** hypothetical protein FLJ13046 similar to exportin 4 Hs.l 17102 T95333
251 ESTs: Hs.181059 AA912032
252 FLJ22009: hypothetical protein FLJ22009 Hs.123253 AA401234
253 ESTs: Hs.62711 AA056377
RAD51C: RAD51 (S. cerevisiae) homolog C Hs.l 1393 R37145
254
RAD5 lC=Recombination/repair Rad51 -related protein
255 ESTs: Hs.268919 H53508
256 Homo sapiens cDNA FLJ11381 fis, clone HEMBA1000501 Hs.127797 AA885096:
257 SAP30: sin3-associated polypeptide, 30kD Hs.20985 AA126982
258 H4FG: H4 histone family, member G Hs.46423 AA868008
259 TUBA1 : tubulin, alpha 1 (testis specific) Hs.75318 AA180742 tubulin-alpha-4
260 DHFR: dihydrofolate reductase Hs.83765 R00884 DHFR=Dihydrofolate reductase
261 DHFR: dihydrofolate reductase Hs.83765 N52980
262 MGC5528: hypothetical protein MGC5528 Hs.315167 AA934904
263 NNMT: nicotinamide N-methyltransferase Hs.76669 T72089
264 TUBB: tubulin, beta polypeptide Hs.179661 AI672565
HSPAIL: heat shock 70kD protein-like 1 Hs.80288 H17513 HSP70-HOM=Heat shock
265
70 KD protein 1
266 TUBA1 : **tubulin, alpha 1 (testis specific) Hs.75318 R36063
267 PRO1073: **PRO1073 protein Hs.6975 AA176999 CIP4=Cdc42-interacting protein 4
268 POLD3: polymerase (DNA directed), delta 3 Hs.82502 AI017254 ESTs,: Moderately similar to T50635 hypothetical protein DKFZp762L0311.1
269
[H.sapiens] Hs.47378 N38809
270 DKFZP564A122: DKFZP564A122 protein Hs.187991 N57723
271 LRRFIP1 : **leucine rich repeat (in FLU) interacting protein 1 Hs.326159 T84633
272 ESTs: Hs.55468 AA165312
273 ESTs: Hs.31444 H16772
274 AFAP: actin filament associated protein Hs.80306 R69355
CXCR4: chemokine (C-X-C motif), receptor 4 (fusin) Hs.89414 T62491 CXC
275
chemokine receptor 4= fusin=neuropeptide Y receptor=L3
MSH2: **mutS (E. coli) homolog 2 (colon cancer, nonpolyposis type 1) Hs.78934
276
AA679697
111 ESTs: Hs.48474 N62074
278 AA677337:
ESTs,: Moderately similar to TBB2 HUMAN TUBULIN BETA-2 CHAIN
279
[H.sapiens] Hs.23189 AA629908
280 HP1-BP74: HP1-BP74 Hs.142442 H79795
281 FLJ20101 : LIS 1 -interacting protein NUDEl, rat homolog Hs.263925 AA459394
Homo sapiens mRNA; cDNA DKFZp434D1428 (from clone DKFZp434D1428);
282
complete cds Hs.321775 AA431268:
283 ESTs: Hs.265592 AA992658
284 ESTs:
DDXl 1 : DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 11 (S.cerevisiae CHL1-
285
like helicase) Hs.27424 AA402879
286 CDC27: cell division cycle 27 Hs.172405 T81764
287 ARGBP2: Arg/Abl-interacting protein ArgBP2 Hs.278626 N89738
288 DKFZP564A122: DKFZP564A122 protein Hs.187991 AA025807
289 OPN3: opsin 3 (encephalopsin) Hs.279926 AA150060
290 DKFZP566C134: DKFZP566C134 protein Hs.20237 AA456319
291 KIAA0855: golgin-67 Hs.182982 H15101
292 PIN: dynein, cytoplasmic, light polypeptide Hs.5120 AA644679
ESTs,: Weakly similar to LIP1 HUMAN PANCREATIC LIPASE RELATED
293
PROTEIN 1 PRECURSO [H.sapiens] Hs.68864 AA088857
294 HDAC3: histone deacetylase 3 Hs.279789 AA973283
295 DONSON: downstream neighbor of SON Hs.17834 AA417895
296 LOC51053: geminin Hs.234896 AA447662
297 FLJ10545: hypothetical protein FLJ10545 Hs.88663 AA460110
MAD2L1 : MAD2 (mitotic arrest deficient, yeast, homolog)-like 1 Hs.79078
298
AA481076 mitotic feedback control protein Madp2 homolog
299 TASR2: TLS-associated serine-arginine protein 2 Hs.3530 HI 1042
300 MCM6: minichromosome maintenance deficient (mis5, S. pombe) 6 Hs.155462 N57722
301 CIT: citron (rho-interacting, serine/threonine kinase 21) Hs.l 5767 W69425
302 **ESTs: Hs.205066 AA284803
ICAM1 : intercellular adhesion molecule 1 (CD54), human rhinovirus receptor
303
Hs.168383 R77293 CD54=ICAM-1
304 KIAA0855: golgin-67 Hs.l 82982 AA456818
305 ESTs,: Weakly similar to putative pi 50 [H.sapiens] Hs.300070 R10422
306 DEEPEST: mitotic spindle coiled-coil related protein Hs.16244 AI652290
MCM2: minichromosome maintenance deficient (S. cerevisiae) 2 (mitotin) Hs.57101
307
AA454572
308 Homo sapiens cDNA: FLJ22272 fis, clone HRC03192 Hs.50740 AA495943:
309 WISP1 : **WNT1 inducible signaling pathway protein 1 Hs.194680 T54850
310 KIAA0855: golgin-67 Hs.l 82982 AA280248
311 TEM8: tumor endothelial marker 8 Hs.8966 H58644
312 BITE: plO-binding protein Hs.42315 H96392
313 RAN: RAN, member RAS oncogene family Hs.10842 AA456636
314 EZH2: enhancer of zeste (Drosophila) homolog 2 Hs.77256 AA428252
315 MCM4: minichromosome maintenance deficient (S. cerevisiae) 4 Hs.154443 W74071
DKFZp434J0310: hypothetical protein Hs.278408 AA279657 Unknown UG Hs.23595
316
ESTs sc_id6950
317 PPP1R10: protein phosphatase 1, regulatory subunit 10 Hs.106019 AA071526
318 HI 1 : protein kinase HI 1; small stress protein-like protein HSP22 Hs. l 11676 H57493
319 ESTs,: Weakly similar to KIAA1074 protein [H.sapiens] Hs.200483 AA463220
ESTs,: Weakly similar to ALU8 HUMAN ALU SUBFAMILY SX SEQUENCE
320
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.226414 N72576
321 AA775033:
322 LOC51004: CGI-10 protein Hs.12239 AA677920
323 ESTs: Hs.150028 AI292036
MCM6: minichromosome maintenance deficient (mis5, S. pombe) 6 Hs.155462
324
AA976533
ESTs,: Moderately similar to T50635 hypothetical protein DKFZp762L0311.1
325
[H.sapiens] Hs.47378 AA406348
326 UCP4: uncoupling protein 4 Hs.40510 H60279
327 MSH5: mutS (E. coli) homolog 5 Hs. l 12193 AA621155
328 ROCKl : Rho-associated, coiled-coil containing protein kinase 1 Hs.17820 AA872143
329 KIAA0855: golgin-67 Hs.l 82982 AA694481
330 AA705332:
331 CDC27: cell division cycle 27 Hs.172405 N47994
332 DONSON: downstream neighbor of SON Hs.17834 AI732249
333 SH3GL2: SH3-domain GRB2-like 2 Hs.75149 R12817 334 PRC1 : protein regulator of cytokinesis 1 Hs.5101 AA449336
ESTs,: Weakly similar to unnamed protein product [H. sapiens] Hs.99807 AA417744
335
Unknown UG Hs.l 19424 ESTs sc_id2235
336 Human: clone 23719 mRNA sequence Hs.80305 AA425722
Homo sapiens mRNA; cDNA DKFZp56402364 (from clone DKFZp56402364)
337
Hs.28893 W90240:
ESTs,: Weakly similar to LIP1 HUMAN PANCREATIC LIPASE RELATED
338
PROTEIN 1 PRECURSO [H.sapiens] Hs.68864 AA132858
339 TUB A3 : Tubulin, alpha, brain-specific Hs.272897 AA864642
340 AI283530:
341 ESTs: Hs.302878 R92512
342 PPP1R10: protein phosphatase 1, regulatory subunit 10 Hs.106019 T75485
343 SFRS5: splicing factor, arginine/serine-rich 5 Hs.l 66975 R73672
344 SFRS3: splicing factor, arginine/serine-rich 3 Hs. l 67460 AA598400
PRIM1 : primase, polypeptide 1 (49kD) Hs.82741 AA025937 DNA primase (subunit
345
p48)
346 FLJ20333: hypothetical protein FLJ20333 Hs.79828 H66982
347 HSPA8: heat shock 70kD protein 8 Hs.180414 AA620511
348 C4A: complement component 4A Hs.l 70250 AA664406
349 DKC1 : dyskeratosis congenita 1, dyskerin Hs.4747 AA052960
350 HP1-BP74: HP1-BP74 Hs.142442 T84669
ETV4: ets variant gene 4 (EIA enhancer-binding protein, EIAF) Hs.77711 AA010400
351
E1A-F=E1A enhancer binding protein=ETS translocation variant
Homo sapiens cDNA: FLJ23037 fis, clone LNG02036, highly similar to HSU68019
352 Homo sapiens mad protein homolog (hMAD-3) mRNA Hs.288261 W42414
Smad3=hMAD-3=Homologue of Mothers Against Decapentaplegic (M:
353 KIAA0952: KIAA0952 protein Hs.7935 AA679150
354 STK9: serine/threonine kinase 9 Hs.50905 N80713
355 NXF1 : **nuclear RNA export factor 1 Hs.323502 R01238
356 FLJ12892: hypothetical protein FL J 12892 Hs.17731 AA449357
357 UNG: uracil-DNA glycosylase Hs.78853 H15111
358 STK17B: ** serine/threonine kinase 17b (apoptosis-inducing) Hs.120996 AA419485
YWHAH: tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein,
359
eta polypeptide Hs.75544 N69107
360 FLJ13154: hypothetical protein FLJ13154 Hs.25303 AA923560
361 LOC51116: CGI-91 protein Hs.20776 AA459419
362 SSXT: synovial sarcoma, translocated to X chromosome Hs.153221 N59206
363 KIAA0978: KIAA0978 protein Hs.3686 AA485878
364 EST: Hs.147907 AI223432
365 FLJ23468: hypothetical protein FLJ23468 Hs.38178 AA431741 366 FLJ10339: ** hypothetical protein FLJ10339 Hs.203963 N95450
367 BMP2: bone morphogenetic protein 2 Hs.73853 AA011061
368 PIR51 : RAD51 -interacting protein Hs.24596 AI214426
369 FLJ20364: hypothetical protein FLJ20364 Hs.32471 AA676296
370 EIF4A2: **eukaryotic translation initiation factor 4A, isoform 2 Hs.173912 H54751
ESTs,: Weakly similar to MCAT HUMAN MITOCHONDRIAL
371 CARNITINE/ ACYLCARNITINE CARRIER PROTEIN [H.sapiens] Hs.27769
AA469975
372 FLJ11323: hypothetical protein FLJ11323 Hs.25625 AA775600
373 DKFZP564D0764: DKFZP564D0764 protein Hs.26799 AA460732
374 CTL2: CTL2 gene Hs.105509 AA454710
375 ESTs: Hs.293419 AA775845
IFITI : interferon-induced protein with tetratricopeptide repeats 1 Hs.20315 AA489640
376
Interferon-induced 56-KDa protein
377 RBBP8: retinoblastoma-binding protein 8 Hs.29287 H23021
378 **Homo sapiens clone 25061 mRNA sequence Hs.183475 R38944:
Human: DNA sequence from clone RP3-383J4 on chromosome lq24.1-24.3 Contains
379 part of a gene encoding a kelch motif containing protein, part of a novel gene encoding a protein similar to Aspartyl-TRNA sy Hs.l 17305 N29457
380 FLJ12888: hypothetical protein FLJ12888 Hs.284137 N68390
ESTs,: Weakly similar to IF38 HUMAN EUKARYOTIC TRANSLATION
381
INITIATION FACTOR 3 SUBUNIT 8 [H.sapiens] Hs.222088 All 39629
382 ESTs: Hs.241101 AA133590
383 H4FI: H4 histone family, member I Hs.143080 AI218900
384 SP38: zona pellucida binding protein Hs.99875 AA400474
GABPB1 : GA-binding protein transcription factor, beta subunit 1 (53kD) Hs.78915
385
H91651
386 LCHN: LCHN protein Hs.12461 AA029330
387 DKFZP564D0462: hypothetical protein DKFZp564D0462 Hs.44197 N32904
LENG8: leukocyte receptor cluster (LRC) encoded novel gene 8 Hs.306121
388
AA464698
HIFIA: hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix transcription
389
factor) Hs. l 97540 AA598526
390 ESTs: Hs.93714 R09201
391 FLJ23468: hypothetical protein FLJ23468 Hs.38178 AA454949
392 DKFZP566C134: DKFZP566C134 protein Hs.20237 AA448164
PPP3CA: protein phosphatase 3 (formerly 2B), catalytic subunit, alpha isoform
393
(calcineurin A alpha) Hs.272458 W60310
394 HMGE: GrpE-like protein cochaperone Hs.151903 H55907 CDK7: cyclin-dependent kinase 7 (homo log of Xenopus MO 15 cdk-activating kinase)
395
Hs.184298 R22624 CAK=cdk7=NRTALRE=sdk=CDK activating kinase
ABCC5: **ATP-binding cassette, sub-family C (CFTR/MRP), member 5 Hs.108660
396
AA186613
397 AA477707:
398 **ESTs: Hs.15607 R92899
399 LOC57209: Kruppel-type zinc finger protein Hs.25275 N50827
400 FLJ20101 : LIS 1 -interacting protein NUDE1, rat homolog Hs.263925 R87716
401 K SL4: kinesin-like 4 Hs.l 19324 AA430503
E2F5: E2F transcription factor 5, pl30-binding Hs.2331 AA455521 E2F-5=pRB-
402
binding transcription factor
403 TMPO: thymopoietin Hs.l 1355 T63980
404 POLQ: polymerase (DNA directed), theta Hs.241517 AI057325
405 TGIF: TG-interacting factor (TALE family homeobox) Hs.90077 H51705
406 TRIP13 : thyroid hormone receptor interactor 13 Hs.6566 AA630784
407 GAS 6: growth arrest-specific 6 Hs.78501 AA461110
408 HN1 : hematological and neurological expressed 1 Hs.109706 AA035429
409 BARD1 : BRCA1 associated RING domain 1 Hs.54089 AA558464
410 DHFR: dihydrofolate reductase Hs.83765 AA424790
411 AA490946:
412 ESTs: Hs.130435 AA167114
413 HSPA8: heat shock 70kD protein 8 Hs.180414 AA629567
414 RRM2: ribonucleotide reductase M2 polypeptide Hs.75319 AA826373
415 FLJ20036: hypothetical protein FLJ20036 Hs.32922 H59114
COPEB: core promoter element binding protein Hs.285313 AA055584
416
CPBP=CBAl=DNA-binding protein
417 FLJ10604: hypothetical protein FLJ10604 Hs.26516 N72697
ESTs,: Weakly similar to cDNA EST yk415cl2.5 comes from this gene [C.elegans]
418
Hs.108824 H97880
UBE2D3: **ubiquitin-conjugating enzyme E2D 3 (homologous to yeast UBC4/5)
419
Hs.l 18797 AA017199
420 FLJ10890: ** hypothetical protein FLJ10890 Hs.17283 AA004210
421 ESTs: Hs.214410 AA579336
422 OLR1 : oxidised low density lipoprotein (lectin-like) receptor 1 Hs.77729 AA682386
423 FLJ13231 : hypothetical protein FLJ13231 Hs.156148 W92787
424 EST: Hs.323101 W40398
425 ESTs,: Weakly similar to R06F6.5b [C.elegans] Hs.180591 N59330
426 Homo sapiens cDNA: FLJ23285 fis, clone HEP09071 Hs.90424 N26163:
Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 42408 Hs.284123
427
AA211446: NFKB1 : nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (pi 05)
428
Hs.83428 AA451716 NFkBl = NF-kappaB pl05=p50
429 LOC58486: transposon-derived Busterl transposase-like protein Hs.25726 AA630256
430 Homo sapiens cDNA FLJ10976 fis, clone PLACE1001399 Hs.296323 AA424756:
431 KIAA0182: KIAA0182 protein Hs.75909 AI023801
432 RAN GAP 1 : Ran GTPase activating protein 1 Hs.183800 AA991855
PKMYT1 : membrane-associated tyrosine- and threonine-specific cdc2-inhibitory
433
kinase Hs.77783 AA478066 Mytl kinase
434 HSPA8: heat shock 70kD protein 8 Hs.180414 H64096
435 LUC7A: cisplatin resistance-associated overexpressed protein Hs.3688 AA411969
436 RRM1 : ribonucleotide reductase Ml polypeptide Hs.2934 AA633549
437 SET07: PR/SET domain containing protein 7 Hs.l 11988 AA421470
**ESTs,: Weakly similar to ALU1 HUMAN ALU SUBFAMILY J SEQUENCE
438
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.l 93452 W96179
439 Homo sapiens clone 25058 mRNA sequence Hs.179397 R38894:
440 ESTs,: Weakly similar to KIAA0973 protein [H.sapiens] Hs.14014 AA780791
441 EST: Hs.l05298 AA489813
442 CTCF: CCCTC-binding factor (zinc finger protein) Hs.57419 H89996
443 HRB: HIV-1 Rev binding protein Hs.171545 AA485958
444 **ESTs: Hs.294083 AA447679
445 KIAA0878: KIAA0878 protein Hs.l 88006 AA599094
ESTs,: Weakly similar to ALUB HUMAN ! ! ! ! ALU CLASS B WARNING ENTRY
446
! ! ! [H.sapiens] Hs. l 80552 AA481283
OGT: O-linked N-acetylglucosamine (GlcNAc) transferase (UDP-N-
447 acetylglucosamine:polypeptide-N-acetylglucosaminyl transferase) Hs.100293
AA425229
448 Homo sapiens mRNA for KIAA1700 protein, partial cds Hs.20281 N40952:
Human: DNA sequence from clone RP1-187J11 on chromosome 6ql 1.1-22.33.
449 Contains the gene for a novel protein similar to S. pombe and S. cerevisiae predicted proteins, the gene for a novel protein simila Hs.72325 AAl 59962
450 KIAA1265: KIAA1265 protein Hs.24936 AA479302
451 H1F0: HI histone family, member 0 Hs.226117 H57830
452 ARGBP2: Arg/Abl-interacting protein ArgBP2 Hs.278626 H02525
453 ODF2: outer dense fibre of sperm tails 2 Hs.129055 AA149882
454 CD97: CD97 antigen Hs.3107 AI651871
455 BMI1 : **murine leukemia viral (bmi-1) oncogene homolog Hs.431 AA193573
456 POLG: polymerase (DNA directed), gamma Hs.80961 AA188629
457 XPR1 : xenotropic and polytropic retrovirus receptor Hs.227656 AA453474
458 ESTs: Hs.128096 AA971179
459 DNAJB1 : DnaJ (Hsp40) homolog, subfamily B, member 1 Hs.82646 AA481022 460 ARL4: ADP-ribosylation factor-like 4 Hs.201672 AI142552
461 SFRS5: splicing factor, arginine/serine-rich 5 Hs.166975 AA598965
462 ESTs: Hs.25933 R11605
463 RIG-I: RNA helicase Hs.145612 AA126958
464 FLJ10339: hypothetical protein FLJ10339 Hs.203963 AA628231
DRl : down-regulator of transcription 1, TBP-binding (negative cofactor 2) Hs.16697
465
AA043503
Homo sapiens, Similar to hypothetical protein FLJ20093, clone MGC: 1076, mRNA,
466
complete cds Hs.298998 AA703249:
467 HSPC163: HSPC163 protein Hs.108854 H98963
468 DKFZP564A122: DKFZP564A122 protein Hs.187991 R27345
FLJ10128: uveal autoantigen with coiled coil domains and ankyrin repeats Hs.49753
469
T47624
470 DSCR1 : Down syndrome critical region gene 1 Hs.184222 AA629707
471 FLJ10342: hypothetical protein FLJ10342 Hs.101514 AA490935
Homo sapiens mRNA; cDNA DKFZp586N1323 (from clone DKFZp586N1323)
472
Hs.24064 R26176:
473 ESTs: Hs.4983 H59921
ESTs,: Weakly similar to ALUB HUMAN ! ! ! ! ALU CLASS B WARNING ENTRY
474
! ! ! [H.sapiens] Hs.117949 H91167
CDC45L: CDC45 (cell division cycle 45, S.cerevisiae, homolog)-like Hs. l 14311
475
AA700904
STAT5B: signal transducer and activator of transcription 5B Hs.244613 AA280647
476
STAT5A/5B
477 Homo sapiens cDNA FLJ14028 fis, clone HEMBA1003838 Hs.281434 AA454682:
478 KIAA1524: KIAA1524 protein Hs.151343 AI248987
479 CTSD: cathepsin D (lysosomal aspartyl protease) Hs.79572 AA485373
Homo sapiens, Similar to hypothetical protein FLJ20093, clone MGC: 1076, mRNA,
480
complete cds Hs.298998 AA682274:
481 GTPBP2: GTP binding protein 2 Hs.13011 T67069
482 LOC51003: CGI-125 protein Hs.27289 AA485945
483 VCL: vinculin Hs.75350 AA486727
484 KIF5B: kinesin family member 5B Hs.149436 AA046613
485 CDC25A: cell division cycle 25A Hs.1634 AA071514
486 LOC51141 : insulin induced protein 2 Hs.7089 AA045308
**ESTs,: Moderately similar to CALD HUMAN CALDESMON [H.sapiens]
487
Hs.l 17774 H48508
488 TBX3-iso: TBX3-iso protein Hs.267182 T48941
489 KIAA0176: KIAA0176 protein Hs.4935 R44371 PRKARl A: protein kinase, cAMP-dependent, regulatory, type I, alpha (tissue specific
490 extinguisher 1) Hs.183037 N25969 PKA-R1 alpha=cAMP-dependent protein kinase type I-alpha-cata
491 ESTs: Hs.268991 H77818
ESTs,: Weakly similar to A53028 isopentenyl-diphosphate Delta-isomerase
492
[H.sapiens] Hs.9270 R17362
493 ESTs,: Weakly similar to B34087 hypothetical protein [H.sapiens] Hs.120946 H50656
494 TRN2: karyopherin beta 2b, transportin Hs.278378 R08897
495 LMNA: lamin A/C Hs.77886 AA489582
496 NFE2L2: nuclear factor (erythroid-derived 2)-like 2 Hs.155396 AA629687
497 DKFZp762L0311 : hypothetical protein DKFZp762L0311 Hs.16520 AA486418
498 ESTs,: Weakly similar to S71752 giant protein p619 [H.sapiens] Hs.14870 T96829
Homo sapiens mRNA; cDNA DKFZp434A1315 (from clone DKFZp434A1315);
499
complete cds Hs.298312 AA991355:
500 E2IG4: hypothetical protein, estradiol-induced Hs.8361 R13844
501 RAN GAP 1 : Ran GTPase activating protein 1 Hs.183800 AA485734
502 H1F0: HI histone family, member 0 Hs.226117 W69399
503 KIAA0239: KIAA0239 protein Hs.9729 AA454740
ESTs,: Weakly similar to ALU7 HUMAN ALU SUBFAMILY SQ SEQUENCE
504
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.68647 R96804
505 PRO0650: PRO0650 protein Hs.177258 N54333
506 DNAJB9: DnaJ (Hsp40) homolog, subfamily B, member 9 Hs.6790 AA045792
507 Homo sapiens cDNA: FLJ21971 fis, clone HEP05790 Hs.71331 AA774678:
508 LOC56996: ** cation-chloride cotransporter-interacting protein Hs. l 19178 AA037466
509 AP3D1 : adaptor-related protein complex 3, delta 1 subunit Hs.75056 AA630776
SGK: serum/glucocorticoid regulated kinase Hs.159640 AA486082 sgk=putative
510
serine/threonine protein kinase transcriptional
511 HSPC148: hypothetical protein Hs.42743 R23666
512 MRPL19: mitochondrial ribosomal protein L19 Hs.75574 AA521243 KIAA0104
513 AA455102:
514 ESTs: Hs.150325 AI278813
515 **ESTs: Hs.40527 AA029844
516 HSPC145: HSPC145 protein Hs. l 8349 AI271431
517 KIAA0170: KIAA0170 gene product Hs.277585 H68789
518 FLJ11127: hypothetical protein Hs.91165 T98200
519 KIAA0182: KIAA0182 protein Hs.75909 H05099
520 FLJ23151 : hypothetical protein FLJ23151 Hs.137260 AA284259
521 AMD1 : S-adenosylmethionine decarboxylase 1 Hs.262476 AA425692
522 FLJ10342: ** hypothetical protein FLJ10342 Hs.101514 AA934516 SPS: SELENOPHOSPHATE SYNTHETASE ; Human selenium donor protein
523
Hs.124027 AA486372
524 KIAA1586: KIAA1586 protein Hs.180663 AA779733
525 ICBP90: transcription factor Hs.108106 AA908902
526 Homo sapiens cDNA: FLJ21971 fis, clone HEP05790 Hs.71331 AI002036:
ABCC2: ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Hs.193852
527
R91502
ARHGDIB: Rho GDP dissociation inhibitor (GDI) beta Hs.83656 AA487426
528
LyGDI=Rho GDP-dissociation inhibitor 2=RHO GDI 2
529 RAD53: protein kinase Chk2 Hs.146329 AI653182
530 R96880:
531 TNFAIP3: tumor necrosis factor, alpha-induced protein 3 Hs.211600 AA433807
532 ESTs: Hs.26979 H23469
533 AOC2: amine oxidase, copper containing 2 (retina-specific) Hs.143102 N50959
Homo sapiens mRNA; cDNA DKFZp586N1323 (from clone DKFZp586N1323)
534
Hs.24064 R30941 :
535 AA452872:
536 ESTs: Hs.124169 R58970
537 ACYP1 : acylphosphatase 1, erythrocyte (common) type Hs.18573 W78754
538 SIL: TALI (SCL) interrupting locus Hs.323032 AA704809
539 AA016234:
Homo sapiens mRNA; cDNA DKFZp566Pl 124 (from clone DKFZp566Pl 124)
540
Hs.321022 N50895:
541 KIAA1067: KIAA1067 protein Hs.325530 AA099138
SMC4L1 : SMC4 (structural maintenance of chromosomes 4, yeast)-like 1 Hs.50758
542
AA283006
543 ESTs: Hs.29074 R70174
544 SNK: serum-inducible kinase Hs.3838 AA460152
545 FANCG: Fanconi anemia, complementation group G Hs.8047 AA427484
546 Homo sapiens cDNA: FLJ21531 fis, clone COL06036 Hs.102941 N95440:
Homo sapiens mRNA; cDNA DKFZp547B086 (from clone DKFZp547B086)
547
Hs.36606 N48700:
C10RF2: chromosome 1 open reading frame 2 Hs.19554 HI 1464 cotel=ORF in
548
glucocerebrosidase locus
549 HTF9C: Hpall tiny fragments locus 9C Hs.63609 HI 7888
ATF4: activating transcription factor 4 (tax-responsive enhancer element B67)
550
Hs.181243 AA600217
551 ESTs: Hs.101014 AA194941
552 CDC25A: cell division cycle 25 A Hs.1634 AA913262
553 TOPK: PDZ-binding kinase; T-cell originated protein kinase Hs.104741 AI002631 554 ASIP: agouti (mouse)-signaling protein Hs.37006 AI220203
555 DKFZP564F013: * hypothetical protein DKFZp564F013 Hs.128653 R14908
556 ZNF265: zinc finger protein 265 Hs.194718 N66014
557 SLC30A1 : solute carrier family 30 (zinc transporter), member 1 Hs.55610 AA195463
558 ESTs: Hs.28462 R63922
559 ESTs: Hs.114055 R27431
560 IL6: interleukin 6 (interferon, beta 2) Hs.93913 N98591 IL-6
561 H3F3B: H3 histone, family 3B (H3.3B) Hs.180877 AA608514
562 ESTs: Hs.81263 W81524
Homo sapiens cDNA: FLJ23538 fis, clone LNG08010, highly similar to BETA2
563
Human MEN1 region clone epsilon/beta mRNA Hs.240443 AA400234:
564 AMD1 : S-adenosylmethionine decarboxylase 1 Hs.262476 R82299
565 MAP3K2: mitogen-activated protein kinase kinase kinase 2 Hs.28827 AA447971
566 NET1 : neuroepithelial cell transforming gene 1 Hs.25155 R24543
567 CHAF1A: chromatin assembly factor 1, subunit A (pl50) Hs.79018 AA704459
568 MGC5585: hypothetical protein MGC5585 Hs.5152 H50655
569 KIAA1598: KIAA1598 protein Hs.23740 HI 7868
570 PNN: pinin, desmosome associated protein Hs.44499 W86139
571 ESTs: Hs.238797 N70848
ESTs,: Weakly similar to ALUB HUMAN ! ! ! ! ALU CLASS B WARNING ENTRY
572
! ! ! [H.sapiens] Hs.180552 AA600192
573 PDGFA: platelet-derived growth factor alpha polypeptide Hs.37040 AA701502
574 Homo sapiens clone FLC0675 PRO2870 mRNA, complete cds Hs.306117 AA443127:
575 ESTs: Hs.143375 AA001841
576 TUBB: tubulin, beta polypeptide Hs.179661 H37989
MSH2: mutS (E. coli) homolog 2 (colon cancer, nonpolyposis type 1) Hs.78934
577
AA219060 MSH2=DNA mismatch repair mutS homologue
578 TOPBP1 : topoisomerase (DNA) II binding protein Hs.91417 R97785
579 KIAA0869: KIAA0869 protein Hs.21543 R43798
580 H4FH: H4 histone family, member H Hs.93758 AA702781
FLJ23293: hypothetical protein FLJ23293 similar to ARL-6 interacting protein-2
581
Hs.31236 AA629027
**Homo sapiens cDNA: FLJ23538 fis, clone LNG08010, highly similar to BETA2
582
Human MEN1 region clone epsilon/beta mRNA Hs.240443 AA053165:
583 KIAA0978: KIAA0978 protein Hs.3686 N64780
584 KIAA1547: KIAA1547 protein Hs.31305 AA057737
585 DKFZP761C169: hypothetical protein DKFZp761C169 Hs.71252 AA608709
586 WS-3: novel RGD-containing protein Hs.39913 AA449975
587 FRZB: frizzled-related protein Hs.153684 H87275 BRCAl : breast cancer 1, early onset Hs.194143 H90415 BRCAl =Mutated in breast
588
and ovarian cancer
589 ESTs: Hs.4983 H22936
HSPC150: HSPC150 protein similar to ubiquitin-conjugating enzyme Hs.5199
590
AA460431
591 Homo sapiens mRNA for KIAA1712 protein, partial cds Hs.29798 H54592:
FLJ11186: hypothetical protein FLJ11186 Hs.89278 AA504111 Unknown UG
592
Hs.89278 ESTs
593 ESTs,: Weakly similar to unnamed protein product [H. sapiens] Hs. l 18338 R25481
APEXL2: apurinic/apyrimidinic endonuclease(APEX nuclease)-like 2 protein
594
Hs.154149 AI674393
595 CDR2: cerebellar degeneration-related protein (62kD) Hs.75124 AA074613
596 ESTs: Hs.69662 AA459724
PSCD2L: pleckstrin homology, Sec7 and coiled/coil domains 2-like Hs.8517
597
AA464957
598 CRK: v-crk avian sarcoma virus CT10 oncogene homolog Hs.306088 H75530
599 CCNE2: cyclin E2 Hs.30464 AA520999 Unknown UG Hs.30464 cyclin E2
600 LOC51240: hypothetical protein Hs.7870 AA988037
601 FLJ11259: hypothetical protein FLJ11259 Hs. l 84465 AA485877
602 PTP4A1 : protein tyrosine phosphatase type IVA, member 1 Hs.227777 AA482193
603 Homo sapiens cDNA: FLJ22355 fis, clone HRC06344 Hs.288283 AA026375:
604 Human: clone 23719 mRNA sequence Hs.80305 H43437
605 Homo sapiens clone FLC0675 PRO2870 mRNA, complete cds Hs.306117 AA485453:
606 MSE55: serum constituent protein Hs.148101 H73234
607 CFLAR: CASP8 and FADD-like apoptosis regulator Hs.195175 AA453766
608 Homo sapiens cDNA: FLJ22844 fis, clone KAIA5181 Hs.296322 AA975103:
Human: DNA sequence from clone RPl 1-371L19 on chromosome 20 Contains two
609 novel genes, the gene for a novel protein similar to 40S ribosomal protein S10
(RPS10), ESTs, STSs, GSSs and five CpG islands Hs. l 9002 R00846
610 ESTs: Hs.60054 R26390
ESTs,: Weakly similar to ALU7 HUMAN ALU SUBFAMILY SQ SEQUENCE
611
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.325158 AA032084
612 FLJ10980: hypothetical protein FLJ10980 Hs.29716 N45467
IFIT1 : **interferon-induced protein with tetratricopeptide repeats 1 Hs.20315
613
AA157787
614 ESTs: Hs.21734 AA429809
615 DKFZP434C245: DKFZP434C245 protein Hs.59461 AA705518
616 RNPSl : RNA-binding protein SI, serine-rich domain Hs.75104 AA496837
617 FLJ13639: hypothetical protein FLJ13639 Hs.101821 AA131681
618 PCF11 : PCFl lp homolog Hs.123654 W73749
619 EIF4G3: eukaryotic translation initiation factor 4 gamma, 3 Hs.25732 N92469 620 Homo sapiens cDNA: FLJ21971 fis, clone HEP05790 Hs.71331 AA130595:
621 STATl : signal transducer and activator of transcription 1, 91kD Hs.21486 AA079495
622 BIRC3: baculoviral IAP repeat-containing 3 Hs.127799 R07870
623 HP1-BP74: HP1-BP74 Hs.142442 N20589
624 HSPC228: hypothetical protein Hs.267288 AI734268
625 KIAA0675: KIAA0675 gene product Hs.165662 AA454867
626 AMD1 : S-adenosylmethionine decarboxylase 1 Hs.262476 AA504772
627 EST: Hs.l49338 AI249089
628 PWPl : nuclear phosphoprotein similar to S. cerevisiae PWPl Hs.172589 AA485992
629 AI336973:
630 DUSP4: dual specificity phosphatase 4 Hs.2359 AA444049
631 FLJ12788: hypothetical protein FLJ12788 Hs.20242 AA497041
HSPC150: HSPC150 protein similar to ubiquitin-conjugating enzyme Hs.5199
632
AA985450
633 FLJ11729: hypothetical protein FLJ11729 Hs.286212 W15533
634 KLF4: Kruppel-like factor 4 (gut) Hs.7934 H45668
635 FLJ11058: hypothetical protein FLJ11058 Hs.180817 N63911
636 FLJ23468: hypothetical protein FLJ23468 Hs.38178 AA460299
637 ESTs: Hs. l 15315 AI278336
EBI3: Epstein-Barr virus induced gene 3 Hs. l 85705 AA425028 EBI3=cytokine
638
receptor
639 ESTs: Hs.293797 N63988
MGAT2: mannosyl (alpha- l,6-)-glycoprotein beta- 1,2-N-
640
acetylglucosaminyltransferase Hs.l 72195 AA485653
641 H2BFQ: H2B histone family, member Q Hs.2178 AA456298
642 NMB: neuromedin B Hs.83321 AI650675
SSR3: signal sequence receptor, gamma (translocon-associated protein gamma)
643
Hs.28707 AA453486
644 HSPC196: hypothetical protein Hs.239938 R78498
645 EST: Hs.44522 N33610
646 BRF1 : butyrate response factor 1 (EGF-response factor 1) Hs.85155 AA723035
647 MAN1A2: mannosidase, alpha, class 1A, member 2 Hs.239114 H97940
648 KIAA1201 : KIAA1201 protein Hs.251278 AA427719
649 NUCKS: similar to rat nuclear ubiquitous casein kinase 2 Hs.l 18064 AA158345
650 MAGEF1 : MAGEF1 protein Hs.306123 AA425302
651 Human: Chromosome 16 BAC clone CIT987SK-A-362G6 Hs.6349 N75498
652 R40377:
653 AP3M2: adaptor-related protein complex 3, mu 2 subunit Hs.77770 R14443
ESTs,: Weakly similar to 1207289A reverse transcriptase related protein [H. sapiens]
654
Hs.272135 AA705010 655 Homo sapiens mRNA for FLJ00116 protein, partial cds Hs.72363 AA159893:
656 EIF4E: eukaryotic translation initiation factor 4E Hs.79306 AA193254
657 Homo sapiens mRNA for hypothetical protein (TR2/D15 gene) Hs.180545 N47285:
658 ESTs: Hs.99542 AA461474
659 CTNND1 : catenin (cadherin-associated protein), delta 1 Hs.166011 AA024656
660 ESTs: Hs.188554 R75884
661 ZNF217: zinc finger protein 217 Hs.155040 R81830
662 FLJ12892: hypothetical protein FLJ12892 Hs.17731 AI243595
663 ETV5: ets variant gene 5 (ets-related molecule) Hs.43697 AA460265
664 EST: Hs.251574 T54821
665 RPS25: ribosomal protein S25 Hs.l 13029 T98662
666 CNN2: calponin 2 Hs.169718 AA284568
667 ESTs,: Weakly similar to plakophilin 2b [H.sapiens] Hs.12705 AA485365
668 PAPPA: pregnancy-associated plasma protein A Hs.75874 AA609463
669 TFF3: trefoil factor 3 (intestinal) Hs.82961 N74131
670 AI204264:
671 DJ328E19.C1.1 : hypothetical protein Hs.218329 AA486041
672 ME3: malic enzyme 3, NADP(+)-dependent, mitochondrial Hs.2838 AA779401
ESTs,: Weakly similar to IEFS HUMAN TRANSFORMATION-SENSITIVE
673
PROTEIN IEF SSP 3521 [H.sapiens] Hs.43213 AA490554
674 FLJ13181 : hypothetical protein FLJ13181 Hs.301526 AA057266
675 KIAA1547: KIAA1547 protein Hs.31305 AA136692
676 ZNF281 : zinc finger protein 281 Hs.59757 N47468
Homo sapiens cDNA: FLJ23260 fis, clone COL05804, highly similar to HSU90911
677
Human clone 23652 mRNA sequence Hs.13996 AA463961 :
678 ESTs: Hs.25933 AA411392
NCBPl : nuclear cap binding protein subunit 1, 80kD Hs.89563 AA278749 nuclear cap
679
binding protein
680 H2BFL: H2B histone family, member L Hs.239884 H70774
681 DKFZP564A122: DKFZP564A122 protein Hs.187991 H66150
682 NASP: nuclear autoantigenic sperm protein (histone -binding) Hs.243886 AA644128
683 **ESTs,: Weakly similar to KIAA0822 protein [H.sapiens] Hs.98368 AA422008
684 MAP2K6: mitogen-activated protein kinase kinase 6 Hs. l 18825 H07920
685 ESTs: Hs.158357 AA865842
GADD45A: growth arrest and DNA-damage-inducible, alpha Hs.80409 AA147214
686
GADD45 alpha=growth arrest and DNA-damage-inducible protein
687 DHFR: dihydrofolate reductase Hs.83765 AA488803
688 AA151930:
Homo sapiens mRNA; cDNA DKFZp434Pl 16 (from clone DKFZp434Pl 16);
689
complete cds Hs.103378 AA431133: Homo sapiens mRNA; cDNA DKFZp564D156 (from clone DKFZp564D156)
690
Hs.9927 T55704:
691 ESTs: Hs.32204 R93719
PRPSAPl : phosphoribosyl pyrophosphate synthetase-associated protein 1 Hs.77498
692
R20005
ZNF42: zinc finger protein 42 (myeloid-specific retinoic acid- responsive) Hs.169832
693
AA987906
694 **ESTs: Hs.43712 N25936
RUNX1 : runt-related transcription factor 1 (acute myeloid leukemia 1; amll oncogene)
695
Hs.129914 AA146826
Homo sapiens mRNA; cDNA DKFZp547C244 (from clone DKFZp547C244) Hs.9460
696
T64452:
697 TYMS: thymidylate synthetase Hs.82962 AA663310
698 MGC5528: hypothetical protein MGC5528 Hs.315167 AA843451
699 ESTs: Hs.268685 R22952
SFPQ: splicing factor proline/glutamine rich (polypyrimidine tract-binding protein-
700
associated) Hs.180610 AA418910
701 ESTs: Hs.155105 AI221390
702 FLJ10624: hypothetical protein FLJ10624 Hs.306000 AA489592
703 TRIP8: thyroid hormone receptor interactor 8 Hs.6685 AA425205
704 DNAJB6: DnaJ (Hsp40) homolog, subfamily B, member 6 Hs.181195 AA496105
705 ESTs: Hs.18331 T98244
706 RBM14: RNA binding motif protein 14 Hs.l 1170 AA421233
SCYA2: small inducible cytokine A2 (monocyte chemotactic protein 1, homologous to
707 mouse Sig-je) Hs.303649 AA425102 MCP-l=MCAF=small inducible cytokine
A2=JE=chemokine
708 MGC4161 : hypothetical protein MGC4161 Hs.177688 AI224867
709 TUBB2: tubulin, beta, 2 Hs.251653 AA888148
710 FLJ20280: hypothetical protein FLJ20280 Hs.270134 N74086
711 TERA: TERA protein Hs. l 80780 AA465096
712 CPS1 : **carbamoyl-phosphate synthetase 1, mitochondrial Hs.50966 N68399
713 KIAA0802: KIAA0802 protein Hs.27657 W55875
714 FYN: FYN oncogene related to SRC, FGR, YES Hs.169370 N22980
715 Homo sapiens PR02751 mRNA, complete cds Hs.283978 H12784:
716 CLTH: Clathrin assembly lymphoid-myeloid leukemia gene Hs.7885 AA441930
717 CHMP1.5: CHMP1.5 protein Hs.42733 W85875
SMARCB1 : SWI/SNF related, matrix associated, actin dependent regulator of
718
chromatin, subfamily b, member 1 Hs.159971 AA446018
719 AA487823: SRF=c-fos serum response element-binding transcription facto
720 **ESTs: Hs.130741 AA608725 721 Homo sapiens cDNA FLJ10976 fis, clone PLACE1001399 Hs.296323 R36085:
722 FLJ20036: hypothetical protein FLJ20036 Hs.32922 N91145
723 C110RF5: chromosome 11 open reading frame 5 Hs.121025 AA776702
724 AF3P21 : SH3 protein Hs.102929 N94372
725 LOC54104: hypothetical protein Hs.12871 H05934
726 DF: D component of complement (adipsin) Hs.155597 AA233549
111 CEP4: Cdc42 effector protein 4; binder of Rho GTPases 4 Hs.3903 AA449061
728 KIF5B: kinesin family member 5B Hs.149436 AA644218
729 MGC5627: hypothetical protein MGC5627 Hs.237971 H02336
G3BP: Ras-GTPase-activating protein SH3 -domain-binding protein Hs.220689
730
AA449834
731 ESTs: Hs.293987 AA229758
732 ESTs: Hs.36828 AA194796
733 Homo sapiens mRNA for FLJ00101 protein, partial cds Hs.221600 W92262:
734 Homo sapiens cDNA: FLJ21288 fis, clone COL01927 Hs.6019 R07184:
ESTs,: Weakly similar to 1207289A reverse transcriptase related protein [H. sapiens]
735
Hs.250594 H86813
736 Homo sapiens cDNA FLJ11941 fis, clone HEMBB1000649 Hs.124106 AI301573:
737 ESTs: Hs.24908 H77726
738 TOB2: transducer of ERBB2, 2 Hs.4994 AA486088
739 ESTs: Hs.143900 AI193212
740 Homo sapiens clone FLC0675 PRO2870 mRNA, complete cds Hs.306117 H16589:
741 ESTs,: Weakly similar to KIAA0638 protein [H.sapiens] Hs.296288 T83657
742 FLJ20039: hypothetical protein FLJ20039 Hs.267448 AA448268
743 RPA2: replication protein A2 (32kD) Hs.79411 R13557
744 GAS1 : growth arrest-specific 1 Hs.65029 AA025819
Human: DNA sequence from clone 967N21 on chromosome 20pl2.3-13. Contains the
745 CHGB gene for chromogranin B (secretogranin 1, SCG1), a pseudogene similar to part of KIAA0172, the gene for a novel protein Hs.88959 R56678
746 ESTs: Hs.21175 AI341642
747 LBC: lymphoid blast crisis oncogene Hs.301946 AA135716
748 ESTs: Hs.194595 R06761
749 MGC4707: hypothetical protein MGC4707 Hs.291003 R14653
750 ZNF183: zinc finger protein 183 (RING finger, C3HC4 type) Hs.64794 AA132766
751 RAD 18: postreplication repair protein hRAD18p Hs.21320 R59197
EIF4EBP2: **eukaryotic translation initiation factor 4E binding protein 2 Hs.278712
752
H15159
**Homo sapiens mRNA; cDNA DKFZp586M0723 (from clone DKFZp586M0723)
753
Hs.27860 AA446650:
754 ORC3L: origin recognition complex, subunit 3 (yeast homolog)-like Hs.74420 H99257 CDK7: cyclin-dependent kinase 7 (homo log of Xenopus MO 15 cdk-activating kinase)
755
Hs.184298 AI311067
756 USP10: ubiquitin specific protease 10 Hs.78829 AA455233
757 KIAA0733: TAKl-binding protein 2; KIAA0733 protein Hs.109727 AA931658
758 R89286:
ALDH4: aldehyde dehydrogenase 4 (glutamate gamma-semialdehyde dehydrogenase;
759
pyrroline-5-carboxylate dehydrogenase) Hs.77448 AA181378
760 IDN3: IDN3 protein Hs.225767 N62911
761 ESTs: Hs.50180 H48143
762 MIG2: mitogen inducible 2 Hs.75260 H29252
763 KIAA0856: KIAA0856 protein Hs.13264 R12847
764 EST: Hs.47763 N54162
Homo sapiens mRNA; cDNA DKFZp547C244 (from clone DKFZp547C244) Hs.9460
765
AA447553:
766 KIAA0855: golgin-67 Hs.182982 AA775625
767 ESTs,: Weakly similar to JH0148 nucleolin - rat [R.norvegicus] Hs.30120 R54659
768 FLJ22313: hypothetical protein FLJ22313 Hs.30211 H52061
769 ESTs: Hs.71818 AI028074
770 KIAA0618: KIAA0618 gene product Hs.295112 AA455506
771 ESTs: Hs.59413 W93056
772 ESTs: Hs.165607 AA992090
773 UBAP: ubiquitin associated protein Hs.75425 AA446016
774 HANI 1 : WD-repeat protein Hs.176600 AA725641
775 USP16: ubiquitin specific protease 16 Hs.99819 AA489619
776 ESTs: Hs.67776 AA464963
777 SM-20: similar to rat smooth muscle protein SM-20 Hs.6523 H56028
778 CCNG2: cyclin G2 Hs.79069 AA489647
Homo sapiens mRNA; cDNA DKFZp566Pl 124 (from clone DKFZp566Pl 124)
779
Hs.321022 N62953:
780 FLJ20094: hypothetical protein FLJ20094 Hs.29700 N95490
781 LOC51174: delta-tubulin Hs.270847 W33133
Homo sapiens mRNA; cDNA DKFZp434I1820 (from clone DKFZp434I1820); partial
782
cds Hs.14235 N52394:
783 FANCA: Fanconi anemia, complementation group A Hs.284153 AA644129
784 P5-1 : MHC class I region ORF Hs.1845 T58146
DNA2L: DNA2 (DNA replication helicase, yeast, homolog)-like Hs.194665
785
AA974495 KIAA0083
786 LOC51578: **adrenal gland protein AD-004 Hs.279586 AA150301
787 ESTs: Hs.326417 AA913304 CDKN2D: cyclin-dependent kinase inhibitor 2D (pl9, inhibits CDK4) Hs.29656
788
R77517 l9-INK4D=Cyclin-dependent kinase 4 inhibitor D
789 FABP1 : fatty acid binding protein 1, liver Hs.5241 AA682392
790 TERA: TERA protein Hs.180780 AA906997
791 ESTs: Hs.145383 AI253072
SLC7A5: solute carrier family 7 (cationic amino acid transporter, y+ system), member
792
5 Hs.184601 AA419176
AXL: AXL receptor tyrosine kinase Hs.83341 H15336 axl=ufo=tyrosine kinase
793
receptor
794 LOC57190: selenoprotein N Hs.8518 AA284276
795 ESTs: Hs.99037 AA443948
796 STCH: stress 70 protein chaperone, microsome-associated, 60kD Hs.288799 H85311
797 ESTs: Hs.88523 AA278591 Unknown UG Hs.88523 ESTs
798 ESD: ** esterase D/formylglutathione hydrolase Hs.82193 AA250931
799 ESTs: Hs.122444 R31021
800 ESTs: Hs.283127 AI291262
801 KIAA0480: **KIAA0480 gene product Hs.92200 H91332
802 HP1-BP74: HP1-BP74 Hs.142442 AA598791
**ESTs,: Moderately similar to ALU1 HUMAN ALU SUBFAMILY J SEQUENCE
803
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.144662 AA987667
804 TTF2: transcription termination factor, RNA polymerase II Hs.142157 AI023603
805 ESTs: Hs.13740 T70541
806 DJ37E16.5: hypothetical protein dJ37E16.5 Hs.5790 AA400021
807 CDH24: cadherin-like 24 Hs.155912 AI732266
808 DJ465N24.2.1 : ** hypothetical protein dJ465N24.2.1 Hs.8084 AA932375
809 ESTs,: Weakly similar to S57447 HPBRII-7 protein [H.sapiens] Hs.16346 AA410490
810 Homo sapiens cDNA: FLJ23285 fis, clone HEP09071 Hs.90424 AI005038:
811 KRAS2: v-Ki-ras2 Kirsten rat sarcoma 2 viral oncogene homolog Hs.184050 N95249
812 FLJ20038: hypothetical protein FLJ20038 Hs.72071 H96090
ESTs,: Weakly similar to ALU4 HUMAN ALU SUBFAMILY SB2 SEQUENCE
813
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.28848 AA486607
814 H2AFN: H2A histone family, member N Hs.134999 AI095013
815 RERE: arginine-glutamic acid dipeptide (RE) repeats Hs.194369 AA490249
816 USP1 : ubiquitin specific protease 1 Hs.35086 T55607
TIP47: cargo selection protein (mannose 6 phosphate receptor binding protein)
817
Hs.140452 AA416787
KIAA0135: KIAA0135 protein Hs.79337 AA427740 KIAA0135=related to pim-1
818
kinase
819 ESTs: Hs.214410 T95273
820 PPP1R2: protein phosphatase 1 , regulatory (inhibitor) subunit 2 Hs.267819 N52605 821 Homo sapiens cDNA: FLJ21210 fis, clone COL00479 Hs.325093 AA978323:
822 CSNK2A2: casein kinase 2, alpha prime polypeptide Hs.82201 AA054996
823 HSRTSBETA: rTS beta protein Hs. l 80433 N66132
824 FLJ13110: hypothetical protein FLJ13110 Hs.7358 AA431233
825 ESTs: Hs.238797 N30704
826 FYN: FYN oncogene related to SRC, FGR, YES Hs.169370 N35086
827 RBM8A: RNA binding motif protein 8A Hs.65648 AA448402
828 ESTs: Hs.21906 AA608546
829 ESTs: Hs.128081 AA971042
830 PP591 : hypothetical protein PP591 Hs. l 18666 AA626336
831 N63866:
832 HM74: putative chemokine receptor; GTP -binding protein Hs.l 37555 R02739
833 MIDI : midline 1 (Opitz/BBB syndrome) Hs.27695 AA598640
834 KIAA1586: KIAA1586 protein Hs.180663 AA938639
835 Homo sapiens clone CDABP0014 mRNA sequence Hs.92679 AA443139:
836 HSU79274: protein predicted by clone 23733 Hs.150555 AA451900
AOC3: amine oxidase, copper containing 3 (vascular adhesion protein 1) Hs.198241
837
AA036974
838 AA548037:
839 FLJ10154: hypothetical protein FLJ10154 Hs.179972 AA457133
840 THBS1 : thrombospondin 1 Hs.87409 AA464532
841 DNAJB6: DnaJ (Hsp40) homolog, subfamily B, member 6 Hs.181195 AA431203
842 KIAA1547: KIAA1547 protein Hs.31305 AI216623
843 GATA2: GATA-binding protein 2 Hs.760 R32405
844 ESTs: Hs.176950 R82522
845 KIAA1018: KIAA1018 protein Hs.5400 AA156859
B4GALT1 : **UDP-Gal:betaGlcNAc beta 1,4- galactosyltransferase, polypeptide 1
846
Hs.198248 AA043795
847 HMGCR: 3-hydroxy-3-methylglutaryl-Coenzyme A reductase Hs. l 1899 AA779417
848 ESTs,: Weakly similar to 1819485A CENP-E protein [H.sapiens] Hs. l 67652 H94466
849 ESTs: Hs.294088 AA971073
KIAA1637: coactivator independent of AF-2 (CIA); KIAA1637 protein Hs.288140
850
AA918007
851 HSPC196: hypothetical protein Hs.239938 H66023
DRl : down-regulator of transcription 1, TBP-binding (negative cofactor 2) Hs.l 6697
852
AA132007
853 CG1I: putative cyclin Gl interacting protein Hs. l 0028 AA486444
854 IGSF4: immunoglobulin superfamily, member 4 Hs.70337 AA487505
855 ESTs: Hs.179309 AA664350
856 HSPC163: HSPC163 protein Hs.108854 AA053139 857 FLJ12788: hypothetical protein FLJ12788 Hs.20242 AI061317
858 FEM1B: FEM-1 (C.elegans) homolog b Hs.6048 H82273
859 FXR1 : fragile X mental retardation, autosomal homolog 1 Hs.82712 N62761
860 NCOA3: nuclear receptor coactivator 3 Hs.225977 AA 156793
861 H2BFB: H2B histone family, member B Hs.180779 N33927
862 ESTs: Hs.23830 AA460601
CDK7: cyclin-dependent kinase 7 (homolog of Xenopus MO 15 cdk-activating kinase)
863
Hs.184298 AA031961 CAK=cdk7=NRTALRE=sdk=CDK activating kinase
864 FLJ20259: hypothetical protein FLJ20259 Hs.9956 T55949
865 Homo sapiens cDNA FLJ20678 fis, clone KAIA4163 Hs.143601 T95823:
866 RPS19: ribosomal protein S19 Hs.298262 T72208
Homo sapiens mRNA; cDNA DKFZp434M0420 (from clone DKFZp434M0420)
867
Hs.326048 AA443976:
868 TP53: tumor protein p53 (Li-Fraumeni syndrome) Hs.1846 R39356 p53
869 FBI1 : HIV-1 inducer of short transcripts binding protein Hs.104640 R06252
GOT1 : glutamic-oxaloacetic transaminase 1, soluble (aspartate aminotransferase 1)
870
Hs.597 H22855
871 FLJ21434: hypothetical protein FLJ21434 Hs.298503 AA680129
872 DNMT2: DNA (cytosine-5-)-methyltransferase 2 Hs.97681 R95731
873 ESTs: Hs.55272 W02785
874 H2BFQ: H2B histone family, member Q Hs.2178 AA010223
875 NFIC: nuclear factor I/C (CCAAT-binding transcription factor) Hs.184771 N20996
876 NPTX1 : neuronal pentraxin I Hs.84154 H22445
877 TLOC1 : translocation protein 1 Hs.8146 AA450205
MGC5302: endoplasmic reticulum resident protein 58; hypothetical protein MGC5302
878
Hs.44970 N39195
879 ACTR2: ARP2 (actin-related protein 2, yeast) homolog Hs.42915 AA032090
880 AI287555:
ABCA7: ATP-binding cassette, sub-family A (ABC1), member 7 Hs.134514
881
AI668632
COL7A1 : collagen, type VII, alpha 1 (epidermolysis bullosa, dystrophic, dominant and
882
recessive) Hs.1640 AA598507
883 RFC2: replication factor C (activator 1) 2 (40kD) Hs.139226 AA663472
884 FLJ22583: hypothetical protein FLJ22583 Hs.287700 AA135836
885 **ESTs,: Weakly similar to ORF2 [M.musculus] Hs.172208 AI820570
886 ESTs: Hs.21667 R15709
887 RBBP4: retinoblastoma-binding protein 4 Hs.16003 AA705035
Homo sapiens mRNA; cDNA DKFZp434J1027 (from clone DKFZp434J1027); partial
888
cds Hs.22908 R20166:
889 ESTs: Hs.166539 AI080987 NKTR: natural killer-tumor recognition sequence Hs.241493 AA279666 NK-tumor
890
recognition protein=cyclophilin-related protein
891 MUC1 : mucin 1, transmembrane Hs.89603 AA486365
892 AP4B1 : adaptor-related protein complex 4, beta 1 subunit Hs.28298 AA481045
893 ESTs: Hs.94943 AA452165
894 MITF: microphthalmia-associated transcription factor Hs.166017 N66177
895 ESTs: Hs.183299 AA286914 Unknown UG Hs.183299 ESTs sc_id2032
896 BAG3: BCL2-associated athanogene 3 Hs.15259 AI269958
897 INSR: insulin receptor Hs.89695 AA001106
898 TRIP: TRAF interacting protein Hs.21254 AA186426
899 EST: Hs.307975 R22182
**Homo sapiens cDNA: FLJ23037 fis, clone LNG02036, highly similar to HSU68019
900
Homo sapiens mad protein homolog (hMAD-3) mRNA Hs.288261 W72201 :
HLA-DNA: major histocompatibility complex, class II, DN alpha Hs.l 1135
901
AA702254 Major histocompatibility complex, class II, DN alpha
902 FLJ10392: ** hypothetical protein FLJ10392 Hs.20887 AI261305
903 MPHOSPH1 : **M-phase phosphoprotein 1 Hs.240 N63752
904 STAG1 : stromal antigen 1 Hs.286148 R36160
905 USP1 : ubiquitin specific protease 1 Hs.35086 AA970066
ESTs,: Moderately similar to ALU4 HUMAN ALU SUBFAMILY SB2 SEQUENCE
906
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.l 81315 AA448251
907 PA26: p53 regulated PA26 nuclear protein Hs.14125 AA447661
908 ESTs,: Weakly similar to zinc finger protein [H.sapiens] Hs.71243 N92478
909 SH3PX1 : SH3 and PX domain-containing protein SH3PX1 Hs.7905 R69163
910 **Homo sapiens cDNA: FLJ22554 fis, clone HSI01092 Hs.93842 H58317:
911 RPS25: ribosomal protein S25 Hs.l 13029 AA779404
ESTs,: Weakly similar to A49134 Ig kappa chain V-I region [H.sapiens] Hs.5890
912
N34799 fra-2=fos-related antigen 2
913 TXNRDl : thioredoxin reductase 1 Hs.l 3046 AA453335 Thioredoxin reductase
914 **ESTs: Hs.184378 N77828
915 GCSH: glycine cleavage system protein H (aminomethyl carrier) Hs.77631 R71327
916 Homo sapiens cDNA FLJ11904 fis, clone HEMBB1000048 Hs.285519 AA447098:
NCOA3: nuclear receptor coactivator 3 Hs.225977 H51992 AIBl=Amplified in Breast
917
Cancer=TRAM- 1 =RAC 3 = ACTR=C AGH 16=nucl
918 FLJ20159: hypothetical protein FLJ20159 Hs.288809 R33122
919 IL7R: interleukin 7 receptor Hs.237868 AA487121
920 RAB23: RAB23, member RAS oncogene family Hs.94769 AA134569
921 ESTs: Hs.132493 AA923168
922 ESTs: Hs.87507 AA236015 SHC1 : SHC (Src homology 2 domain-containing) transforming protein 1 Hs.81972
923
R52960
924 KIAA1321 : KIAA1321 protein Hs.24336 W37999
925 GLI: glioma-associated oncogene homolog (zinc finger protein) Hs.2693 AI373071
926 ESTs: Hs.183299 AA291137 Unknown UG Hs.183299 ESTs sc_id2032
927 GPR 6: G protein-coupled receptor kinase 6 Hs.76297 AA291284
928 ESTs: Hs.93704 AA702684
929 CAPS: calcyphosine Hs.26685 AA858390
930 Homo sapiens cDNA FLJ10976 fis, clone PLACE1001399 Hs.296323 R27711 :
931 C6: complement component 6 Hs.1282 N59396
UBE2D3: ubiquitin-conjugating enzyme E2D 3 (homologous to yeast UBC4/5)
932
Hs.l 18797 AA465196
DDX8: DEAD/H (Asp-Glu- Ala-Asp/His) box polypeptide 8 (R A helicase)
933
Hs.171872 AA465387 RNA helicase (HRH1)
934 DKFZP434B168: DKFZP434B168 protein Hs.48604 N62684
935 FLJ10512: hypothetical protein FLJ10512 Hs.93581 T39933
Homo sapiens mRNA; cDNA DKFZp564F093 (from clone DKFZp564F093)
936
Hs.l 8724 W87709:
937 F8A: coagulation factor Vlll-associated (intronic transcript) Hs.83363 AA463924
938 HSU53209: transformer-2 alpha (htra-2 alpha) Hs.24937 AA465172
939 UBQLN2: ubiquilin 2 Hs.4552 R43580
940 EIF2C2: eukaryotic translation initiation factor 2C, 2 Hs.193053 N93082
941 Homo sapiens mRNA for FLJ00012 protein, partial cds Hs.21051 HI 7645:
942 KIAA0841 : KIAA0841 protein Hs.7426 R20299
KCNAB2: potassium voltage-gated channel, shaker-related subfamily, beta member 2
943
Hs.298184 H14383
KIAA1637: coactivator independent of AF-2 (CIA); KIAA1637 protein Hs.288140
944
AA521358
945 ESTs: Hs.27379 H17455
946 FLJ11323: hypothetical protein FLJ11323 Hs.25625 R49707
947 SSP29: acidic protein rich in leucines Hs.84264 AA489201
948 ESTs: Hs.69280 AA486011
ADAMTS 1 : a disintegrin-like and metalloprotease (reprolysin type) with
949
thrombospondin type 1 motif, 1 Hs.8230 AA057170
950 ESTs: Hs.43466 N23889
MLLT4: myeloid/lymphoid or mixed-lineage leukemia (trithorax (Drosophila)
951
homolog); translocated to, 4 Hs.l 00469 AA010818
952 ESTs: Hs.271034 AA406581
953 LMNB1 : lamin Bl Hs.89497 AA983462
954 Homo sapiens cDNA FLJ13547 fis, clone PLACE1007053 Hs.7984 AA629264: 955 PTMS: parathymosin Hs.171814 R10451
956 H2AFL: H2A histone family, member L Hs.28777 AI268551
957 FLJ21603: hypothetical protein FLJ21603 Hs.129691 R72794
958 FLJ13287: hypothetical protein FLJ13287 Hs.53263 AA621725
959 CXCR4: chemokine (C-X-C motif), receptor 4 (fusin) Hs.89414 AA479357
960 INSM1 : insulinoma-associated 1 Hs.89584 R38640
961 FREQ: frequenin (Drosophila) homolog Hs.301760 H16821
962 LOC58486: transposon-derived Busterl transposase-like protein Hs.25726 AA868020
SMARCD1 : SWI/SNF related, matrix associated, actin dependent regulator of
963
chromatin, subfamily d, member 1 Hs.79335 H91691
964 ESTs: Hs.242998 T96522
965 INADL: PDZ domain protein (Drosophila inaD-like) Hs.321197 AA005153
966 ESTs,: Weakly similar to putative pl50 [H.sapiens] Hs.37751 AA436174
967 MGC5338: hypothetical protein MGC5338 Hs.99598 H50550
968 W85890:
969 NUCKS: similar to rat nuclear ubiquitous casein kinase 2 Hs.l 18064 AI053436
970 Homo sapiens clone 25110 mRNA sequence Hs.27262 H18031 :
971 AI333214:
972 GAS41 : glioma-amplified sequence-41 Hs.4029 T62072
973 LOC51170: retinal short-chain dehydrogenase/reductase retSDR2 Hs.12150 N79745
974 H2BFG: **H2B histone family, member G Hs. l 82137 R98472
ABCC1 : **ATP-binding cassette, sub-family C (CFTR/MRP), member 1 Hs.89433
975
AA424804
976 EFNA1 : ephrin-Al Hs.1624 AA857015
Homo sapiens mRNA; cDNA DKFZp434A1014 (from clone DKFZp434A1014);
977
partial cds Hs.278531 H00596:
PPP2CA: protein phosphatase 2 (formerly 2A), catalytic subunit, alpha isoform
978
Hs.91773 AA599092
979 ESTs,: Weakly similar to unnamed protein product [H.sapiens] Hs. l 18338 W85843
980 Homo sapiens cDNA FLJ11643 fis, clone HEMBA1004366 Hs.l 11496 AA598803:
ESTs,: Moderately similar to ALUE HUMAN ! ! ! ! ALU CLASS E WARNING
981
ENTRY ! ! ! [H.sapiens] Hs.125407 AA878944
ESTs,: Moderately similar to ALU1 HUMAN ALU SUBFAMILY J SEQUENCE
982
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.144662 AI191290
983 KIAA0916: KIAA0916 protein Hs. l 51411 R91388
984 CDC25A: cell division cycle 25 A Hs.l 634 R09062
985 PRIM2A: primase, polypeptide 2A (58kD) Hs.74519 R61073
986 DSP: desmoplakin (DPI, DPII) Hs.74316 H90899
987 KIAA0101 : KIAA0101 gene product Hs.81892 W68219
988 ESTs,: Weakly similar to putative pl50 [H.sapiens] Hs.268026 AA411454 989 ESTs: Hs. l 8140 T97707
990 H2AFL: H2A histone family, member L Hs.28777 AA457566
991 Homo sapiens mRNA for KIAA1700 protein, partial cds Hs.20281 H00287:
992 STAG3: stromal antigen 3 Hs.20132 AA453028
993 ZNF207: zinc finger protein 207 Hs.62112 N59119
994 BMP6: bone morphogenetic protein 6 Hs.285671 AA424833
995 ESTs,: Moderately similar to sertolin [R.norvegicus] Hs.91192 H60690
996 LOC51064: ** glutathione S-transferase subunit 13 homolog Hs.279952 W88497
997 NUCKS: similar to rat nuclear ubiquitous casein kinase 2 Hs.l 18064 AA927182
ESTs,: Weakly similar to T00370 hypothetical protein KIAA0659 [H. sapiens]
998
Hs.131899 W93155
999 FLJ13057: hypothetical protein FLJ13057 similar to germ cell-less Hs.243122 R23254
1000 ESTs: Hs.144796 AI219737
1001 FLJ10511 : hypothetical protein FL J 10511 Hs.106768 R25877
1002 DKFZP564A122: DKFZP564A122 protein Hs.187991 N31577
1003 ODF2: outer dense fibre of sperm tails 2 Hs.129055 AA400407
1004 AMY2A: amylase, alpha 2A; pancreatic Hs.278399 R64129
1005 **ESTs,: Weakly similar to plakophilin 2b [H.sapiens] Hs.12705 N91589
CYP1B1 : cytochrome P450, subfamily I (dioxin-inducible), polypeptide 1 (glaucoma
1006
3, primary infantile) Hs.154654 AA029776
1007 CAPN7: calpain 7 Hs.7145 N46420
1008 FLJ20069: hypothetical protein FLJ20069 Hs.273294 AA229966
1009 FLJ10618: hypothetical protein FLJ10618 Hs.42484 AA478847
KIAA1637: **coactivator independent of AF-2 (CIA); KIAA1637 protein Hs.288140
1010
AA452531
1011 FLJ20004: ** hypothetical protein FLJ20004 Hs.17311 AA487895
1012 FLJ12892: hypothetical protein FL J 12892 Hs.17731 AA670363
1013 PLU-1 : putative DNA/chromatin binding motif Hs.143323 AA464869
1014 **ESTs: Hs.36828 AA418448
1015 KIAA0586: KIAA0586 gene product Hs.77724 AA905278
MTHFD2: methylene tetrahydro folate dehydrogenase (NAD+ dependent),
1016
methenyltetrahydrofolate cyclohydrolase Hs.l 54672 AA480994
1017 BRF1 : **butyrate response factor 1 (EGF-response factor 1) Hs.85155 AA424743
TFAP2A: transcription factor AP-2 alpha (activating enhancer-binding protein 2 alpha)
1018
Hs.l8387 R38044
1019 VIL2: villin 2 (ezrin) Hs.155191 AA411440
1020 SDC1 : syndecan 1 Hs.82109 AA074511
1021 RNTRE: related to the N terminus of tre Hs.278526 AA281057
1022 HSPC207: hypothetical protein Hs.75798 H99997
1023 FLJ22376: hypothetical protein FLJ22376 Hs.29341 AI199155 1024 RNF10: ring finger protein 10 Hs.5094 H73586
1025 PNN: pinin, desmosome associated protein Hs.44499 AA707321
1026 FLJ20516: hypothetical protein FLJ20516 Hs.70811 AA122393
1027 RPL13A: ribosomal protein L13a Hs. l 19122 AI254200
1028 H2BFB: H2B histone family, member B Hs.l 80779 AA885642
OGT: O-linked N-acetylglucosamine (GlcNAc) transferase (UDP-N-
1029
acetylglucosamine:polypeptide-N-acetylglucosaminyl transferase) Hs.l 00293 R13317
1030 KIAA0155: KIAA0155 gene product Hs.l 73288 AA133684
1031 ILF2: interleukin enhancer binding factor 2, 45kD Hs.75117 H95638
Homo sapiens mRNA; cDNA DKFZp586I1518 (from clone DKFZp586I1518)
1032
Hs.21739 AA287917:
1033 PKNOX1 : PBX/knotted 1 homeobox 1 Hs.158225 AI350546
KMO: **kynurenine 3-monooxygenase (kynurenine 3 -hydroxylase) Hs.107318
1034
AA044326
1035 VCAMl : vascular cell adhesion molecule 1 Hs.109225 H16591 CD106=VCAM-1
1036 N54811 :
1037 KIAA0618: KIAA0618 gene product Hs.295112 H81940
MAFG: v-maf musculoaponeurotic fibrosarcoma (avian) oncogene family, protein G
1038
Hs.252229 N21609 MafG=basic-leucine zipper transcription factor
1039 MATN2: matrilin 2 Hs.19368 AA071473
1040 HOXB4: homeo box B4 Hs.126666 AA918749
1041 FLJ10466: hypothetical protein FLJ10466 Hs.121073 AA453607
1042 FLJ22557: hypothetical protein FLJ22557 Hs.106101 AA127879
1043 EST: Hs.149260 AI247680
1044 KIAA0677: KIAA0677 gene product Hs.155983 AA026751
1045 EST: Hs.l04123 AA197344
1046 UCP4: uncoupling protein 4 Hs.40510 H94680
1047 EST: Hs.144224 N93807
1048 GATA2: GATA-binding protein 2 Hs.760 H00625 GATA-binding protein 2
1049 ESTs: Hs.14743 H61082
1050 EST: Hs.l 16174 AA626786
ITGB3: integrin, beta 3 (platelet glycoprotein Ilia, antigen CD61) Hs.87149
1051
AA666269
1052 FLJ23399: hypothetical protein FLJ23399 Hs.299883 R19895
1053 ESTs: Hs.21734 N72976
1054 FLJ20425: hypothetical protein FLJ20425 Hs.71040 AA424566
1055 CUL4A: cullin 4A Hs. l 83874 AA598836
PTP4A1 : protein tyrosine phosphatase type IVA, member 1 Hs.227777 R61007
1056
protein tyrosine phosphatase PTPCAAX1 (hPTPCAAXl)
1057 ESTs: Hs.7913 N35592 GROl : GROl oncogene (melanoma growth stimulating activity, alpha) Hs.789
1058
W46900
1059 ESTs,: Moderately similar to NRD2 convertase [H.sapiens] Hs.309734 H78796
1060 FLJ10826: hypothetical protein FLJ10826 Hs.24809 AA486738
1061 TOM34: translocase of outer mitochondrial membrane 34 Hs.76927 AA457118
1062 H2AFL: H2A histone family, member L Hs.28777 AA452933
D10S170: **DNA segment, single copy, probe pH4 (transforming sequence, thyroid-
1063
l, Hs.315591 N35493
SCYA2: small inducible cytokine A2 (monocyte chemotactic protein 1, homologous to
1064 mouse Sig-je) Hs.303649 T77816 MCP-l=MCAF=small inducible cytokine
A2=JE=chemokine
1065 FLJ10688: hypothetical protein FLJ10688 Hs. l 18793 AA465358
1066 PTD017: PTD017 protein Hs.274417 AA160498
1067 KIAA0026: MORF-related gene X Hs.173714 AA676604
1068 BMP2: bone morphogenetic protein 2 Hs.73853 AA489383
1069 MNT: MAX binding protein Hs.25497 AA455508
1070 KIAA1170: KIAA1170 protein Hs.268044 H80507
1071 CRYBA1 : crystallin, beta Al Hs.46275 AA487614
1072 KATNAl : katanin p60 (ATPase-containing) subunit A 1 Hs.289099 AA609740
1073 Homo sapiens cDNA FLJ20796 fis, clone COL00301 Hs. l 13994 N53458:
1074 CEP4: Cdc42 effector protein 4; binder of Rho GTPases 4 Hs.3903 W32509
1075 ESTs: Hs. l 17261 AA682521
CYP1B1 : cytochrome P450, subfamily I (dioxin-inducible), polypeptide 1 (glaucoma
1076
3, primary infantile) Hs.154654 AA040872
1077 ALTE: Ac-like transposable element Hs.9933 AA630498
1078 RAD51 : RAD51 (S. cerevisiae) homolog (E coli RecA homolog) Hs.23044 AA873056
1079 MAN1A2: mannosidase, alpha, class 1A, member 2 Hs.239114 R78501
1080 H53763:
1081 MET: met proto-oncogene (hepatocyte growth factor receptor) Hs.285754 AA410591
DYRK1 A: dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1 A
1082
Hs.75842 AA676749
1083 ARHGAP8: **Rho GTPase activating protein 8 Hs.102336 AA037410
1084 LM04: LIM domain only 4 Hs.3844 H27986
1085 ADCY6: adenylate cyclase 6 Hs.12373 AA148044
1086 EST: Hs.l35448 AI078552
1087 NCOA3: nuclear receptor coactivator 3 Hs.225977 W46433
1088 DNAJB4: DnaJ (Hsp40) homolog, subfamily B, member 4 Hs.41693 AA081471
1089 NAB 1 : NGFI-A binding protein 1 (ERG1 binding protein 1) Hs. l 07474 AA486027
ESTs,: Weakly similar to T08663 hypothetical protein DKFZp547G0910.1 [H.sapiens]
1090
Hs.172084 N63646 KIAA0735: KIAA0735 gene product; synaptic vesicle protein 2B homolog Hs.8071
1091
R56082
GNBl : guanine nucleotide binding protein (G protein), beta polypeptide 1 Hs.215595
1092
AA487912
1093 Homo sapiens mRNA for KIAA1716 protein, partial cds Hs.21446 R49763:
1094 KINESIN: HEAVY CHAIN
CCND1 : cyclin Dl (PRAD1 : parathyroid adenomatosis 1) Hs.82932 AA487486
1095
Cyclin Dl=BCLl=PRADl=Translocated in mantle cell leukemia
1096 ESTs: Hs.106129 R56716
1097 AA431931 :
1098 PSEN1 : presenilin 1 (Alzheimer disease 3) Hs.3260 AA403083
1099 ESTs: Hs.193804 AA010918
1100 DKFZp762P2111 : hypothetical protein DKFZp762P2111 Hs.14217 AA429586
1101 KIAA1350: KIAA1350 protein Hs.101799 W37627
1102 FLJ20847: hypothetical protein FLJ20847 Hs.13479 H16996
1103 HDCMA18P: HDCMA18P protein Hs.278635 N64387
1104 FLJ12890: hypothetical protein FLJ12890 Hs.43299 N62475
1105 ESTs: Hs.127453 AA973625
1106 BAIAP2: BAI1 -associated protein 2 Hs.7936 R60328
1107 ESTs: Hs.317584 AA191424
1108 DKFZP434J046: DKFZP434J046 protein Hs. l 16244 AI024401
1109 ESTs: Hs. l 14055 AA701352
1110 ESTs: Hs.44380 N93122
1111 ESTs: Hs.20142 AA625570
1112 UBL3: ubiquitin-like 3 Hs.173091 T82438
1113 H2AFL: H2A histone family, member L Hs.28777 N50797
1114 SUCLG2: **succinate-CoA ligase, GDP-forming, beta subunit Hs.247309 N68557
1115 ZWINT: ZW10 interactor Hs.42650 AA706968
1116 FLJ10583: hypothetical protein FLJ10583 Hs.105633 R00425
1117 FLJ20552: hypothetical protein FLJ20552 Hs.69554 AA463982
FADD: Fas (TNFRSF6)-associated via death domain Hs.86131 AA430751
1118
FADD=MORT
1119 SFRS7: splicing factor, arginine/serine-rich 7 (35kD) Hs.184167 AA418813
1120 RAD54L: RAD54 (S.cerevisiae)-like Hs.66718 AI372035
1121 MYLE: MYLE protein Hs.l 1902 T68845
1122 LOC51334: mesenchymal stem cell protein DSC54 Hs.157461 R63841
1123 PRIM2A: primase, polypeptide 2A (58kD) Hs.74519 AA434404
1124 KIAA0056: KIAA0056 protein Hs.13421 AA430545
ESTs,: Moderately similar to ALU7 HUMAN ALU SUBFAMILY SQ SEQUENCE
1125
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.82590 N53024 1126 ESTs: Hs.117269 AA705050
1127 NSAP1 : NS1 -associated protein 1 Hs.155489 AA186327
CEACAM5: carcinoembryonic antigen-related cell adhesion molecule 5 Hs.220529
1128
AA130547
FLJ11021 : hypothetical protein FLJ11021 similar to splicing factor, arginine/serine-
1129 rich 4 Hs.81648 AA291183 Unknown UG Hs.202583 ESTs, Weakly similar to
arginine-rich
1130 FOSL1 : FOS-like antigen-1 Hs.283565 T82817 fra-l=fos-related antigen 1
1131 U3-55K: U3 snoRNP-associated 55-kDa protein Hs.153768 AA465355
1132 DNAJC6: DnaJ (Hsp40) homolog, subfamily B, member 6 Hs.44896 AA455940
1133 KIAA1382: amino acid transporter 2 Hs.298275 R27255 Similar to transporter protein
PCAF: p300/CBP-associated factor Hs.199061 N74637 P/CAF=p300/CBP-associated
1134
factor
1135 ESTs: Hs.130460 AA927252
1136 ESTs: Hs. l 12570 AI014667
1137 FLJ10209: hypothetical protein FLJ10209 Hs.260150 AA454626
1138 ESTs: Hs.99014 AA485679
1139 ESTs: Hs.99621 AA464707
1140 Homo sapiens cDNA FLJ11904 fis, clone HEMBB1000048 Hs.285519 N74617:
1141 AA928536:
1142 SQSTM1 : **sequestosome 1 Hs.182248 AA931964
**Homo sapiens cDNA FLJ13700 fis, clone PLACE2000216, highly similar to
1143
SPECTRIN BETA CHAIN, BRAIN Hs.324648 AA018591 :
SLC22A3: solute carrier family 22 (extraneuronal monoamine transporter), member 3
1144
Hs.81086 AA460012
1145 FLJ22557: hypothetical protein FLJ22557 Hs.106101 H00595
1146 FLJ20539: hypothetical protein FLJ20539 Hs. l 18552 R36152
1147 AA991624:
TRAP150: thyroid hormone receptor-associated protein, 150 kDa subunit Hs.108319
1148
W85832
1149 ESTs: Hs.221847 R91557
1150 TCFL1 : transcription factor-like 1 Hs.2430 AA443950
ESTs,: Highly similar to oxytocinase splice variant 1 [H. sapiens] Hs.203271
1151
AA487918
1152 PLAB: prostate differentiation factor Hs.296638 AA450062
1153 RBM14: RNA binding motif protein 14 Hs.l 1170 AA417283
1154 EGFL5: EGF-like-domain, multiple 5 Hs.5599 W67981
1155 H2AFO: H2A histone family, member O Hs.795 AA047260
ESTs,: Weakly similar to A46661 leukotriene B4 omega-hydroxylase [H. sapiens]
1156
Hs.l69001 N45556
1157 W78784: 1158 TOP3A: topoisomerase (DNA) III alpha Hs.91175 N21546
1159 W73732: Host cell factor-l=VP16 transactivator interacting protein
CYP1B1 : cytochrome P450, subfamily I (dioxin-inducible), polypeptide 1 (glaucoma
1160
3, primary infantile) Hs.154654 AA448157 Cytochrome P450 IBl (dioxin-inducible)
1161 ESTs: Hs.135276 AI092102
1162 RHEB2: Ras homolog enriched in brain 2 Hs.279903 AA482117
1163 ESTs,: Highly similar to EF-9 [M.musculus] Hs.8366 H94467
1164 POLA: polymerase (DNA directed), alpha Hs.267289 AA707650
1165 KIAA1008: KIAA1008 protein Hs.323346 AA863115
PIK3CD: phosphoinositide-3-kinase, catalytic, delta polypeptide Hs.162808
1166
AA281652
1167 T53625:
**Homo sapiens mRNA; cDNA DKFZp434Al 114 (from clone DKFZp434Al 114)
1168
Hs.326292 AA417274:
1169 ESTs: Hs.26744 H16988
1170 FLJ13912: hypothetical protein FLJ13912 Hs.47125 W74133
Homo sapiens mRNA; cDNA DKFZp762B195 (from clone DKFZp762B195)
1171
Hs.284158 AA625574:
SSA2: Sjogren syndrome antigen A2 (60kD, ribonucleoprotein autoantigen SS-A/Ro)
1172
Hs.554 AA010351
1173 BK1048E9.5: hypothetical protein bK1048E9.5 Hs.6657 N68512
1174 TOPI : topoisomerase (DNA) I Hs.317 AA232856 Topoisomerase I
1175 ESTs: Hs. l5386 H18472
1176 KPNB1 : karyopherin (importin) beta 1 Hs.180446 AA121732
1177 MGC861 : hypothetical protein MGC861 Hs.208912 N69694
1178 PMS2L8: **postmeiotic segregation increased 2-like 8 Hs.323954 T62577
TSC22: * transforming growth factor beta-stimulated protein TSC-22 Hs. l 14360
1179
R16390
1180 C80RF1 : chromosome 8 open reading frame 1 Hs.40539 AA278836
1181 ESTs: Hs.129165 AA989211
1182 DMTF: cyclin D binding Myb-like transcription factor 1 Hs.5671 AA129860
CDC7L1 : CDC7 (cell division cycle 7, S. cerevisiae, homolog)-like 1 Hs.28853
1183
N62245 Cdc7-related kinase
1184 LOC51700: cytochrome b5 reductase b5R.2 Hs.22142 AA425316
1185 FLNA: filamin A, alpha (actin-binding protein-280) Hs.l 95464 AA598978
1186 FLJ20257: hypothetical protein FLJ20257 Hs.178011 H78675
1187 Homo sapiens cDNA FLJ13604 fis, clone PLACE1010401 Hs.23193 AA406599:
1188 ESTs: Hs.205227 R73480
SCYB14: small inducible cytokine subfamily B (Cys-X-Cys), member 14 (BRAK)
1189
Hs.24395 AA953842 MAPK8IP2: **mitogen-activated protein kinase 8 interacting protein 2 Hs.80545
1190
AA418293
ZNF42: zinc finger protein 42 (myeloid-specific retinoic acid- responsive) Hs.169832
1191
AA932642
1192 ESTs: Hs.127054 AA862450
NUDT4: nudix (nucleoside diphosphate linked moiety X)-type motif 4 Hs.92381
1193
AA425630
1194 Homo sapiens cDNA FLJ10632 fis, clone NT2RP2005637 Hs.202596 H82421 :
1195 LOC51042: zinc finger protein Hs.102419 AA033532
1196 NUMA1 : nuclear mitotic apparatus protein 1 Hs.301512 AA679293
ESTs,: Highly similar to A56429 I-kappa-B-related protein [H. sapiens] Hs.144614
1197
AA293771
1198 ESTs: Hs.127703 AA947258
1199 Homo sapiens cDNA FLJ14214 fis, clone NT2RP3003576 Hs.321236 AA903913:
NFKBIA: nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor,
1200
alpha Hs.81328 W55872 IkB alpha
1201 ESTs: Hs.120029 AA707598
ESTs,: Moderately similar to A Chain A, Human Glucosamine-6-Phosphate
1202
Deaminase Isomerase At 1.75 A [H. sapiens] Hs.21398 AA172012
1203 NFIA: nuclear factor I/A Hs.173933 AI912047
1204 RECQL4: RecQ protein-like 4 Hs.31442 AA620446
**ESTs,: Weakly similar to ALU1 HUMAN ALU SUBFAMILY J SEQUENCE
1205
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.318894 R96212
1206 Homo sapiens cDNA: FLJ21686 fis, clone COL09379 Hs.20787 R11371 :
1207 LOC57168: similar to aspartate beta hydroxylase (ASPH) Hs.184390 H17272
1208 ESTs: Hs.26096 R54109
1209 Homo sapiens OSBP-related protein 6 niRNA, complete cds Hs.318775 AA680281 :
1210 APACD: ATP binding protein associated with cell differentiation Hs.153884 N80741
1211 VIM: **vimentin Hs.297753 AI668662
1212 Homo sapiens cDNA FLJ13618 fis, clone PLACE1010925 Hs.17448 AA427980:
NR3C1 : nuclear receptor subfamily 3, group C, member 1 Hs.75772 N30428
1213
Glucocorticoid receptor
1214 Homo sapiens cDNA: FLJ21814 fis, clone HEP01068 Hs.289008 R12808:
1215 BRD7: bromodomain-containing 7 Hs.279762 AA488428
1216 MAP3K8: **mitogen-activated protein kinase kinase kinase 8 Hs.248 W42450
1217 ESTs: Hs.23213 H29336
1218 ESTs: Hs.122444 AA939019
1219 TUSP: tubby super-family protein Hs.102237 H78234
1220 KIAA1117: KIAA1117 protein Hs.278398 H01516
1221 Human: clone 137308 niRNA, partial cds Hs.322149 H91303 1222 ESTs: Hs.130214 AA456631
1223 RAB3A: RAB3A, member RAS oncogene family Hs.27744 H14230
1224 AA598795: Protein phosphatase 2 (formerly 2A), regulatory subunit B (P
1225 H2BFC: H2B histone family, member C Hs.137594 AI340654
1226 CFLAR: CASP8 and FADD-like apoptosis regulator Hs.195175 N94588
1227 CD24: CD24 antigen (small cell lung carcinoma cluster 4 antigen) Hs.286124 H59915
1228 EST: Hs.48532 N62402
1229 CCRK: cell cycle related kinase Hs.26322 H17616
1230 HECH: heterochromatin-like protein 1 Hs.278554 All 39106
1231 DKFZp5470146: hypothetical protein DKFZp5470146 Hs.91246 T80848
1232 ESTs: Hs.71574 AA135328
1233 HLXB9: homeo box HB9 Hs.37035 AI459915
1234 AA600222:
1235 SPINK5: serine protease inhibitor, Kazal type, 5 Hs.5476 W92134
1236 RNUT1 : RNA, U transporter 1 Hs.21577 AA447799
1237 Homo sapiens cDNA: FLJ23013 fis, clone LNG00740 Hs.13075 AA464543:
1238 KIAA0063: KIAA0063 gene product Hs.3094 T82263
DYRK2: dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 2 Hs.173135
1239
R63622
1240 R94947:
1241 Homo sapiens cDNA FLJ14337 fis, clone PLACE4000494 Hs.180187 AA004903:
1242 FLJ20624: hypothetical protein FLJ20624 Hs.52256 AA431909
1243 ESTs: Hs.43838 R38261
1244 FLJ23053: hypothetical protein FLJ23053 Hs.94037 R25654
1245 MGC11266: hypothetical protein MGC11266 Hs.293943 AA400456
ESTs,: Moderately similar to ALU8 HUMAN ALU SUBFAMILY SX SEQUENCE
1246
CONTAMINATION WARNING ENTRY [H.sapiens] Hs.34174 AA126603
1247 PLAUR: plasminogen activator, urokinase receptor Hs.179657 AA 147962
1248 TSG101 : tumor susceptibility gene 101 Hs.l 18910 AA670215
1249 HCNGP: transcriptional regulator protein Hs.27299 AA457232
1250 KIAA0978: KIAA0978 protein Hs.3686 AA857017
1251 ESTs: Hs.61708 AA033867
1252 ESTs: Hs.120734 AA827482
1253 ESTs: Hs.5909 AA972654
1254 CDH24: cadherin-like 24 Hs.155912 AI668564
1255 CCND1 : cyclin Dl (PRAD1 : parathyroid adenomatosis 1) Hs.82932 T77237
1256 ESTs: Hs.43148 AA284775
1257 ESTs: Hs.222566 T50982
1258 ESTs: Hs.194125 N52822
1259 EST: Hs.l54621 AI138644 1260 MAN1A2: mannosidase, alpha, class 1A, member 2 Hs.239114 R22905
1261 MAN2A2: mannosidase, alpha, class 2A, member 2 Hs.295605 AA454175
Human DNA sequence from clone 967N21 on chromosome 20pl2.3-13. Contains the
1262 CHGB gene for chromogranin B (secretogranin 1, SCG1), a pseudogene similar to part of KIAA0172, the gene for a novel protein Hs.88959 W94690
ESTs,: Highly similar to CIKG HUMAN VOLTAGE-GATED POTASSIUM
1263
CHANNEL PROTEIN KV3.4 [H.sapiens] Hs.106486 HI 1376
1264 Homo sapiens HT023 mRNA, complete cds Hs.237225 AA169496:
1265 FLJ10339: ** hypothetical protein FLJ10339 Hs.203963 H72354
1266 N66278:
1267 ESTs: Hs.6195 AA454745
1268 KIAA1404: KIAA1404 protein Hs.200317 W72798
PMAIP1 : phorbol-12-myristate- 13 -acetate-induced protein 1 Hs.96 AA458838
1269
APR=immediate-early-response gene=ATL-derived PMA-responsive
G3BP: Ras-GTPase-activating protein SH3 -domain-binding protein Hs.220689
1270
AA598628
1271 Homo sapiens cDNA: FLJ22807 fis, clone KAIA2887 Hs.261734 R26854:
1272 Homo sapiens, clone IMAGE:3535294, mRNA, partial cds Hs.80449 T57359:
1273 CDC16: CDC 16 (cell division cycle 16, S. cerevisiae, homolog) Hs.1592 AA410559
1274 FGA: **fibrinogen, A alpha polypeptide Hs.90765 AA026626
1275 ESTs: Hs.33446 N53560
1276 Homo sapiens cDNA FLJ14175 fis, clone NT2RP2002979 Hs.288613 AA054704:
1277 ESTs: Hs.44243 AA011390
Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 42408 Hs.284123
1278
R61732:
1279 ESTs: Hs.53455 AA454165
1280 FLJ11264: hypothetical protein FLJ11264 Hs. l 1260 AI219094
1281 MBD4: methyl-CpG binding domain protein 4 Hs.35947 AA010492
1282 FLJ11305: hypothetical protein FLJ11305 Hs.7049 N94612
Homo sapiens, Similar to CG5057 gene product, clone MGC:5309, mRNA, complete
1283
cds Hs.13885 AA460004:
1284 ARHB: ras homolog gene family, member B Hs.204354 H88963
1285 ITPR3: inositol 1,4,5-triphosphate receptor, type 3 Hs.77515 AA865667
1286 HMG20B: high-mobility group 20B Hs.32317 AA775743
1287 ESTs: Hs.146276 AI214204
1288 PTPN9: protein tyrosine phosphatase, non-receptor type 9 Hs.147663 AA434420
1289 Homo sapiens clone FLB9213 PR02474 mRNA, complete cds Hs.21321 AA486770:
1290 H21107:
1291 HSPC157: HSPC157 protein Hs.279842 N20480 Homo sapiens mRNA; cDNA DKFZp56402363 (from clone DKFZp56402363)
1292
Hs.321403 AA406332:
1293 ESTs: Hs.150623 AA693532
1294 EST: Hs.l88697 AA199733
CLECSF2: C-type (calcium dependent, carbohydrate-recognition domain) lectin,
1295 superfamily member 2 (activation-induced) Hs.85201 HI 1732 AICL=activation- induced C-type lectin
1296 ITPR1 : inositol 1,4,5-triphosphate receptor, type 1 Hs.198443 AA035450
1297 CHML: choroideremia-like (Rab escort protein 2) Hs.170129 R91881
1298 CDC42: cell division cycle 42 (GTP-binding protein, 25kD) Hs.146409 AA668681
1299 FKBP5: **FK506-binding protein 5 Hs.7557 AA872767
[00204] All publications and patent applications mentioned in the specification are indicative of the level of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The mere mentioning of the publications and patent applications does not necessarily constitute an admission that they are prior art to the instant application.
[00205] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims.

Claims

CLAIMS What is claimed is:
1. A method of determining gene expression in a tumor sample, comprising:
obtaining a tumor sample from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer;
determining the expression levels of a panel of genes in said tumor sample including at least 4 cell-cycle genes; and
providing a test value by (1) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (2) combining the weighted expression to provide said test value, wherein at least 75%, at least 85% or at least 95% of said plurality of test genes are cell-cycle genes.
2. The method of Claim 1, wherein at least 90% of said plurality of test genes are cell- cycle genes.
3. The method of Claim 1 or 2, wherein said determining step comprises:
measuring the amount of mR A in said tumor sample transcribed from each of between 6 and 200 cell-cycle genes; and
measuring the amount of mRNA of one or more housekeeping genes in said tumor sample.
4. The method of Claim 1 or 2 or 3, wherein the expression of at least 8 cell-cycle genes are determined and weighted.
5. A method of prognosing prostate cancer, lung cancer, bladder cancer or brain cancer, comprising:
determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of a panel of genes in said tumor sample including at least 4 cell-cycle genes; providing a test value by (1) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (2) combining the weighted expression to provide said test value, wherein at least 75%, at least 85% or at least 95% of said plurality of test genes are cell-cycle genes; and
correlating an increased level of expression of said plurality of test genes to a poor prognosis.
6. The prognosis method of Claim 5, further comprising comparing said test value to a reference value, and correlating to an increased likelihood of poor prognosis if said test value is greater than said reference value.
7. The prognosis method of Claim 5, wherein the expression levels of from 6 to about 200 cell-cycle genes are measured.
8. The method of any one of Claims 5-7, wherein said determining step comprises: measuring the amount of mR A of from 6 to about 200 cell-cycle genes in said tumor sample; and
measuring the amount of mRNA of one or more housekeeping genes in said tumor sample.
9. A method of treating cancer in a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer, comprising:
determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of a panel of genes in said tumor sample including at least 4 cell-cycle genes;
providing a test value by (1) weighting the determined expression of each of a plurality of test genes selected from said panel of genes with a predefined coefficient, and (2) combining the weighted expression to provide said test value, wherein at least 60%> or 75% of said plurality of test genes are cell-cycle genes, wherein an increased level of expression of said plurality of test genes indicates a poor prognosis; and
administering to said patient an anti-cancer drug, or recommending or prescribing or initiating active treatment if a poor prognosis is determined.
10. A diagnostic kit for prognosing cancer in a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, comprising, in a compartmentalized container:
a plurality of PCR primer pairs for PCR amplification of at least 5 test genes, wherein less than 10%, 30%> or less than 40%> of all of said at least 8 test genes are non-cell-cycle genes; and one or more PCR primer pairs for PCR amplification of at least one housekeeping gene.
11. A diagnostic kit for prognosing cancer in a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, comprising, in a compartmentalized container:
a plurality of probes for hybridizing to at least 5 test genes under stringent hybridization conditions, wherein less than 10%>, 30%> or less than 40%> of all of said at least 8 test genes are non- cell-cycle genes; and
one or more probes for hybridizing to at least one housekeeping gene.
12. A kit consisting essentially of, in a compartmentalized container:
a first plurality of PCR reaction mixtures for PCR amplification of between 5 or 10 and 300 test genes, wherein at least 50%>, at least 60%> or at least 80%> of said 5 or 10 to 300 test genes are cell-cycle genes, and wherein each reaction mixture comprises a PCR primer pair for PCR amplifying one of said test genes; and
a second plurality of PCR reaction mixtures for PCR amplification of at least one housekeeping gene.
13. The kit of any one of Claims 10-12, wherein cell-cycle genes constitute no less than 10%) of the total number of said test genes.
14. The kit of any one of Claims 10-12, wherein cell-cycle genes constitute no less than 20%) of the total number of said test genes.
15. Use of
(1) a plurality of PCR primer pairs suitable for PCR amplification of at least 4 cell-cycle genes; and
(2) one or more PCR primer pairs suitable for PCR amplification of at least one
housekeeping gene, for the manufacture of a diagnostic product for determining the expression of said test genes in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, to predict the prognosis of cancer, wherein an increased level of said expression indicates a poor prognosis or an increased likelihood of recurrence of cancer in the patient.
16. The use of Claim 15, wherein said plurality of PCR primer pairs are suitable for PCR amplification of at least 8 cell-cycle genes.
17. The use of Claim 15 or 16, wherein said plurality of PCR primer pairs are suitable for PCR amplification of from 4 to about 300 test genes, no greater than 10%, 30% or less than 50% of which being non-cell-cycle genes.
18. The use of Claim 15 or 16, wherein said plurality of PCR primer pairs are suitable for PCR amplification of from 20 to about 300 test genes, at least 25% of which being cell-cycle genes.
19. Use of
(1) a plurality of probes for hybridizing to at least 4 cell-cycle genes under stringent hybridization conditions; and
(2) one or more probes for hybridizing to at least one housekeeping gene under stringent hybridization conditions,
for the manufacture of a diagnostic product for determining the expression of said test genes in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, to predict the prognosis of cancer, wherein an increased level of said expression indicates a poor prognosis or an increased likelihood of recurrence of cancer in the patient.
20. The use of Claim 18, wherein said plurality of probes are suitable for hybridization to at least 8 different cell-cycle genes.
21. The use of Claim 18 or 19, wherein said plurality of probes are suitable for hybridization to from 4 to about 300 test genes, no greater than 10%, 30% or less than 50% of which being non-cell-cycle genes.
22. The use of Claim 18 or 19, wherein said plurality of probes are suitable for hybridization to from 20 to about 300 test genes, at least 25% of which being cell-cycle genes.
23. A system for prognosing cancer selected from prostate cancer, lung cancer, bladder cancer or brain cancer, comprising:
a sample analyzer for determining the expression levels of a panel of genes in said tumor sample including at least 4 cell-cycle genes, wherein the sample analyzer contains the tumor sample which is from a patient identified as having prostate cancer, lung cancer, bladder cancer or brain cancer, or cDNA molecules from mRNA expressed from the panel of genes; and
a first computer program means for (a) receiving gene expression data on at least 4 test genes selected from the panel of genes, (b) weighting the determined expression of each of the test genes, and (c) combining the weighted expression to provide a test value, wherein at least 50%, at least at least 75%) of at least 4 test genes are cell-cycle genes; and
a second computer program means for comparing the test value to one or more reference values each associated with a predetermined degree of risk of cancer recurrence or progression of the prostate cancer, lung cancer, bladder cancer or brain cancer.
24. A system for prognosing cancer selected from lung cancer or colon cancer, comprising:
(1) a sample analyzer for determining the expression levels of a panel of genes including at least 4 cell-cycle genes in a tumor sample from a patient identified as having lung cancer or colon cancer, wherein the sample analyzer contains the tumor sample, R A expressed from the panel of genes, or DNA synthesized from such RNA; and
(2) a first computer subsystem programmed for (a) receiving gene expression data on at least 4 test genes selected from the panel of genes, (b) weighting the determined expression of each of the test genes, and (c) combining the weighted expression to provide a test value, wherein the combined weight given to said at least 4 cell-cycle genes is at least 40% of the total weight given to the expression of all of said plurality of test genes; and
(3) a second computer subsystem programmed for comparing the test value to one or more reference values each associated with a predetermined degree of risk of cancer recurrence or progression of the lung cancer or colon cancer.
25. The system of Claim 23 or Claim 24, further comprising a display module displaying the comparison between the test value to the one or more reference values, or displaying a result of the comparing step.
26. A method of prognosing cancer selected from prostate cancer, lung cancer, bladder cancer or brain cancer, comprising:
determining in a tumor sample from a patient diagnosed of prostate cancer, lung cancer, bladder cancer or brain cancer, the expression of at least 4 cell-cycle genes;
correlating overexpression of said at least 4 cell-cycle genes to a poor prognosis or an increased likelihood of recurrence of cancer in the patient.
PCT/US2011/043228 2010-07-07 2011-07-07 Gene signatures for cancer prognosis WO2012006447A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA2804391A CA2804391A1 (en) 2010-07-07 2011-07-07 Gene signatures for cancer prognosis
EP11804357.9A EP2591126B1 (en) 2010-07-07 2011-07-07 Gene signatures for cancer prognosis
EP20210783.5A EP3812469A1 (en) 2010-07-07 2011-07-07 Gene signatures for cancer prognosis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US36220910P 2010-07-07 2010-07-07
US61/362,209 2010-07-07

Publications (2)

Publication Number Publication Date
WO2012006447A2 true WO2012006447A2 (en) 2012-01-12
WO2012006447A3 WO2012006447A3 (en) 2012-05-18

Family

ID=45441805

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/043228 WO2012006447A2 (en) 2010-07-07 2011-07-07 Gene signatures for cancer prognosis

Country Status (4)

Country Link
US (5) US20120053253A1 (en)
EP (2) EP3812469A1 (en)
CA (1) CA2804391A1 (en)
WO (1) WO2012006447A2 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2611941A2 (en) * 2010-08-30 2013-07-10 Myriad Genetics, Inc. Gene signatures for cancer diagnosis and prognosis
WO2013104921A1 (en) * 2012-01-13 2013-07-18 Isis Innovation Limited Biomarker and uses thereof
EP2691547A1 (en) * 2011-03-26 2014-02-05 Oregon Health & Science University Gene expression predictors of cancer prognosis
WO2014078700A1 (en) * 2012-11-16 2014-05-22 Myriad Genetics, Inc. Gene signatures for cancer prognosis
WO2015042570A1 (en) 2013-09-23 2015-03-26 The University Of Chicago Methods and compositions relating to cancer therapy with dna damaging agents
EP2744919A4 (en) * 2011-08-19 2015-04-08 Myriad Genetics Inc Gene signatures for lung cancer prognosis and therapy selection
WO2015175692A1 (en) 2014-05-13 2015-11-19 Myriad Genetics, Inc. Gene signatures for cancer prognosis
WO2017120456A1 (en) 2016-01-06 2017-07-13 Myriad Genetics, Inc. Genes and gene signatures for diagnosis and treatment of melanoma
US9790556B2 (en) 2012-01-05 2017-10-17 Centre National De La Recherche Scientifique (Cnrs) Signature for the diagnosis of lung cancer aggressiveness and genetic instability
WO2017193062A1 (en) 2016-05-06 2017-11-09 Myriad Genetics, Inc. Gene signatures for renal cancer prognosis
EP3134548A4 (en) * 2014-04-23 2018-02-28 Myriad Genetics, Inc. Cancer prognosis signatures
US9976188B2 (en) 2009-01-07 2018-05-22 Myriad Genetics, Inc. Cancer biomarkers
US10954568B2 (en) 2010-07-07 2021-03-23 Myriad Genetics, Inc. Gene signatures for cancer prognosis
EP3798316A2 (en) 2013-03-15 2021-03-31 Myriad Genetics, Inc. Genes and gene signatures for diagnosis and treatment of melanoma
EP4250299A2 (en) 2014-07-02 2023-09-27 Myriad myPath, LLC Genes and gene signatures for diagnosis and treatment of melanoma

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013192616A1 (en) 2012-06-22 2013-12-27 Htg Molecular Diagnostics, Inc. Molecular malignancy in melanocytic lesions
GB201322034D0 (en) 2013-12-12 2014-01-29 Almac Diagnostics Ltd Prostate cancer classification
US20170058354A1 (en) * 2014-02-19 2017-03-02 The Trustees Of Columbia University In The City Of New York Method and composition for diagnosis of aggressive prostate cancer
GB201616912D0 (en) 2016-10-05 2016-11-16 University Of East Anglia Classification of cancer
CN112888459B (en) * 2018-06-01 2023-05-23 格里尔公司 Convolutional neural network system and data classification method
US20210395832A1 (en) 2018-11-13 2021-12-23 Bracco Imaging S.P.A. Gene signatures for the prediction of prostate cancer recurrence
US11581062B2 (en) 2018-12-10 2023-02-14 Grail, Llc Systems and methods for classifying patients with respect to multiple cancer classes
EP3901288A1 (en) * 2020-04-20 2021-10-27 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Multi-gene expression assay for prostate carcinoma

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5593839A (en) 1994-05-24 1997-01-14 Affymetrix, Inc. Computer-aided engineering system for design of sequence arrays and lithographic masks
US5733729A (en) 1995-09-14 1998-03-31 Affymetrix, Inc. Computer-aided probability base calling for arrays of nucleic acid probes on chips
US5795716A (en) 1994-10-21 1998-08-18 Chee; Mark S. Computer-aided visualization and analysis system for sequence evaluation
US6090555A (en) 1997-12-11 2000-07-18 Affymetrix, Inc. Scanned image alignment systems and methods
US6185561B1 (en) 1998-09-17 2001-02-06 Affymetrix, Inc. Method and apparatus for providing and expression data mining database
US6188783B1 (en) 1997-07-25 2001-02-13 Affymetrix, Inc. Method and system for providing a probe array chip design database
US6223127B1 (en) 1997-08-15 2001-04-24 Affymetrix, Inc. Polymorphism detection utilizing clustering analysis
US6355906B1 (en) 1998-07-07 2002-03-12 Nissan Motor Co., Ltd. Production system using combination jigs and jig replacement method and apparatus therefor
US6420108B2 (en) 1998-02-09 2002-07-16 Affymetrix, Inc. Computer-aided display for comparative gene expression
US20020183936A1 (en) 2001-01-24 2002-12-05 Affymetrix, Inc. Method, system, and computer software for providing a genomic web portal
US20030097222A1 (en) 2000-01-25 2003-05-22 Craford David M. Method, system, and computer software for providing a genomic web portal
US20030100995A1 (en) 2001-07-16 2003-05-29 Affymetrix, Inc. Method, system and computer software for variant information via a web portal
US20030120432A1 (en) 2001-01-29 2003-06-26 Affymetrix, Inc. Method, system and computer software for online ordering of custom probe arrays
US6586806B1 (en) 1997-06-20 2003-07-01 Cypress Semiconductor Corporation Method and structure for a single-sided non-self-aligned transistor
US6585606B2 (en) 2001-07-16 2003-07-01 Thomas S. Penrose Golf club accessory
US20040049354A1 (en) 2002-04-26 2004-03-11 Affymetrix, Inc. Method, system and computer software providing a genomic web portal for functional analysis of alternative splice variants

Family Cites Families (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5993388A (en) 1997-07-01 1999-11-30 Kattan; Michael W. Nomograms to aid in the treatment of prostatic cancer
US6409664B1 (en) 1997-07-01 2002-06-25 Michael W. Kattan Nomograms to aid in the treatment of prostatic cancer
WO2000040749A2 (en) * 1999-01-06 2000-07-13 Genenews Inc. Method for the detection of gene transcripts in blood and uses thereof
EP2258872B1 (en) 2002-03-13 2013-08-14 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
US20030235816A1 (en) 2002-03-14 2003-12-25 Baylor College Of Medicine (By Slawin And Shariat) Method to determine outcome for patients with prostatic disease
US6809118B2 (en) 2002-07-25 2004-10-26 Yih-Lin Chung Methods for therapy of radiation cutaneous syndrome
EP1422526A1 (en) * 2002-10-28 2004-05-26 MTM Laboratories AG Method for improved diagnosis of dysplasias
US20040231909A1 (en) 2003-01-15 2004-11-25 Tai-Yang Luh Motorized vehicle having forward and backward differential structure
PT1597391E (en) 2003-02-20 2008-12-19 Genomic Health Inc Use of intronic rna to measure gene expression
WO2004111603A2 (en) 2003-05-28 2004-12-23 Genomic Health, Inc. Gene expression markers for predicting response to chemotherapy
CA3061769C (en) 2003-06-24 2021-10-26 Genomic Health, Inc. Methods of predicting the likelihood of long-term survival of a human patient with node-negative, estrogen receptor (er) positive, invasive ductal breast cancer without the recurrence of breast cancer
US20070048738A1 (en) 2003-07-14 2007-03-01 Mayo Foundation For Medical Education And Research Methods and compositions for diagnosis, staging and prognosis of prostate cancer
WO2005059109A2 (en) * 2003-12-15 2005-06-30 The Regents Of The University Of California Molecular signature of the pten tumor suppressor
EP1737980A2 (en) 2004-04-09 2007-01-03 Fondazione IRCCS Istituto Nazionale dei Tumori Gene expression markers for predicting response to chemotherapy
WO2005106043A2 (en) 2004-04-23 2005-11-10 Exagen Diagnostics, Inc. Breast cancer gene expression biomarkers
DE602005024021D1 (en) * 2004-08-10 2010-11-18 Oncotherapy Science Inc GEN ANLN IN COMBINATION WITH NON-SMALL CELL LUNG CANCER AND ITS INTERACTION WITH RhoA
DK1815014T3 (en) 2004-11-05 2012-06-18 Genomic Health Inc Molecular indicators of breast cancer prognosis and prediction of treatment response
JP5020088B2 (en) 2004-11-05 2012-09-05 ジェノミック ヘルス, インコーポレイテッド Predicting response to chemotherapy using gene expression markers
US20070059720A9 (en) 2004-12-06 2007-03-15 Suzanne Fuqua RNA expression profile predicting response to tamoxifen in breast cancer patients
US7666595B2 (en) 2005-02-25 2010-02-23 The Brigham And Women's Hospital, Inc. Biomarkers for predicting prostate cancer progression
US20070026424A1 (en) 2005-04-15 2007-02-01 Powell Charles A Gene profiles correlating with histology and prognosis
WO2006119593A1 (en) 2005-05-13 2006-11-16 Universite Libre De Bruxelles Gene-based algorithmic cancer prognosis
US20080275652A1 (en) 2005-05-13 2008-11-06 Universite Libre De Bruxelles Gene-based algorithmic cancer prognosis
US20070099209A1 (en) 2005-06-13 2007-05-03 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US20100152055A1 (en) 2005-09-02 2010-06-17 Satoko Kozono Composition and method for diagnosing kidney cancer and for predicting prognosis for kidney cancer patient
GB0519405D0 (en) * 2005-09-23 2005-11-02 Univ Aberdeen Cancer therapy prognosis and target
WO2007067500A2 (en) 2005-12-05 2007-06-14 Genomic Health, Inc. Predictors of patient response to treatment with egfr inhibitors
WO2007067672A2 (en) 2005-12-06 2007-06-14 Baylor College Of Medicine Method to predict systemic progression in prostate cancer patients
US20090215054A1 (en) 2005-12-13 2009-08-27 Carter Scott L Prognosis indicators for solid human tumors
BRPI0706511A2 (en) 2006-01-11 2011-03-29 Genomic Health Inc gene expression markers for colorectal cancer prognosis
US7914988B1 (en) 2006-03-31 2011-03-29 Illumina, Inc. Gene expression profiles to predict relapse of prostate cancer
WO2008058018A2 (en) 2006-11-02 2008-05-15 Mayo Foundation For Medical Education And Research Predicting cancer outcome
AT504702A1 (en) 2006-12-22 2008-07-15 Arc Austrian Res Centers Gmbh SET OF TUMOR MARKERS
CA2939539A1 (en) 2007-02-23 2008-08-28 General Hospital Corporation Prostate cancer survival and recurrence
WO2008104805A2 (en) 2007-02-26 2008-09-04 Oxford Genome Sciences (Uk) Limited Proteins
EP2140020A2 (en) 2007-03-15 2010-01-06 Genomic Health, Inc. Gene expression markers for prediction of patient response to chemotherapy
ES2304306B1 (en) * 2007-03-20 2009-07-07 Indas Biotech, S.L.U. METHOD OF DIAGNOSIS AND / OR FORECAST OF CANCER VESICAL.
CA2688477C (en) 2007-06-01 2020-12-22 The Regents Of The University Of California Multigene prognostic assay for lung cancer
US20100247528A1 (en) 2007-09-06 2010-09-30 Kent Hunter Arrays, kits and cancer characterization methods
WO2009036427A2 (en) 2007-09-14 2009-03-19 Ventana Medical Systems, Inc. Prostate cancer biomarkers
NZ562237A (en) * 2007-10-05 2011-02-25 Pacific Edge Biotechnology Ltd Proliferation signature and prognosis for gastrointestinal cancer
US8067178B2 (en) 2008-03-14 2011-11-29 Genomic Health, Inc. Gene expression markers for prediction of patient response to chemotherapy
DK2294215T3 (en) 2008-05-12 2013-04-22 Genomic Health Inc Tests to predict cancer patients' response to various chemotherapeutic treatment options
US20100184063A1 (en) 2008-05-14 2010-07-22 Ming-Sound Tsao Prognostic and predictive gene signature for non-small cell lung cancer and adjuvant chemotherapy
WO2010003773A1 (en) 2008-06-16 2010-01-14 Siemens Medical Solutions Diagnostics Gmbh Algorithms for outcome prediction in patients with node-positive chemotherapy-treated breast cancer
US20120053842A9 (en) 2008-06-16 2012-03-01 Sividon Diagnostics Molecular markers for cancer prognosis
AU2009262022A1 (en) 2008-06-26 2009-12-30 Dana-Farber Cancer Institute, Inc. Signatures and determinants associated with metastasis and methods of use thereof
CN102272324A (en) 2008-10-01 2011-12-07 诺维奥根迪克斯研究有限公司 Molecular markers in prostate cancer
US20120041274A1 (en) 2010-01-07 2012-02-16 Myriad Genetics, Incorporated Cancer biomarkers
CA2749103A1 (en) * 2009-01-07 2010-07-15 Steve Stone Cancer biomarkers
US20120190565A1 (en) 2009-02-20 2012-07-26 Pangea Biosciences, Inc. Method Of Diagnosis Or Prognosis Of A Neoplasm Comprising Determining The Level Of Expression Of A Protein In Stromal Cells Adjacent To The Neoplasm
US20120039805A1 (en) 2009-02-20 2012-02-16 Pangea Biosciences, Inc. Therapeutics And Methods For Treating Neoplastic Diseases Comprising Determining The Level Of Caveolin-1 And/Or Caveolin-2 In A Stromal Cell Sample
EP2241634A1 (en) 2009-04-16 2010-10-20 Université Libre de Bruxelles Diagnostic method and tools to predict the effiacy of targeted agents against IGF-1 pathway activation in cancer
WO2010127322A1 (en) 2009-05-01 2010-11-04 Genomic Health Inc. Gene expression profile algorithm and test for likelihood of recurrence of colorectal cancer and response to chemotherapy
DK2504451T3 (en) 2009-11-23 2019-08-05 Genomic Health Inc Methods for predicting the clinical course of cancer
EP2531619A4 (en) 2010-02-05 2013-08-21 Myriad Genetics Inc Hypoxia-related gene signatures for cancer classification
CA2804391A1 (en) 2010-07-07 2012-01-12 Myriad Genetics, Inc. Gene signatures for cancer prognosis
EP2409989A1 (en) 2010-07-19 2012-01-25 International-Drug-Development-Biotech Method to improve glycosylation profile for antibody
WO2012012225A2 (en) 2010-07-22 2012-01-26 Mayo Foundation For Medical Education And Research Predicting cancer outcome
EP2913405B1 (en) 2010-07-27 2016-11-09 Genomic Health, Inc. Method for using gene expression to determine prognosis of prostate cancer
EP2611941A4 (en) 2010-08-30 2014-01-22 Myriad Genetics Inc Gene signatures for cancer diagnosis and prognosis
EP2671076A4 (en) 2011-02-04 2016-11-16 Bioarray Genetics Inc Methods of using gene expression signatures to select a method of treatment, predict prognosis, survival, and/or predict response to treatment
US20140011701A1 (en) 2011-03-14 2014-01-09 National Research Council Of Canada Prognostic Marker Sets For Prostate Cancer
CA2831074A1 (en) 2011-03-26 2012-10-04 Oregon Health And Science University Gene expression predictors of cancer prognosis
US20140073535A1 (en) 2011-05-12 2014-03-13 Noviogendix Research B.V. Molecular markers in prostate cancer
CA2835728A1 (en) 2011-05-12 2012-11-15 Noviogendix Research B.V. Molecular markers in prostate cancer
WO2013028554A2 (en) 2011-08-19 2013-02-28 Myriad Genetics, Inc. Gene signatures for lung cancer prognosis and therapy selection
EP4190918A1 (en) 2012-11-16 2023-06-07 Myriad Genetics, Inc. Gene signatures for cancer prognosis

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5593839A (en) 1994-05-24 1997-01-14 Affymetrix, Inc. Computer-aided engineering system for design of sequence arrays and lithographic masks
US5795716A (en) 1994-10-21 1998-08-18 Chee; Mark S. Computer-aided visualization and analysis system for sequence evaluation
US5974164A (en) 1994-10-21 1999-10-26 Affymetrix, Inc. Computer-aided visualization and analysis system for sequence evaluation
US5733729A (en) 1995-09-14 1998-03-31 Affymetrix, Inc. Computer-aided probability base calling for arrays of nucleic acid probes on chips
US6066454A (en) 1995-09-14 2000-05-23 Affymetrix, Inc. Computer-aided probability base calling for arrays of nucleic acid probes on chips
US6586806B1 (en) 1997-06-20 2003-07-01 Cypress Semiconductor Corporation Method and structure for a single-sided non-self-aligned transistor
US6308170B1 (en) 1997-07-25 2001-10-23 Affymetrix Inc. Gene expression and evaluation system
US6188783B1 (en) 1997-07-25 2001-02-13 Affymetrix, Inc. Method and system for providing a probe array chip design database
US6229911B1 (en) 1997-07-25 2001-05-08 Affymetrix, Inc. Method and apparatus for providing a bioinformatics database
US6223127B1 (en) 1997-08-15 2001-04-24 Affymetrix, Inc. Polymorphism detection utilizing clustering analysis
US6090555A (en) 1997-12-11 2000-07-18 Affymetrix, Inc. Scanned image alignment systems and methods
US6420108B2 (en) 1998-02-09 2002-07-16 Affymetrix, Inc. Computer-aided display for comparative gene expression
US6355906B1 (en) 1998-07-07 2002-03-12 Nissan Motor Co., Ltd. Production system using combination jigs and jig replacement method and apparatus therefor
US6185561B1 (en) 1998-09-17 2001-02-06 Affymetrix, Inc. Method and apparatus for providing and expression data mining database
US20030097222A1 (en) 2000-01-25 2003-05-22 Craford David M. Method, system, and computer software for providing a genomic web portal
US20020183936A1 (en) 2001-01-24 2002-12-05 Affymetrix, Inc. Method, system, and computer software for providing a genomic web portal
US20030120432A1 (en) 2001-01-29 2003-06-26 Affymetrix, Inc. Method, system and computer software for online ordering of custom probe arrays
US20030100995A1 (en) 2001-07-16 2003-05-29 Affymetrix, Inc. Method, system and computer software for variant information via a web portal
US6585606B2 (en) 2001-07-16 2003-07-01 Thomas S. Penrose Golf club accessory
US20040049354A1 (en) 2002-04-26 2004-03-11 Affymetrix, Inc. Method, system and computer software providing a genomic web portal for functional analysis of alternative splice variants

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"COMPUTATIONAL METHODS IN MOLECULAR BIOLOGY", 1998, ELSEVIER
DESMEDT ET AL., CLIN. CANCER RES., vol. 13, 2007, pages 4407 - 14
FRANK ET AL., J. CLIN. ONCOL., vol. 20, 2002, pages 1480 - 1490
FREIJE ET AL., CANCER RES., vol. 64, 2004, pages 6503 - 10
GALEA ET AL., BREAST CANCER RES. & TREAT., vol. 22, 1992, pages 207 - 19
JABLONSKI ET AL., NUCLEIC ACIDS RES., vol. 14, 1986, pages 6115 - 6128
KATTAN ET AL., J. CLIN. ONCOL., vol. 17, 1999, pages 1499 - 1507
LIU JIANGANG ET AL., BMC MEDICAL GENOMICS, vol. 1, 2008, pages 11
NGUYEN ET AL., BIOTECHNIQUES, vol. 13, 1992, pages 116 - 123
OUELETTEBZEVANIS: "BIOINFORMATICS: A PRACTICAL GUIDE FOR ANALYSIS OF GENE AND PROTEINS", 2001, WILEY & SONS, INC.
PHILLIPS ET AL., CANCER CELL, vol. 9, 2006, pages 157 - 73
QUINN ET AL., EUROPEAN JOURNAL OF CANCER, vol. 41, 2005, pages 858 - 887
RASHIDIBUEHLER: "BIOINFORMATICS BASICS: APPLICATION IN BIOLOGICAL SCIENCE AND MEDICINE", 2000, CRC PRESS
RIGBY ET AL., J. MOL. BIOL., vol. 113, 1977, pages 237 - 251
SCHMIDT ET AL., CANCER RES., vol. 68, 2008, pages 5405 - 13
See also references of EP2591126A4
SETUBAL ET AL.: "INTRODUCTION TO COMPUTATIONAL BIOLOGY METHODS", 1997, PWS PUBLISHING COMPANY
SHEDDEN ET AL., NATURE MED., vol. 14, 2008, pages 822
STEPHENSON ET AL., J. CLIN. ONCOL., vol. 23, 2005, pages 7005 - 7012
SWANSON ET AL., UROL ONCOL., vol. 25, 2007, pages 110 - 114
SWANSON ET AL., UROL. ONCOL., vol. 25, 2007, pages 110 - 114
WANG LIANG ET AL., CANCER RESEARCH, vol. 69, 2009, pages 9490 - 9497
WHITFIELD ET AL., MOL. BIOL. CELL, vol. 13, 2002, pages 1977 - 2000
WHITFIELD ET AL., MOL. CELL. BIOL., vol. 20, 2000, pages 4188 - 4198

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9976188B2 (en) 2009-01-07 2018-05-22 Myriad Genetics, Inc. Cancer biomarkers
US10519513B2 (en) 2009-01-07 2019-12-31 Myriad Genetics, Inc. Cancer Biomarkers
US10954568B2 (en) 2010-07-07 2021-03-23 Myriad Genetics, Inc. Gene signatures for cancer prognosis
EP2611941A4 (en) * 2010-08-30 2014-01-22 Myriad Genetics Inc Gene signatures for cancer diagnosis and prognosis
EP2611941A2 (en) * 2010-08-30 2013-07-10 Myriad Genetics, Inc. Gene signatures for cancer diagnosis and prognosis
US9605319B2 (en) 2010-08-30 2017-03-28 Myriad Genetics, Inc. Gene signatures for cancer diagnosis and prognosis
EP2691547A1 (en) * 2011-03-26 2014-02-05 Oregon Health & Science University Gene expression predictors of cancer prognosis
EP2691547A4 (en) * 2011-03-26 2015-01-14 Univ Oregon Health & Science Gene expression predictors of cancer prognosis
EP2744919A4 (en) * 2011-08-19 2015-04-08 Myriad Genetics Inc Gene signatures for lung cancer prognosis and therapy selection
US9790556B2 (en) 2012-01-05 2017-10-17 Centre National De La Recherche Scientifique (Cnrs) Signature for the diagnosis of lung cancer aggressiveness and genetic instability
WO2013104921A1 (en) * 2012-01-13 2013-07-18 Isis Innovation Limited Biomarker and uses thereof
EP4190918A1 (en) 2012-11-16 2023-06-07 Myriad Genetics, Inc. Gene signatures for cancer prognosis
WO2014078700A1 (en) * 2012-11-16 2014-05-22 Myriad Genetics, Inc. Gene signatures for cancer prognosis
US10876164B2 (en) 2012-11-16 2020-12-29 Myriad Genetics, Inc. Gene signatures for cancer prognosis
EP3798316A2 (en) 2013-03-15 2021-03-31 Myriad Genetics, Inc. Genes and gene signatures for diagnosis and treatment of melanoma
EP4043580A1 (en) 2013-03-15 2022-08-17 Myriad myPath, LLC Genes and gene signatures for diagnosis and treatment of melanoma
WO2015042570A1 (en) 2013-09-23 2015-03-26 The University Of Chicago Methods and compositions relating to cancer therapy with dna damaging agents
EP3049536B1 (en) * 2013-09-23 2019-05-22 The University of Chicago Methods and compositions relating to cancer therapy with dna damaging agents
EP3134548A4 (en) * 2014-04-23 2018-02-28 Myriad Genetics, Inc. Cancer prognosis signatures
EP3623482A1 (en) 2014-05-13 2020-03-18 Myriad Genetics, Inc. Gene signatures for cancer prognosis
US11174517B2 (en) 2014-05-13 2021-11-16 Myriad Genetics, Inc. Gene signatures for cancer prognosis
WO2015175692A1 (en) 2014-05-13 2015-11-19 Myriad Genetics, Inc. Gene signatures for cancer prognosis
EP4250299A2 (en) 2014-07-02 2023-09-27 Myriad myPath, LLC Genes and gene signatures for diagnosis and treatment of melanoma
WO2017120456A1 (en) 2016-01-06 2017-07-13 Myriad Genetics, Inc. Genes and gene signatures for diagnosis and treatment of melanoma
WO2017193062A1 (en) 2016-05-06 2017-11-09 Myriad Genetics, Inc. Gene signatures for renal cancer prognosis

Also Published As

Publication number Publication date
EP2591126A4 (en) 2014-01-01
EP2591126A2 (en) 2013-05-15
CA2804391A1 (en) 2012-01-12
EP2591126B1 (en) 2020-12-30
US20220259675A1 (en) 2022-08-18
US20150167103A1 (en) 2015-06-18
US20180334722A1 (en) 2018-11-22
US20120053253A1 (en) 2012-03-01
WO2012006447A3 (en) 2012-05-18
US10954568B2 (en) 2021-03-23
US20160355884A1 (en) 2016-12-08
EP3812469A1 (en) 2021-04-28

Similar Documents

Publication Publication Date Title
US20220259675A1 (en) Gene signatures for cancer prognosis
US10519513B2 (en) Cancer Biomarkers
AU2019210509B2 (en) Cancer biomarkers
US9605319B2 (en) Gene signatures for cancer diagnosis and prognosis
US20210071269A1 (en) Gene signatures for cancer prognosis
US20220112565A1 (en) Gene signatures for cancer prognosis

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11804357

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2804391

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: 2011804357

Country of ref document: EP