US20180312926A9 - Biomarker panels for predicting prostate cancer outcomes - Google Patents

Biomarker panels for predicting prostate cancer outcomes Download PDF

Info

Publication number
US20180312926A9
US20180312926A9 US14/857,658 US201514857658A US2018312926A9 US 20180312926 A9 US20180312926 A9 US 20180312926A9 US 201514857658 A US201514857658 A US 201514857658A US 2018312926 A9 US2018312926 A9 US 2018312926A9
Authority
US
United States
Prior art keywords
psa
later
systemic disease
prostate cancer
score
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
US14/857,658
Other versions
US10407731B2 (en
US20160010162A1 (en
Inventor
George G. Klee
Robert B. Jenkins
Thomas M. Kollmeyer
Karla V. Ballman
Eric J. Bergstralh
Bruce W. Morlan
S. Keith Anderson
Tohru Nakagawa
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Mayo Foundation for Medical Education and Research
Original Assignee
Mayo Foundation for Medical Education and Research
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 Mayo Foundation for Medical Education and Research filed Critical Mayo Foundation for Medical Education and Research
Priority to US14/857,658 priority Critical patent/US10407731B2/en
Publication of US20160010162A1 publication Critical patent/US20160010162A1/en
Publication of US20180312926A9 publication Critical patent/US20180312926A9/en
Assigned to CRG SERVICING LLC reassignment CRG SERVICING LLC SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DECIPHER BIOSCIENCES, INC.
Application granted granted Critical
Publication of US10407731B2 publication Critical patent/US10407731B2/en
Assigned to MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH reassignment MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NAKAGAWA, TOHRU, ANDERSON, S. KEITH, BALLMAN, KARLA V., BERGSTRALH, ERIC J., JENKINS, ROBERT B., KLEE, GEORGE G., KOLLMEYER, THOMAS M., MORLAN, BRUCE W.
Assigned to DECIPHER BIOSCIENCES, INC. reassignment DECIPHER BIOSCIENCES, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: CRG SERVICING LLC
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

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
    • G06F19/3431
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/20ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for computer-aided diagnosis, e.g. based on medical expert systems
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/30ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for calculating health indices; for individual health risk assessment
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16ZINFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS, NOT OTHERWISE PROVIDED FOR
    • G16Z99/00Subject matter not provided for in other main groups of this subclass
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • 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
    • C12Q2600/16Primer sets for multiplex assays

Definitions

  • This document relates to methods and materials involved in predicting the outcome of prostate cancer.
  • Prostate cancer occurs when a malignant tumor forms in the tissue of the prostate.
  • the prostate is a gland in the male reproductive system located below the bladder and in front of the rectum.
  • the main function of the prostate gland which is about the size of a walnut, is to make fluid for semen.
  • This type of cancer is known as adenocarcinoma.
  • Prostate cancer is the second leading cause of cancer-related death in American men. Most of the time, prostate cancer grows slowly. Autopsy studies show that many older men who died of other diseases also had prostate cancer that neither they nor their doctor were aware of. Sometimes, however, prostate cancer can grow and spread quickly. It is important to be able to distinguish prostate cancers that will grow slowly from those that will grow quickly since treatment can be especially effective when the cancer has not spread beyond the region of the prostate. Finding ways to detect cancers early can improve survival rates.
  • This document provides methods and materials related to assessing male mammals (e.g., humans) with prostate cancer. For example, this document provides methods and materials for predicting (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer.
  • one aspect of this document features a method for predicting whether or not a human, at the time of PSA reoccurrence or retropubic radial prostatectomy, will later develop systemic disease.
  • the method comprises, or consists essentially of, (a) determining an expression profile score for cancer tissue from the human, wherein the expression profile score is based on at least the expression levels of RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 nucleic acid, and (b) prognosing the human as later developing systemic disease or as not later developing systemic disease based on at least the expression profile score.
  • the method can be performed at the time of the PSA reoccurrence.
  • the method can be performed at the time of the retropubic radial prostatectomy.
  • the expression levels can be mRNA expression levels.
  • the prognosing step (b) can comprise prognosing the human as later developing systemic disease or as not later developing systemic disease based on at least the expression profile score and a clinical variable.
  • the clinical variable can be selected from the group consisting of a Gleason score and a revised Gleason score.
  • the clinical variable can be selected from the group consisting of a Gleason score, a revised Gleason score, the pStage, age at surgery, initial PSA at recurrence, use of hormone or radiation therapy after radical retropubic prostatectomy, age at PSA recurrence, the second PSA level at time of PSA recurrence, and PSA slope.
  • the method can comprise prognosing the human as later developing systemic disease based on at least the expression profile score.
  • the method can comprise prognosing the human as not later developing systemic disease based on at least the expression profile score.
  • this document features a method for predicting whether or not a human, at the time of systemic disease, will later die from prostate cancer.
  • the method comprises, or consists essentially of, (a) determining an expression profile score for cancer tissue from the human, wherein the expression profile score is based on at least the expression levels of RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 nucleic acid, and (b) prognosing the human as later dying of the prostate cancer or as not later dying of the prostate cancer based on at least the expression profile score.
  • the expression levels can be mRNA expression levels.
  • the prognosing step (b) can comprise prognosing the human as later developing systemic disease or as not later developing systemic disease based on at least the expression profile score and a clinical variable.
  • the clinical variable can be selected from the group consisting of a Gleason score and a revised Gleason score.
  • the clinical variable can be selected from the group consisting of a Gleason score, a revised Gleason score, the pStage, age at surgery, initial PSA at recurrence, use of hormone or radiation therapy after radical retropubic prostatectomy, age at PSA recurrence, the second PSA level at time of PSA recurrence, and PSA slope.
  • the method can comprise prognosing the human as later dying of the prostate cancer based on at least the expression profile score.
  • the method can comprise prognosing the human as not later dying of the prostate cancer based on at least the expression profile score.
  • this document features a method for (1) predicting whether or not a patient, at the time of PSA reoccurrence, will later develop systemic disease, (2) predicting whether or not a patient, at the time of retropubic radial prostatectomy, will later develop systemic disease, or (3) predicting whether or not a patient, at the time of systemic disease, will later die from prostate cancer.
  • the method comprises, or consists essentially of, determining whether or not cancer tissue from the patient contains an RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 expression profile indicative of a later development of the systemic disease or the death.
  • FIG. 1 Nine genes with significantly different expression in cases with systemic disease progression (SYS) versus controls with PSA recurrence (PSA). P-values (t-test) for the SYS case/PSA control comparison are shown. Controls with no evidence of disease recurrence (NED) are also included.
  • SYS systemic disease progression
  • PSA PSA recurrence
  • FIG. 2 (A to D) Areas under the curve (AUCs) for three clinical models, the final 17 gene/probe model and the combined clinical probe models.
  • A. The training set AUCs for three clinical models, the final 17 gene/probe model and the combined clinical/17 gene/probe model.
  • B. The validation set AUCs for three clinical models, the final 17 gene/probe model and the combined clinical/17 gene/probe model.
  • C. The training set AUCs of four previously reported gene expression models of prostate cancer aggressiveness compared with the Clinical model C alone and with the 17 gene/probe model.
  • D The validation set AUCs of four previously reported gene expression models of prostate cancer aggressiveness compared with the clinical model C alone and with the 17 gene/probe model.
  • E and F A comparison of the training and validation set AUCs for each of the model.
  • E. AUCs of the each of the gene/probe models alone.
  • F. AUCs of each of the gene/probe models with the inclusion of clinical model C.
  • FIG. 3 Systemic progression-free and overall prostate cancer-specific survival in the PSA Control and SYS Case groups.
  • FIG. 4 Expression results for ERG, ETV1 and ETV4 among the men with no evidence of disease progression (NED), PSA recurrence (PSA) and systemic progression (SYS).
  • NED disease progression
  • PSA PSA recurrence
  • SYS systemic progression
  • FIG. 5 is a summary of the nested case-control study design.
  • FIG. 6 Reproducibility of DASL assay and the effect of RNA quantity on the DASL assay.
  • A An example of DASL interplate reproducibility.
  • B Effect of reduced RNA quantity on the DASL assay.
  • F Summary of quantitative RT-PCR and DASL data comparisons.
  • FIG. 8 Comparison of genes having multiple probe sets on the Cancer Panel v1 and/or the Custom panel.
  • A Comparison of three probe sets (Cancer Panel ERG, Custom Panel ERG and Custom panel ERG splice variant) for ERG.
  • B Comparison of two probe sets (Custom Panel SRD5A2 and Custom panel terparbo) for SRD5A2/terparbo.
  • This document provides methods and materials related to assessing male mammals (e.g., humans) with prostate cancer. For example, this document provides methods and materials for predicting (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer.
  • the expression level of any of the genes listed in the tables provided herein can be assessed as described herein to predict (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer.
  • the combination of genes set forth in Table 3 can be assessed as described herein to predict (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer.
  • any appropriate type of sample e.g., cancer tissue
  • prostate cancer tissue can be collected and assessed to determine the expression level of a gene listed in any of the tables provided herein.
  • the expression level for a particular nucleic acid can be used as a raw number or can be normalized using appropriate calculations and controls.
  • the expression levels for groups of nucleic acids can be combined to obtain an expression level score that is based on the measured expression levels (e.g., raw expression level number or normalized number). In some cases, the expression levels of the individual nucleic acids that are used to obtain an expression level score can be weighted.
  • An expression level score can be a whole number, an integer, an alphanumerical value, or any other representation capable of indicating whether or not a condition is met.
  • an expression level score is a number that is based on the mRNA expression levels of at least the seventeen nucleic acids listed in Table 3.
  • an expression level score can be based on the mRNA expression levels of the seventeen nucleic acids listed in Table 3 and no other nucleic acids.
  • the seventeen nucleic acids listed in Table 3 can be used together to determine, at the time of PSA reoccurrence or at the time of retropubic radial prostatectomy, whether or not a mammal will later develop systemic disease.
  • the seventeen nucleic acids listed in Table 3 can be used together to determine, at the time of systemic disease, whether or not a mammal will later die of prostate cancer.
  • the seventeen nucleic acids listed in Table 3 can have the nucleic acid sequence set forth in GenBank as follows: RAD21 (GenBank Accession No. NM_006265; GI No. 208879448; probe sequences GGGATAAGAAGCTAACCAAAG-CCCATGTGTTCGAGTGTAATTTAGAGAG (SEQ ID NO:1), GAGGAAAATCGGG-AAGCAGCTTATAATGCCATTACTTTACCTGAAG (SEQ ID NO:2), and TGATT-TTGGAATGGATGATCGTGAGATAATGAGAGAAGGCAGTGCTT (SEQ ID NO:3)), CDKN3 (GenBank Accession Nos. NM_005192 and NM_001130851; GI Nos.
  • probe sequences TGCAGCTGGTTGGTGTCACTGCCATGTTTATTGCAAGCAAATAT (SEQ ID NO:7), AACAAGTATGCCACATCGAAGCATGCTAAGATCAGCACTCTACCAC-AG (SEQ ID NO:8), and TTTAGCCAAGGCTGTGGCAAAGGTGTAACTT-GTAAACTTGAGTTGGA (SEQ ID NO:9)), SEC14L1 (GenBank Accession Nos. NM_001039573, NM_001143998, NM_001143999, NM_001144001, and NM_003003; GI Nos.
  • probe sequences CAGACTCCCTC-ATCACCAAAAAGCAAGTGTCAGTCTGGTGCAGTAAT (SEQ ID NO:16), CAG-GCCTTTCTGCAGAAAGCAGGCAAATCTCTGTTGTTCTATGCC (SEQ ID NO:17), and TTCCAGGACATCATGCAAAAGCAAAGACCAGAAAGAGTGTCTCATC (SEQ ID NO:18)), KIAA0196 (GenBank Accession No. NM_014846; GI No.
  • probe sequences AATGCCATCATTGCTGAACTTTTGAGACTCTCTGAGTTTATT-CCTGCT SEQ ID NO:19
  • TGGGAAAGCAAACTGGATGCTAAGCCAGAGC-TACAGGATTTAGATGAA SEQ ID NO:20
  • CAACCAGGTGCCAAAAG-ACCATCCAACTATCCCGAGAGCTATTTC SEQ ID NO:21
  • TAF2 GenBank Accession No. NM_003184; GI No.
  • RT-PCR reverse transcription-PCR
  • the term “elevated level” as used herein with respect to the level of mRNA for a nucleic acid listed herein is any mRNA level that is greater than a reference mRNA level for that nucleic acid.
  • the term “reference level” as used herein with respect to an mRNA for a nucleic acid listed herein is the level of mRNA for a nucleic acid listed herein that is typically expressed by mammals with prostate cancer that does not progress to systemic disease or result in prostate cancer-specific death.
  • a reference level of an mRNA biomarker listed herein can be the average mRNA level of that biomarker that is present in samples obtained from a random sampling of 50 males without prostate cancer.
  • levels from comparable samples are used when determining whether or not a particular level is an elevated level.
  • the average mRNA level present in bulk prostate tissue from a random sampling of mammals may be X units/g of prostate tissue, while the average mRNA level present in isolated prostate epithelial cells may be Y units/number of prostate cells.
  • the reference level in bulk prostate tissue would be X units/g of prostate tissue, and the reference level in isolated prostate epithelial cells would be Y units/number of prostate cells.
  • the measured level would be compared to the reference level in bulk prostate tissue.
  • the reference level can be a ratio of an expression value of a biomarker in a sample to an expression value of a control nucleic acid or polypeptide in the sample.
  • a control nucleic acid or polypeptide can be any polypeptide or nucleic acid that has a minimal variation in expression level across various samples of the type for which the nucleic acid or polypeptide serves as a control.
  • GAPDH, HPRT, NDUFA7, and RPS16 nucleic acids or polypeptides can be used as control nucleic acids or polypeptides, respectively, in prostate samples.
  • nucleic acids or polypeptides can be used as control nucleic acids or polypeptides, respectively, as described elsewhere (Ohl et al., J. Mol. Med., 83:1014-1024 (2005)).
  • the level of mRNA expression for a particular nucleic acid listed herein (or the degree of which the level is elevated over a reference level) can be combined with the levels of mRNA expression for other particular nucleic acids listed herein to obtain an expression level score.
  • the mRNA levels for each nucleic acid listed in Table 3 can be added together to obtain an expression level score. If this expression level score is greater than the sum of corresponding mRNA reference levels for each nucleic acid listed in Table 3, then the patient, at the time of PSA reoccurrence or retropubic radial prostatectomy, can be classified as later developing systemic disease or, at the time of systemic disease, can be classified as later dying from prostate cancer.
  • the levels of biomarkers can be used in combination with one or more other factors to assess a prostate cancer patient.
  • expression level scores can be used in combination with the clinical stage, the serum PSA level, and/or the Gleason score of the prostate cancer to determine, at the time of PSA reoccurrence or at the time of retropubic radial prostatectomy, whether or not a mammal will later develop systemic disease.
  • such combinations can be used together to determine, at the time of systemic disease, whether or not a mammal will later die of prostate cancer.
  • Additional information about the mammal such as information concerning genetic predisposition to develop cancer, SNPs, chromosomal abnormalities, gene amplifications or deletions, and/or post translational modifications, can also be used in combination with the level of one or more biomarkers provided herein (e.g., the list of nucleic acids set forth in Table 3) to assess prostate cancer patients.
  • biomarkers provided herein (e.g., the list of nucleic acids set forth in Table 3) to assess prostate cancer patients.
  • This document also provides methods and materials to assist medical or research professionals in determining, at the time of PSA reoccurrence or at the time of retropubic radial prostatectomy, whether or not a mammal will later develop systemic disease or in determining, at the time of systemic disease, whether or not a mammal will later die of prostate cancer.
  • Medical professionals can be, for example, doctors, nurses, medical laboratory technologists, and pharmacists.
  • Research professionals can be, for example, principle investigators, research technicians, postdoctoral trainees, and graduate students.
  • a professional can be assisted by (1) determining the level of one or more than one biomarker in a sample, and (2) communicating information about that level to that professional.
  • Any method can be used to communicate information to another person (e.g., a professional).
  • information can be given directly or indirectly to a professional.
  • any type of communication can be used to communicate the information.
  • mail, e-mail, telephone, and face-to-face interactions can be used.
  • the information also can be communicated to a professional by making that information electronically available to the professional.
  • the information can be communicated to a professional by placing the information on a computer database such that the professional can access the information.
  • the information can be communicated to a hospital, clinic, or research facility serving as an agent for the professional.
  • Illumina DASL expression microarrays Two Illumina DASL expression microarrays were utilized for the experiments: (1) The standard commercially available Illumina DASL expression microarray (Cancer PanelTM v1) containing 502 oncogenes, tumor suppressor genes and genes in their associated pathways. Seventy-eight of the targets on the commercial array have been associated with prostate cancer progression. (2) A custom Illumina DASLTM expression microarray containing 526 gene targets for RNAs, including genes whose expression is altered in association with prostate cancer progression. Four different sets of prostate cancer aggressiveness genes were included in the study. If the genes were not present on the Cancer Panel v1 array, then they were included in the design of the custom array:
  • Markers associated with prostate cancer aggressiveness from publicly available expression microarray datasets e.g. EZH2, AMACR, hepsin, PRLz, PRL3: Sufficiently large datasets from 9 prior microarray studies of prostate cancer of varying grades and metastatic potential (Dhanasekaran et al., Nature. 412, 822-826 (2001); Luo et al., Cancer Res. 61, 4683-4688 (2001); Magee et al., Cancer Res. 61, 5692-5696 (2001); Welsh et al., Cancer Res. 61, 5974-5978 (2001); LaTulippe et al., Cancer Res.
  • EZH2 EZH2, AMACR, hepsin, PRLz, PRL3
  • the custom array also included probe sets for 47 genes that were not expected to differ between case and control groups. Thirty-eight of these genes were also present on the commercial array (see Supplemental Tables 1 and 2 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008).
  • RRP Mayo Radical Retropubic Prostatectomy
  • This group of 2,131 men comprises the underlying cohort from which SYS cases and PSA controls were selected.
  • SYS cases systemic progression
  • NED No Evidence of Disease
  • the list of 639 cases and controls was randomized. An attempt was made to identify all available blocks from the RRP (including apparently normal and abnormal lymph nodes) from the randomized list of 639 eligible cases and controls. Maintaining the randomization, each available block was assessed for tissue content by pathology review, and the block containing the dominant Gleason pattern cancer was selected for RNA isolation.
  • Probes for the custom DASL® panel were designed and synthesized by Illumina, Inc. (San Diego, Calif.). RNA samples were processed in following the manufacturer's manual. Samples were hybridized to Sentrix Universal 96-Arrays and scanned using Illumina's BeadArray Reader.
  • mRNA was reverse-transcriptized with SuperScript III First Strand Synthesis SuperMix (Invitrogen, Carlsbad, Calif.) for first strand synthesis using random hexamer. Expression of each gene was measured (the number of cycles required to achieve a threshold, or Ct) in triplicate and then normalized relative to the set of four reference genes.
  • the Gleason score in the Mayo Clinic RRP Registry was the initial diagnostic Gleason score. Since there have been changes in pathologic interpretation of the Gleason Score over time, a single pathologist (JCC) reviewed the Gleason score of each of the blocks selected for expression analysis. This clinical variable was designated as the revised Gleason Score.
  • Discrete clinical factors were tested using Chi-square analysis.
  • Continuous clinical variables (Gleason score (revised), age at PSA recurrence, first rising PSA value, second rising PSA value, and PSA slope) were tested using Wilcoxon rank sum.
  • array normalization is to remove systemic biases introduced during the sample preparation, hybridization, and scanning process. Since different samples were randomly assigned to arrays and positions on arrays, the data was normalized by total fluorescence separately within each disease group within each array type. The normalization technique used was fast cyclic loess (fastlo) (Ballman et al., Bioinformatics. 20, 2778-2786 (2004)).
  • the training data were analyzed using random forests (Breiman, Machine Learning. 45, 5-32 (2001)) using R Version 2.3.1 (http://www.r-project.org) and randomForest version 4.5-16 (http://stat-www.berkeley.edu/users/breiman/RandomForests).
  • the data were analyzed by panel (Cancer, Custom and Merged, where Merged was the Cancer and Custom data treated as a single array). By testing the ntree parameter of the randomForest function, it was determined that 4000 random forests were sufficient to generate a stable list of markers.
  • the resulting scoring function was then analyzed using Receiver Operating Characteristic (ROC) methods, and the cut-off was chosen that assumed an equal penalty for false positives and false negatives.
  • ROC Receiver Operating Characteristic
  • a review of the models permitted a subset of markers to be identified, and a subset of supporting clinical data identified.
  • the number of features in the model was determined by leave 1 ⁇ 3 out Monte Carlo Cross Validation (MCCV) using 100 iterations. The number of features was selected to maximize AUC and minimize random variation in the model.
  • FIG. 5 summarizes the study design.
  • SYS cases were 213 men who developed systemic progression between 90 days and 5.0 years following the PSA rise.
  • PSA control were a random sample of 213 men post-radical prostatectomy with PSA recurrence with no evidence of further clinical progression within 5 years.
  • NED controls were a random sample of 213 men post-radical prostatectomy without PSA rise within 7 years (the comparison of PSA controls with NED controls—to assess markers of PSA recurrence—will be presented in a subsequent paper).
  • the list of eligible cases and controls was scrambled for the blind ascertainment of blocks, isolation of RNA and performance of the expression array experiments.
  • Table 1B summarizes the final block availability, the RNA isolation success rate, and the success rates of the expression array analyses.
  • paraffin blocks were available on 623 (97.5%).
  • RNA was successfully isolated and the DASL assays successfully performed on a very high proportion of patients/specimens: Usable RNA was prepared from all 623 blocks, and the Cancer Panel and custom prostate cancer panel DASL arrays were both successful (after repeating some specimens—see below) on 596 RNA specimens (95.7% of RNAs; 93.3% of design patients). Only 9 (1.4%) RNA specimens failed both expression panels.
  • RNA quality was poor RNA quality—as measured by qRT-PCR of the RPL13A gene expression (Bibikova et al., Genomics, 89(6):666-72 (2007)). Of the 1246 initial samples run on both panels, 87 (7.0%) specimens failed. Those specimens for which there was residual RNA were repeated with a success rate of 77.2% (61 of 79 samples).
  • the study design included several intra- and inter-run array replicates. To determine inter-run array variability, two specimens were run on each of 8 Cancer Panel v1 array runs. The median (range) inter-run correlation coefficients (r2) comparing these two specimen replicates were 0.94 (0.89-0.95) and 0.98 (0.90-0.98), respectively. The same two specimens were run on each of 8 custom prostate cancer panel array runs. The median (range) inter-run correlation coefficients (r2) comparing these specimen replicates were 0.97 (0.95-0.98) and 0.98 (0.96-0.99), respectively.
  • FIG. 6A summarizes the inter-run replicates for one of the specimens on the custom panel. Twelve specimens were evaluated as intra-run array replicates.
  • the median (range) intra-run r2 values comparing these paired specimens on the Cancer Panel v1 was 0.98 (0.93-0.99).
  • the median (range) intra-run r2 values comparing these paired specimens on the custom panel was 0.98 (0.88-0.99).
  • Two specimens were serially diluted, and the expression results of the diluted RNA specimens compared to that of the standard 200 ng of the parental RNA specimen.
  • the r2 for RNA specimens of 25, 50, and 100 ng ranged from 0.98-0.99 ( FIG. 6B ) with slopes near 1.0.
  • RT-PCR analyses were performed for 9 genes (CDH1, VEGF, MUC1, IGFBP3, ERG, TPD52, YWHAZ, FAM13C1, and PAGE4) on 210 samples.
  • Example results are illustrated in FIG. 7 .
  • Comparison of the quantitative RT-PCR and the DASL results gave r2 values of 0.72-0.94 for genes with dynamic range of at least 7 ⁇ CTs. Genes with a smaller dynamic range of ⁇ CT gave r2 values of 0.15-0.79 ( FIG. 7 ).
  • both the DASL and RT-PCR measurements appear to be highly correlated with each other when there is a broad range of RNA expression values.
  • Inter- and Intra-Panel Gene Expression Comparisons By design several genes were evaluated twice on the custom and/or cancer panels. As an example of a specific inter-panel gene expression comparison, probe sets for ERG were present on both the custom (two 3 probe sets) and cancer (one 3 probe set) panels. The r2 comparing the 2 custom probe sets with the commercial probe set for all 596 patients was 0.96 in both cases ( FIG. 8A ). As an example of a specific intra-custom panel gene expression comparison are the probe sets for SRD5A2 and terparbo. Terparbo is a “novel” gene which is likely a variant of the SRD5A2 transcript (UCSC browser, http://genome.ucsc.edu). The r2 comparing the two custom probe sets for SRD5A2 and terparbo was 0.91 ( FIG. 8B ).
  • FIG. 1 illustrates nine genes with significantly different expression in the SYS cases and PSA controls.
  • the training data were analyzed by panel (cancer, custom and merged), by gene (the average expression for all gene-specific probes), and by individual probes.
  • a statistical model to predict systemic progression was developed using random forests (Breiman, Machine Learning. 45, 5-32 (2001)) and logistic regression as described herein. Table 3 lists the 15 genes and 2 individual probes selected for the final model.
  • the node impurity is measured by the Gini index.
  • the Gini index is the weighted average of the impurity in each branch, with impurity being the proportion of incorrectly classified samples in that branch. The larger the Gini decrease, the fewer the misclassification impurities. **Genes mapped to 8q24 ***Single probes for C8orf53 and CDK10 were selected. The Mean Gini Decrease for these probes are derived from an independent random forest analysis of the all probes separately.
  • Table 4 and FIG. 2A summarize the areas under the curve (AUCs) for three clinical models, the final 17 gene/probe model and the combined clinical probe models.
  • the variables in the clinical models were those items of clinical information that would be available at specific times in a patient's course.
  • Clinical model A included revised Gleason score and pathologic stage—information available immediately after RRP. The addition of diagnostic PSA and age at surgery did not significantly add to the AUC and was left out of this model.
  • Clinical model B added age at surgery, preoperative PSA value, and any adjuvant or hormonal therapy within 90 days after RRP—information available at RRP after RRP but before PSA recurrence.
  • Clinical model C added age at PSA recurrence, the second PSA level at time of PSA recurrence, and the PSA slope—information available at the time of PSA recurrence.
  • a pStage or TNM staging system can be used as described elsewhere (e.g., on the World Wide Web at “upmccancercenters.com/cancer/prostate/TNMsystem.html”).
  • clinical models A, B and C alone had AUCs of 0.74 (95% CI 0.68-0.80), 0.76 (95% CI 0.70-0.82) and 0.78 (95% CI 0.73-0.84), respectively.
  • the 17 gene/probe model alone had an AUC of 0.85 (95% CI 0.81-0.90).
  • clinical models A, B, and C had AUCs of 0.86 (95% CI 0.81-0.90), 0.87 (95% CI 0.83-0.91) and 0.88 (95% CI 0.84-0.92), respectively.
  • a 19 gene model that included the 17 gene/probe model as well as the averaged probe sets for TOP2A and survivin (BIRC5) was tested. Expression alterations have previously been reported to be associated with prostate cancer progression for both genes, and they were included in the top 68 gene list (see Table 2). The addition of these two genes did not improve the prediction of systemic progression in the training set.
  • the arrays were designed to contain probe sets for several previously published prostate aggressiveness models (Singh et al., 2002, Glinsky et al., 2004, Lapointe et al., 2004, Yu et al., 2004, Glinsky et al., 2005).
  • Table 4 also summarizes the AUCs for array expression results for these models, with and without the inclusion of the three clinical models.
  • FIG. 2C illustrates the AUCs for four of these models with the appropriate comparison with the clinical model C alone and with the 17 gene/probe model. With the clinical data, each of these models generated AUCs that were less than the developed model. However several of the models generated AUCs (e.g. Lapointe et al. 2004 recurrence model, Yu et al. 2004 model, and Singh et al. 2002 model) that were within or close to the 95% confidence limits of our AUC training set estimates.
  • FIGS. 2B and 2D The 17 gene/probe model and the other previously published models were then applied to the reserved 205 patient validation set ( FIGS. 2B and 2D ).
  • FIG. 2E compares the training and validation set AUCs of the each of the gene/probe models alone. With the exception of the Glinsky et al. 2004 Signature 1, all of the gene/probe models had significantly lower AUCs in the validation set compared to the training set.
  • FIG. 2F compares the training and validation set AUCs of each of the gene/probe models including clinical model C. While the 17 gene/probe model and three of the previously published models (LaPointe et al. 2004 Recurrence model, Yu et al. 2004 model and Glinsky et al. 2005 model) outperformed the clinical model alone, the AUCs were significantly lower in the validation set compared to the training set.
  • the models were compared for their classification of patients into the known PSA progression control and SYS progression case groups.
  • the Cramér's V-statistic (Cramér, 1999) was used.
  • Cramér's V-statistic measures how well two models agree. It is calculated by creating a contingency table (2 ⁇ 2 in this case) and computing a statistic from that table.
  • the lowest Cramér's V value was between the true state (perfect prediction) and the Glinsky et al. 2005 model with clinical data.
  • the highest Cramér's V value was between our 17 gene/probe model and Singh et al. 2002 model, both with clinical data.
  • Most of the models classified the same patients into the known groups (e.g. classifying a patient in the PSA control group as a PSA progression and a patient in the SYS case group as a systemic progression). They also tended to incorrectly classify the same patients (e.g., classifying a patient in the PSA control group as a systemic progression and vice versa).
  • the 17 gene/probe model correctly classified 5-15 more patients into their known category (PSA controls or SYS cases) compared to the other models.
  • the 17 gene/probe model and the previously reported models each classified some of the SYS cases in the good outcome category (e.g. to be PSA recurrences, not systemic progressors) and some of the PSA controls in the poor outcome category (e.g. to go on to systemic progression).
  • good outcome category e.g. to be PSA recurrences, not systemic progressors
  • poor outcome category e.g. to go on to systemic progression
  • FIG. 3A illustrates the systemic progression-free survival for the good and poor outcome groups in the PSA controls.
  • FIG. 3C illustrates the prostate cancer-specific overall survival for the good and poor outcome groups in the SYS cases.
  • the custom array included 82 8q genes on the custom array.
  • ETS-family oncogenes are associated with the development of prostate cancer (Tomilins et al., Science. 310, 644-648 (2005); Tomlins et al., Cancer Res. 66, 3396-3400 (2006); and Demichelis et al., Oncogene. 26:4596-4599 (2007)).
  • Oligonucleotide probe sets for the three major members of the ETS family involved in prostate cancer were included: ERG, ETV1, and ETV4, as well as their translocation partner TMPRSS2.
  • FIG. 4 summarizes the expression results for these genes for the SYS cases and the PSA and NED controls.
  • ETV1 overexpression was 11.5%, 6.5% and 5.1% in the SYS cases, PSA controls and NED controls, respectively (using the cutoff of 6000 normalized fluorescence units—see FIG. 4 ).
  • the prevalence of ETV4 overexpression ranged from 2.5%-5.5% among the three groups and was not significantly different. 6) None of the genes were selected by the formal statistical modeling (see Table 3). In fact, the 17 gene/probe model predicted similar rates of progression in ERG+ and ERG ⁇ patients.
  • IPA Ingenuity Pathway Analysis
  • IPA Ingenuity Systems Inc., Redwood City, Calif.
  • IPA identified 101 canonical pathways that are associated with the focus genes, 51 of which are over-represented with p ⁇ 0.05 (see Supplemental Table 5 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008).
  • the p values from IPA analysis may not accurately quantify the degree of over-representation of focus genes in each pathway.
  • GSEA Gene Set Enrichment Analysis

Abstract

This document provides methods and materials related to assessing male mammals (e.g., humans) with prostate cancer. For example, methods and materials for predicting (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer are provided.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of priority to U.S. Provisional Application Ser. No. 61/057,698, filed May 30, 2008. The disclosure of the prior application is considered part of (and is incorporated by reference in) the disclosure of this application.
  • STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
  • Funding for the work described herein was provided by the federal government under grant number 90966043 awarded by the National Institute of Health. The federal government has certain rights in the invention.
  • BACKGROUND
  • 1. Technical Field
  • This document relates to methods and materials involved in predicting the outcome of prostate cancer.
  • 2. Background Information
  • Prostate cancer occurs when a malignant tumor forms in the tissue of the prostate. The prostate is a gland in the male reproductive system located below the bladder and in front of the rectum. The main function of the prostate gland, which is about the size of a walnut, is to make fluid for semen. Although there are several cell types in the prostate, nearly all prostate cancers start in the gland cells. This type of cancer is known as adenocarcinoma.
  • Prostate cancer is the second leading cause of cancer-related death in American men. Most of the time, prostate cancer grows slowly. Autopsy studies show that many older men who died of other diseases also had prostate cancer that neither they nor their doctor were aware of. Sometimes, however, prostate cancer can grow and spread quickly. It is important to be able to distinguish prostate cancers that will grow slowly from those that will grow quickly since treatment can be especially effective when the cancer has not spread beyond the region of the prostate. Finding ways to detect cancers early can improve survival rates.
  • SUMMARY
  • This document provides methods and materials related to assessing male mammals (e.g., humans) with prostate cancer. For example, this document provides methods and materials for predicting (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer.
  • The majority of men with prostate cancer are diagnosed with cancers with low mortality. Initial treatment is typically radical prostatectomy, external beam radiotherapy, or brachytherapy and followed by serial serum PSA measurements. Not every man who suffers PSA recurrence is destined to suffer systemic progression or to die of his prostate cancer. Thus, it is not clear whether men with PSA recurrence should be simply observed or should receive early androgen ablation. The methods and materials provided herein can be used to predict which men with a rising PSA post-definitive therapy might benefit from additional therapy.
  • In general, one aspect of this document features a method for predicting whether or not a human, at the time of PSA reoccurrence or retropubic radial prostatectomy, will later develop systemic disease. The method comprises, or consists essentially of, (a) determining an expression profile score for cancer tissue from the human, wherein the expression profile score is based on at least the expression levels of RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 nucleic acid, and (b) prognosing the human as later developing systemic disease or as not later developing systemic disease based on at least the expression profile score. The method can be performed at the time of the PSA reoccurrence. The method can be performed at the time of the retropubic radial prostatectomy. The expression levels can be mRNA expression levels. The prognosing step (b) can comprise prognosing the human as later developing systemic disease or as not later developing systemic disease based on at least the expression profile score and a clinical variable. The clinical variable can be selected from the group consisting of a Gleason score and a revised Gleason score. The clinical variable can be selected from the group consisting of a Gleason score, a revised Gleason score, the pStage, age at surgery, initial PSA at recurrence, use of hormone or radiation therapy after radical retropubic prostatectomy, age at PSA recurrence, the second PSA level at time of PSA recurrence, and PSA slope. The method can comprise prognosing the human as later developing systemic disease based on at least the expression profile score. The method can comprise prognosing the human as not later developing systemic disease based on at least the expression profile score.
  • In another aspect, this document features a method for predicting whether or not a human, at the time of systemic disease, will later die from prostate cancer. The method comprises, or consists essentially of, (a) determining an expression profile score for cancer tissue from the human, wherein the expression profile score is based on at least the expression levels of RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 nucleic acid, and (b) prognosing the human as later dying of the prostate cancer or as not later dying of the prostate cancer based on at least the expression profile score. The expression levels can be mRNA expression levels. The prognosing step (b) can comprise prognosing the human as later developing systemic disease or as not later developing systemic disease based on at least the expression profile score and a clinical variable. The clinical variable can be selected from the group consisting of a Gleason score and a revised Gleason score. The clinical variable can be selected from the group consisting of a Gleason score, a revised Gleason score, the pStage, age at surgery, initial PSA at recurrence, use of hormone or radiation therapy after radical retropubic prostatectomy, age at PSA recurrence, the second PSA level at time of PSA recurrence, and PSA slope. The method can comprise prognosing the human as later dying of the prostate cancer based on at least the expression profile score. The method can comprise prognosing the human as not later dying of the prostate cancer based on at least the expression profile score.
  • In another aspect, this document features a method for (1) predicting whether or not a patient, at the time of PSA reoccurrence, will later develop systemic disease, (2) predicting whether or not a patient, at the time of retropubic radial prostatectomy, will later develop systemic disease, or (3) predicting whether or not a patient, at the time of systemic disease, will later die from prostate cancer. The method comprises, or consists essentially of, determining whether or not cancer tissue from the patient contains an RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 expression profile indicative of a later development of the systemic disease or the death.
  • 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 to practice the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. 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.
  • The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1: Nine genes with significantly different expression in cases with systemic disease progression (SYS) versus controls with PSA recurrence (PSA). P-values (t-test) for the SYS case/PSA control comparison are shown. Controls with no evidence of disease recurrence (NED) are also included.
  • FIG. 2: (A to D) Areas under the curve (AUCs) for three clinical models, the final 17 gene/probe model and the combined clinical probe models. A. The training set AUCs for three clinical models, the final 17 gene/probe model and the combined clinical/17 gene/probe model. B. The validation set AUCs for three clinical models, the final 17 gene/probe model and the combined clinical/17 gene/probe model. C. The training set AUCs of four previously reported gene expression models of prostate cancer aggressiveness compared with the Clinical model C alone and with the 17 gene/probe model. D. The validation set AUCs of four previously reported gene expression models of prostate cancer aggressiveness compared with the clinical model C alone and with the 17 gene/probe model. For an explanation of the clinical models see Table 4. (E and F) A comparison of the training and validation set AUCs for each of the model. E. AUCs of the each of the gene/probe models alone. F. AUCs of each of the gene/probe models with the inclusion of clinical model C.
  • FIG. 3: Systemic progression-free and overall prostate cancer-specific survival in the PSA Control and SYS Case groups. A) Systemic progression-free survival for the patients classified in the poor outcome category and for those in the good outcome category in the PSA control group—17 gene/probe model. B) Prostate cancer-specific overall survival for the patients classified in the poor outcome category and for those in the good outcome category in the SYS case group—17 gene/probe model. C) Prostate cancer-specific overall survival for patients classified in the poor outcome category and for those in the good outcome category in the SYS case group—Lapointe et al. 2004 recurrence model.
  • FIG. 4: Expression results for ERG, ETV1 and ETV4 among the men with no evidence of disease progression (NED), PSA recurrence (PSA) and systemic progression (SYS). (A) Each overlapping set of three bars represent a different a different case or control. Thresholds for overexpression are ERG>3200, ETV1>6000 and ETV4>1400. (B) The numbers of cases showing overexpression of one or more of ERG, ETV1 and ETV4 are shown.
  • FIG. 5 is a summary of the nested case-control study design.
  • FIG. 6: Reproducibility of DASL assay and the effect of RNA quantity on the DASL assay. (A) An example of DASL interplate reproducibility. (B) Effect of reduced RNA quantity on the DASL assay.
  • FIG. 7: (A to E) Example results of the comparison of quantitative RT-PCR and DASL data on ERG—Cancer Panel ver1 (A, R2=0.94), ERG—Custom Panel (B, R2=0.94), PAGE4 (C, R2=0.89), MUC1 (D, R2=0.82), and FAM13C1 (E, R2=0.75). (F) Summary of quantitative RT-PCR and DASL data comparisons.
  • FIG. 8: Comparison of genes having multiple probe sets on the Cancer Panel v1 and/or the Custom panel. (A) Comparison of three probe sets (Cancer Panel ERG, Custom Panel ERG and Custom panel ERG splice variant) for ERG. (B) Comparison of two probe sets (Custom Panel SRD5A2 and Custom panel terparbo) for SRD5A2/terparbo.
  • DETAILED DESCRIPTION
  • This document provides methods and materials related to assessing male mammals (e.g., humans) with prostate cancer. For example, this document provides methods and materials for predicting (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer. As described herein, the expression level of any of the genes listed in the tables provided herein (e.g., Tables 2 and 3) or any combination of the genes listed in the tables provided herein can be assessed as described herein to predict (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer. For example, the combination of genes set forth in Table 3 can be assessed as described herein to predict (1) which patients, at the time of PSA reoccurrence, will later develop systemic disease, (2) which patients, at the time of retropubic radial prostatectomy, will later develop systemic disease, and (3) which patients, at the time of systemic disease, will later die from prostate cancer.
  • Any appropriate type of sample (e.g., cancer tissue) can be used to assess the level of gene expression. For example, prostate cancer tissue can be collected and assessed to determine the expression level of a gene listed in any of the tables provided herein. Once obtained, the expression level for a particular nucleic acid can be used as a raw number or can be normalized using appropriate calculations and controls. In addition, the expression levels for groups of nucleic acids can be combined to obtain an expression level score that is based on the measured expression levels (e.g., raw expression level number or normalized number). In some cases, the expression levels of the individual nucleic acids that are used to obtain an expression level score can be weighted. An expression level score can be a whole number, an integer, an alphanumerical value, or any other representation capable of indicating whether or not a condition is met. In some cases, an expression level score is a number that is based on the mRNA expression levels of at least the seventeen nucleic acids listed in Table 3. In some cases, an expression level score can be based on the mRNA expression levels of the seventeen nucleic acids listed in Table 3 and no other nucleic acids. As described herein, the seventeen nucleic acids listed in Table 3 can be used together to determine, at the time of PSA reoccurrence or at the time of retropubic radial prostatectomy, whether or not a mammal will later develop systemic disease. In addition, the seventeen nucleic acids listed in Table 3 can be used together to determine, at the time of systemic disease, whether or not a mammal will later die of prostate cancer.
  • For humans, the seventeen nucleic acids listed in Table 3 can have the nucleic acid sequence set forth in GenBank as follows: RAD21 (GenBank Accession No. NM_006265; GI No. 208879448; probe sequences GGGATAAGAAGCTAACCAAAG-CCCATGTGTTCGAGTGTAATTTAGAGAG (SEQ ID NO:1), GAGGAAAATCGGG-AAGCAGCTTATAATGCCATTACTTTACCTGAAG (SEQ ID NO:2), and TGATT-TTGGAATGGATGATCGTGAGATAATGAGAGAAGGCAGTGCTT (SEQ ID NO:3)), CDKN3 (GenBank Accession Nos. NM_005192 and NM_001130851; GI Nos. 195927023 and 195927024; probe sequences TGAGTTTGACTCATCAGATGAAGAG-CCTATTGAAGATGAACAGACTCCAA (SEQ ID NO:4), TCCTGACATAGCC-AGCTGCTGTGAAATAATGGAAGAGCTTACAACC (SEQ ID NO:5), and TTCGG-GACAAATTAGCTGCACATCTATCATCAAGAGATTCACAATCA (SEQ ID NO:6)), CCNB1 (GenBank Accession No. NM_031966; GI No. 34304372; probe sequences TGCAGCTGGTTGGTGTCACTGCCATGTTTATTGCAAGCAAATAT (SEQ ID NO:7), AACAAGTATGCCACATCGAAGCATGCTAAGATCAGCACTCTACCAC-AG (SEQ ID NO:8), and TTTAGCCAAGGCTGTGGCAAAGGTGTAACTT-GTAAACTTGAGTTGGA (SEQ ID NO:9)), SEC14L1 (GenBank Accession Nos. NM_001039573, NM_001143998, NM_001143999, NM_001144001, and NM_003003; GI Nos. 221316683, 221316675, 221316679, 221316686, and 221316681; probe sequences CATGGTGCAAAAATACCAGTCCCCAGTGAGAGTGTACAA-ATACCCCT (SEQ ID NO:10), TCCTTTGATTCCGATGTTCGTGGGCAGTGAC-ACTGTGAGTGAAT (SEQ ID NO:11), and CACCCTGAAAATGAAGATTG-GACCTGTTTTGAACAGTCTGCAAGTTTA (SEQ ID NO:12)), BUB1 (GenBank Accession No. NM 004336; GI No. 211938448; probe sequences CATGATTGAGC-AAGTGCATGACTGTGAAATCATTCATGGAGACATTAA (SEQ ID NO:13), CTTG-GAAACGGATTTTTGGAACAGGATGATGAAGATGATTTATCTGC (SEQ ID NO:14), and TGAGATGCTCAGCAACAAACCATGGAACTACCAGATCGAT-TACTTT (SEQ ID NO:15)), ALAS1 (GenBank Accession Nos. NM_000688 and NM_199166; GI Nos. 40316942 and 40316938; probe sequences CAGACTCCCTC-ATCACCAAAAAGCAAGTGTCAGTCTGGTGCAGTAAT (SEQ ID NO:16), CAG-GCCTTTCTGCAGAAAGCAGGCAAATCTCTGTTGTTCTATGCC (SEQ ID NO:17), and TTCCAGGACATCATGCAAAAGCAAAGACCAGAAAGAGTGTCTCATC (SEQ ID NO:18)), KIAA0196 (GenBank Accession No. NM_014846; GI No. 120952850; probe sequences AATGCCATCATTGCTGAACTTTTGAGACTCTCTGAGTTTATT-CCTGCT (SEQ ID NO:19), TGGGAAAGCAAACTGGATGCTAAGCCAGAGC-TACAGGATTTAGATGAA (SEQ ID NO:20), and CAACCAGGTGCCAAAAG-ACCATCCAACTATCCCGAGAGCTATTTC (SEQ ID NO:21)), TAF2 (GenBank Accession No. NM_003184; GI No. 115527086; probe sequences TTTGGTTCCC-TTGTGTTGATTCATACTCTGAATTGTGTACATGGAAA (SEQ ID NO:22), TTT-CCCACAGTTGCAAACTTGAATAGAATCAAGTTGAACAGCAAAC (SEQ ID NO:23), and GGCAGAGAGAGGTGCTCATGTTTTCTGTGTGGGTATCAA-AATTCTA (SEQ ID NO:24)), SFRP4 (GenBank Accession No. NM_003014; GI No. 170784837; probe sequences CCATCCCTCGAACTCAAGTCCCGCTCATTACA-AATTCTTCTTGCC (SEQ ID NO:25), AAGAGAGGCTGCAGGAACAG-CGGAGAACAGTTCAGGACAAGAAG (SEQ ID NO:26), and CCAAACCAGCC-AGTCCCAAGAAGAACATTAAAACTAGGAGTGCC (SEQ ID NO:27)), STIP1 (GenBank Accession No. NM_006819; GI No. 110225356; probe sequences CAACA-AGGCCCTGAGCGTGGGTAACATCGATGATGCCTTACA (SEQ ID NO:28), TCAT-GAACCCTTTCAACATGCCTAATCTGTATCAGAAGTTGGAGAGT (SEQ ID NO:29), and AAAAAGAGCTGGGGAACGATGCCTACAAGAAGAAAGACTTTG-ACACA (SEQ ID NO:30)), CTHRC1 (GenBank Accession No. NM_138455; GI No. 34147546; probe sequences CCTGGACACCCAACTACAAGCAGTGTTCATG-GAGTTCATTGAATTAT (SEQ ID NO:31), AGAAATGCATGCTGTCAGCG-TTGGTATTTCACATTCAATGGAGCT (SEQ ID NO:32), ACCAAGGAAGCCCTG-AAATGAATTCAACAATTAATATTCATCGCACT (SEQ ID NO:33)), SLC44A1 (GenBank Accession No. NM_080546; GI No. 112363101; probe sequences CAGTCCT-GTTCAGAATGAGCAAGGCTTTGTGGAGTTCAAAATTTCTG (SEQ ID NO:34), CAATAGCAACAGGTGCAGCAGCAAGACTAGTGTCAGGATACGACAG (SEQ ID NO:35), and GATCCATGCAACCTGGACTTGATAAACCGGAAGATTAAGTCT-GTAG (SEQ ID NO:36)), IGFBP3 (GenBank Accession Nos. NM_000598 and NM_001013398; GI Nos. 62243067 and 62243247; probe sequences CAGCCTCCACA-TTCAGAGGCATCACAAGTAATGGCACAATTCTTC (SEQ ID NO:37), TTCTGAA-ACAAGGGCGTGGATCCCTCAACCAAGAAGAATGTTTATG (SEQ ID NO:38), and TGCTTGGGGACTATTGGAGAAAATAAGGTGGAGTCCTACTTGTTTAA (SEQ ID NO:39)), EDG7 (GenBank Accession No. NM_012152; GI No. 183396778; probe sequences AGTGCCTATGGAACATCCAGCTGATAATCTTGCCTAGTAAGAGC-AAA (SEQ ID NO:40), TTCTGGCACCATTTCGTAGCCATTCTCTTTGTATTTTAA-AAGGACG (SEQ ID NO:41), and CCTCAAAGAAACCATGGCCAGTAGCTAG-GTGTTCAGTAGGAATCAAA (SEQ ID NO:42)), FAM49B (GenBank Accession No. NM_016623; GI No. 42734437; probe sequences TTGCACACCTGTTAGCAAGA-AACAGAAGTTGAAGGACTGGAACAAGT (SEQ ID NO:43), TCCTGTGAAAT-CTCCGAGGAGAAGAAAGAATGATGGACAGTTTATCC (SEQ ID NO:44), and GCAGCATTAAGAGGTCTTCTGGGAGCCTTAACAAGTACCCCATATTCT (SEQ ID NO:45)), C8orf53 (GenBank Accession No. NM_032334; GI No. 223468686; probe sequence GAATTCGGAACAGATCTAACCCAAAAGTACTTTCTGAGAAGCA-GAATG (SEQ ID NO:46)), and CDK10 (GenBank Accession Nos. NM_001098533, NM_001160367, NM_052987, and NM_052988; GI Nos. 237858579, 237858581, 237858574, and 237858573; probe sequence AGGGGTCTCATGTGGTCCTCCTCG-CTATGTTGGAAATGTGCAAC (SEQ ID NO:47)).
  • Any appropriate method can be used to determine the expression level of a gene listed herein. For example, reverse transcription-PCR (RT-PCR) techniques can be performed to detect the level of gene expression.
  • The term “elevated level” as used herein with respect to the level of mRNA for a nucleic acid listed herein is any mRNA level that is greater than a reference mRNA level for that nucleic acid. The term “reference level” as used herein with respect to an mRNA for a nucleic acid listed herein is the level of mRNA for a nucleic acid listed herein that is typically expressed by mammals with prostate cancer that does not progress to systemic disease or result in prostate cancer-specific death. For example, a reference level of an mRNA biomarker listed herein can be the average mRNA level of that biomarker that is present in samples obtained from a random sampling of 50 males without prostate cancer.
  • It will be appreciated that levels from comparable samples are used when determining whether or not a particular level is an elevated level. For example, the average mRNA level present in bulk prostate tissue from a random sampling of mammals may be X units/g of prostate tissue, while the average mRNA level present in isolated prostate epithelial cells may be Y units/number of prostate cells. In this case, the reference level in bulk prostate tissue would be X units/g of prostate tissue, and the reference level in isolated prostate epithelial cells would be Y units/number of prostate cells. Thus, when determining whether or not the level in bulk prostate tissue is elevated, the measured level would be compared to the reference level in bulk prostate tissue. In some cases, the reference level can be a ratio of an expression value of a biomarker in a sample to an expression value of a control nucleic acid or polypeptide in the sample. A control nucleic acid or polypeptide can be any polypeptide or nucleic acid that has a minimal variation in expression level across various samples of the type for which the nucleic acid or polypeptide serves as a control. For example, GAPDH, HPRT, NDUFA7, and RPS16 nucleic acids or polypeptides can be used as control nucleic acids or polypeptides, respectively, in prostate samples. In some cases, nucleic acids or polypeptides can be used as control nucleic acids or polypeptides, respectively, as described elsewhere (Ohl et al., J. Mol. Med., 83:1014-1024 (2005)).
  • Once determined, the level of mRNA expression for a particular nucleic acid listed herein (or the degree of which the level is elevated over a reference level) can be combined with the levels of mRNA expression for other particular nucleic acids listed herein to obtain an expression level score. For example, the mRNA levels for each nucleic acid listed in Table 3 can be added together to obtain an expression level score. If this expression level score is greater than the sum of corresponding mRNA reference levels for each nucleic acid listed in Table 3, then the patient, at the time of PSA reoccurrence or retropubic radial prostatectomy, can be classified as later developing systemic disease or, at the time of systemic disease, can be classified as later dying from prostate cancer.
  • In some cases, the levels of biomarkers (e.g., an expression level score) can be used in combination with one or more other factors to assess a prostate cancer patient. For example, expression level scores can be used in combination with the clinical stage, the serum PSA level, and/or the Gleason score of the prostate cancer to determine, at the time of PSA reoccurrence or at the time of retropubic radial prostatectomy, whether or not a mammal will later develop systemic disease. In addition, such combinations can be used together to determine, at the time of systemic disease, whether or not a mammal will later die of prostate cancer. Additional information about the mammal, such as information concerning genetic predisposition to develop cancer, SNPs, chromosomal abnormalities, gene amplifications or deletions, and/or post translational modifications, can also be used in combination with the level of one or more biomarkers provided herein (e.g., the list of nucleic acids set forth in Table 3) to assess prostate cancer patients.
  • This document also provides methods and materials to assist medical or research professionals in determining, at the time of PSA reoccurrence or at the time of retropubic radial prostatectomy, whether or not a mammal will later develop systemic disease or in determining, at the time of systemic disease, whether or not a mammal will later die of prostate cancer. Medical professionals can be, for example, doctors, nurses, medical laboratory technologists, and pharmacists. Research professionals can be, for example, principle investigators, research technicians, postdoctoral trainees, and graduate students. A professional can be assisted by (1) determining the level of one or more than one biomarker in a sample, and (2) communicating information about that level to that professional.
  • Any method can be used to communicate information to another person (e.g., a professional). For example, information can be given directly or indirectly to a professional. In addition, any type of communication can be used to communicate the information. For example, mail, e-mail, telephone, and face-to-face interactions can be used. The information also can be communicated to a professional by making that information electronically available to the professional. For example, the information can be communicated to a professional by placing the information on a computer database such that the professional can access the information. In addition, the information can be communicated to a hospital, clinic, or research facility serving as an agent for the professional.
  • The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
  • EXAMPLES Example 1 A Tissue Biomarker Panel that Predicts which Men with a Rising PSA Post-Definitive Prostate Cancer Therapy Will have Systemic Progression
  • After therapy for prostate cancer many men develop a rising PSA. Such men may develop a local or metastatic recurrence that warrants further therapy. However many men will have no evidence of disease progression other than the rising PSA and will have a good outcome. A case-control design, incorporating test and validation cohorts, was used to test the association of gene expression results with outcome after PSA progression. Using arrays optimized for paraffin-embedded tissue RNAs, a gene expression model significantly associated with systemic progression after PSA progression was developed. The model also predicted prostate cancer death (in men with systemic progression) and systemic progression beyond 5 years (in PSA controls) with hazard ratios 2.5 and 4.7, respectively (log-rank p-values of 0.0007 and 0.0005). The measurement of gene expression pattern may be useful for determining which men may benefit from additional therapy after PSA recurrence.
  • Gene Selection and Array Design for the DASL™ Assay:
  • Two Illumina DASL expression microarrays were utilized for the experiments: (1) The standard commercially available Illumina DASL expression microarray (Cancer Panel™ v1) containing 502 oncogenes, tumor suppressor genes and genes in their associated pathways. Seventy-eight of the targets on the commercial array have been associated with prostate cancer progression. (2) A custom Illumina DASL™ expression microarray containing 526 gene targets for RNAs, including genes whose expression is altered in association with prostate cancer progression. Four different sets of prostate cancer aggressiveness genes were included in the study. If the genes were not present on the Cancer Panel v1 array, then they were included in the design of the custom array:
  • 1) Markers of prostate cancer aggressiveness identified by a Mayo/University of Minnesota Partnership (Kube et al., BMC Mol. Biol., 8:25 (2007)): The expression profiles of 100 laser-capture microdissected prostate cancer lesions and matched normal and BPH control lesions were analyzed using Affymetrix HG-U133 Plus 2.0 microarrays. Ranked lists of significantly over- and under-expressed genes comparing 10 Gleason 5 and 7 metastatic lesions to 31 Gleason 3 cancer lesions were generated. The top 500 genes on this list were compared to lists generated from prior expression microarray studies and other marker studies of prostate cancer (see 2-4 next). After this analysis there was space for 204 novel targets with potential association with aggressive prostate cancer on the custom array.
  • 2) Markers associated with prostate cancer aggressiveness from publicly available expression microarray datasets (e.g. EZH2, AMACR, hepsin, PRLz, PRL3): Sufficiently large datasets from 9 prior microarray studies of prostate cancer of varying grades and metastatic potential (Dhanasekaran et al., Nature. 412, 822-826 (2001); Luo et al., Cancer Res. 61, 4683-4688 (2001); Magee et al., Cancer Res. 61, 5692-5696 (2001); Welsh et al., Cancer Res. 61, 5974-5978 (2001); LaTulippe et al., Cancer Res. 62, 4499-4506 (2002), Singh et al., Cancer Cell. 1, 203-209 (2002); Glinsky et al., J Clin Invest. 113, 913-923 (2004); Lapointe et al., Proc Natl Acad Sci USA. 101, 811-816 (2004); and Yu et al., J Clin Oncol. 22, 2790-2799 (2004)) were available from the OncoMine internet site (Rhodes et al., Neoplasia. 6, 1-6 (2004); Rhodes et al., Proc Natl Acad Sci USA. 101, 9309-9314 (2004); www.oncomine.org) when the array was designed. From ordered lists of these data, 32 genes were selected for inclusion on the array.
  • 3) Previously published markers associated with prostate cancer aggressiveness (e.g. PSMA, PSCA, Cav-1): Expression microarray data has also been published. This literature was evaluated for additional tissue biomarkers. For example, at the time of array design 13 high quality expression microarray studies of prostate cancer aggressiveness were identified (See Supplemental Tables 1 and 2 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008, for full reference list). In addition, among the 13 reports, 5 papers presented 8 expression biomarker panels to predict prostate cancer aggressiveness (Singh et al., Cancer Cell. 1, 203-209 (2002); Glinsky et al., J Clin Invest. 113, 913-923 (2004); Lapointe et al., Proc Natl Acad Sci USA. 101, 811-816 (2004); Yu et al., J Clin Oncol. 22, 2790-2799 (2004); and Glinsky et al., J Clin Invest. 115, 1503-1521 (2005)). When appropriate probes suitable for the DASL chemistry could be designed for these panels they were included on the custom array. 12 articles were identified reviewing genes associated with prostate cancer. These criteria resulted in the selection of 150 genes.
  • 4) Markers derived from Mayo SPORE research (including genes and ESTs mapped to 8q24). Ninety-three additional biomarkers were identified (see Supplemental Tables 1 and 2 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008).
  • The custom array also included probe sets for 47 genes that were not expected to differ between case and control groups. Thirty-eight of these genes were also present on the commercial array (see Supplemental Tables 1 and 2 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008).
  • After enumerating the potentially prostate cancer relevant genes on the commercially available cancer panel, 557 potentially prostate cancer relevant genes and 424 other cancer-related genes were evaluated across both arrays.
  • Design of Nested Case-Control Study:
  • Since training and validation analysis requires tissue from patients with sufficient follow-up time, for this study individuals from the Mayo Radical Retropubic Prostatectomy (RRP) Registry were sampled. The registry consists of a population of men who received prostatectomy as their first treatment for prostate cancer at the Mayo Clinic (For a current description and use of the registry; see Tollefson et al., Mayo Clin Proc. 82, 422-427 (2007)). As systemic progression is relatively infrequent, a case-control study nested within a cohort of men with a rising PSA was designed. Between 1987-2001, inclusive, 9,989 previously-untreated men had RRP at Mayo. On follow-up, 2,131 developed a rising PSA (>30 days after RRP) in the absence of concurrent clinical recurrence. PSA rise was defined as a follow-up PSA>=0.20 ng/ml, with the next PSA at least 0.05 ng/ml higher or the initiation of treatment for PSA recurrence (for patients whose follow-up PSA was high enough to warrant treatment). This group of 2,131 men comprises the underlying cohort from which SYS cases and PSA controls were selected.
  • Within 5 years of PSA rise, 213 men developed systemic progression (SYS cases), defined as a positive bone scan or CT scan. Of these, 100 men succumbed to a prostate cancer-specific death, 37 died from other causes, and 76 remain at risk.
  • PSA progression controls (213) were selected from those men without systemic progression within 5 years after the PSA rise and were matched (1:1) on birth year, calendar year of PSA rise and initial diagnostic pathologic Gleason score (<=6, 7+). Twenty of these men developed systemic progression greater than 5 years after initial PSA rise and 9 succumbed to a prostate cancer-specific death.
  • A set of 213 No Evidence of Disease (NED) Progression controls were also selected from the Mayo RRP Registry of 9,989 men and used for some comparisons. These controls had RRP from 1987-1998 with no evidence of PSA rise within 7 years of RRP. The median (25th, 75th percentile) follow-up from RRP was 11.3 (9.3, 13.8) years. The NED controls were matched to the systemic progression cases on birth-year, calendar year of RRP and initial diagnostic Gleason Score. Computerized optimal matching was performed to minimize the total “distance” between cases and controls in terms of the sum of the absolute difference in the matching factors (Bergstralh et al., Epidemiology. 6, 271-275 (1995)).
  • Block Identification, RNA Isolation, and Expression Analysis:
  • The list of 639 cases and controls was randomized. An attempt was made to identify all available blocks from the RRP (including apparently normal and abnormal lymph nodes) from the randomized list of 639 eligible cases and controls. Maintaining the randomization, each available block was assessed for tissue content by pathology review, and the block containing the dominant Gleason pattern cancer was selected for RNA isolation.
  • Four freshly cut 10 μm sections of FFPE tissue were deparaffinized and the Gleason dominant cancer focus was macrodissected. RNA was extracted using the High Pure RNA Paraffin Kit from Roche (Indianapolis, Ind.). RNA was quantified using ND-1000 spectrophotometer from NanoDrop Technologies (Wilmington, Del.). The RNAs were distributed on 96-well plates in the randomized order for DASL analysis (including within-run and between-run duplicates).
  • Probes for the custom DASL® panel were designed and synthesized by Illumina, Inc. (San Diego, Calif.). RNA samples were processed in following the manufacturer's manual. Samples were hybridized to Sentrix Universal 96-Arrays and scanned using Illumina's BeadArray Reader.
  • In order to evaluate the accuracy of the gene expression levels defined by the DASL technology, quantitative SYBR Green RT-PCR reactions were performed for 9 selected “target” genes (CDH1, MUC1, VEGF, IGFBP3, ERG, TPD52, YWHAZ, FAM13C1, and PAGE4) and four commonly-used endogenous control genes (GAPDH, B2M, PPIA and RPL13a) in 384-well plates, with the use of Prism 7900HT instruments (Applied Biosystems, Foster City, Calif.). 210 RNA samples with abundant mRNA from the group of total 639 patients were analyzed. For the PAGE4 assay, only 77 samples were subjected to the assay because of mRNA shortage. mRNA was reverse-transcriptized with SuperScript III First Strand Synthesis SuperMix (Invitrogen, Carlsbad, Calif.) for first strand synthesis using random hexamer. Expression of each gene was measured (the number of cycles required to achieve a threshold, or Ct) in triplicate and then normalized relative to the set of four reference genes.
  • Pathology Review:
  • The Gleason score in the Mayo Clinic RRP Registry was the initial diagnostic Gleason score. Since there have been changes in pathologic interpretation of the Gleason Score over time, a single pathologist (JCC) reviewed the Gleason score of each of the blocks selected for expression analysis. This clinical variable was designated as the revised Gleason Score.
  • Statistical Methodology:
  • Collection of gene expression data was attempted for the 623 patients as described herein. Of these, there were 596 (nSYS=200, nPSA=201, nNED=195) patients for whom data was collected, the rest having failed one or both expression panels as described herein. To assure selection of similar training and validation sets, 100 case-control-control cohorts comprised of 133 randomly chosen SYS patients (two-thirds of 200 for training) along with their matched PSA and NED controls were selected as a proposed training set. The remaining cases and controls were treated as a proposed validation set. The clinical variables were tested for independence between the proposed training and validation sets separately within the SYS cases and the PSA controls. Discrete clinical factors (pathologic stage, hormonal treatment adjuvant to RRP, radiation treatment adjuvant to RRP, hormonal treatment adjuvant to PSA recurrence, and radiation therapy adjuvant to PSA recurrence) were tested using Chi-square analysis. Continuous clinical variables (Gleason score (revised), age at PSA recurrence, first rising PSA value, second rising PSA value, and PSA slope) were tested using Wilcoxon rank sum. Six of the one hundred randomly sampled sets failed to show dependency for any of the clinical variables at the 0.2 level, and the first of these was chosen as the training set: 391 patients (nSYS=133, nPSA=133, nNED=125). This reserved 205 patients for the validation set (nSYS=67, nPSA=68, nNED=70).
  • The purpose of array normalization is to remove systemic biases introduced during the sample preparation, hybridization, and scanning process. Since different samples were randomly assigned to arrays and positions on arrays, the data was normalized by total fluorescence separately within each disease group within each array type. The normalization technique used was fast cyclic loess (fastlo) (Ballman et al., Bioinformatics. 20, 2778-2786 (2004)).
  • The training data were analyzed using random forests (Breiman, Machine Learning. 45, 5-32 (2001)) using R Version 2.3.1 (http://www.r-project.org) and randomForest version 4.5-16 (http://stat-www.berkeley.edu/users/breiman/RandomForests). The data were analyzed by panel (Cancer, Custom and Merged, where Merged was the Cancer and Custom data treated as a single array). By testing the ntree parameter of the randomForest function, it was determined that 4000 random forests were sufficient to generate a stable list of markers. The top markers as sorted for significance by the randomForest program were combined with various combinations of clinical variables using logistic regression R program (glm( ) with family=binary (a logistic model), where glm refers to generalized linear model). The resulting scoring function was then analyzed using Receiver Operating Characteristic (ROC) methods, and the cut-off was chosen that assumed an equal penalty for false positives and false negatives. A review of the models permitted a subset of markers to be identified, and a subset of supporting clinical data identified. The number of features in the model was determined by leave ⅓ out Monte Carlo Cross Validation (MCCV) using 100 iterations. The number of features was selected to maximize AUC and minimize random variation in the model. The final model was then applied to the 391 patient training set and the reserved 205 patient validation set. For comparison, other previously reported gene expression models were also tested against the training and validation sets (Singh et al., Cancer Cell. 1, 203-209 (2002); Glinsky et al., J Clin Invest. 113, 913-923 (2004); Lapointe et al., Proc Natl Acad Sci USA. 101, 811-816 (2004); Yu et al., J Clin Oncol. 22, 2790-2799 (2004); and Glinsky et al., J Clin Invest. 115, 1503-1521 (2005)).
  • Study Design/Paraffin Block Recovery/RNA Isolation and Expression Panel Success
  • Briefly, a nested case-control study was performed using the large, well-defined cohort of men with rising PSA following radical prostatectomy at our institution. FIG. 5 summarizes the study design. SYS cases were 213 men who developed systemic progression between 90 days and 5.0 years following the PSA rise. PSA control were a random sample of 213 men post-radical prostatectomy with PSA recurrence with no evidence of further clinical progression within 5 years. NED controls were a random sample of 213 men post-radical prostatectomy without PSA rise within 7 years (the comparison of PSA controls with NED controls—to assess markers of PSA recurrence—will be presented in a subsequent paper). SYS cases and PSA controls were matched (1:1) on birth year, calendar year of PSA rise, initial diagnostic pathologic Gleason score (<=6 vs. >=7). The list of eligible cases and controls was scrambled for the blind ascertainment of blocks, isolation of RNA and performance of the expression array experiments.
  • Table 1A summarizes the distribution of clinical parameters between the SYS cases and the PSA and NED control groups. As expected, there was no significant difference between the groups for the variables used for matching (there was no significant difference in Gleason score when the <=6 and >7 groups—the matching criteria—were compared). Because Gleason scoring may have changed over time, all of the macrodissected lesions were blindly re-graded by a single experienced pathologist (providing a revised Gleason score). As expected, Gleason scores have increased over time. In addition, the proportion of Gleason 8-10 tumors increased comparing NED controls to PSA controls, and PSA controls to SYS cases. Because of this change in grade, the revised Gleason score was used in all the biomarker modeling.
  • TABLE 1A
    Systemic progression (SYS) Case and PSA recurrence (PSA) and
    no evidence of disease (NED) control patient demographics
    Progression group p-value
    NED PSA SYS NED vs. PSA vs.
    controls controls cases PSA SYS
    Year of surgery 0.707 0.592
    N 213 213 213
    Median 1992 1992 1992
    Q1, Q3 1989, 1995 1990, 1995 1989, 1995
    Age at RRP 0.682 0.496
    N 213 213 213
    Median 67 67 67
    Q1, Q3 61, 70 61, 70 61, 70
    PSA at RRP 0.001 0.957
    N 205 208 204
    Median 8.1 10.5 10.6
    Q1, Q3  5.1, 13.1  6.4, 21.4  6.5, 20.7
    Gleason score, original 0.411 0.024
    Missing 12 6 14
    <=6 45 (22.4%) 48 (23.2%) 46 (23.1%)
      7 139 (69.2%) 129 (62.3%) 94 (47.2%)
    8-10 17 (8.5%) 30 (14.5%) 59 (29.6%)
    Gleason score, revised 0.002 <0.001
    Missing 8 2 6
    <=6 50 (22.4%) 32 (15.2%) 8 (3.9%)
      7 114 (55.6%) 113 (53.6%) 75 (36.2%)
    8-10 41 (20.0%) 66 (31.3%) 124 (59.9%)
    Stage 0.138 <0.001
    T2N0 118 (55.4%) 95 (44.6%) 59 (27.7%)
    T3aN0 43 (20.2%) 53 (24.9%) 47 (22.1%)
    T3bN0 21 (9.9%) 54 (25.4%) 56 (26.3%)
    T3xN+ 31 (14.6%) 11 (5.2%) 51 (23.9%)
    Ploidy 0.525 0.001
    Missing 13 9 1
    Diploid 136 (68.0%) 128 (62.7%) 97 (45.8%)
    Tetraploid 53 (26.5%) 61 (29.9%) 84 (39.6%)
    Aneuploid 11 (5.5%) 15 (7.4%) 31 (14.6%)
    Age at PSA recurrence NA 0.558
    N 213 213
    Median 69.1 69.6
    Q1, Q3 64.2, 73.4 64.7, 73.8
  • All paraffin-embedded blocks from eligible men were identified, and each block was surveyed for the tissue present (primary and secondary Gleason cancer regions, normal and metastatic lymph nodes, etc.). The dominant Gleason pattern region was macrodissected from the available blocks, and RNA was isolated from that region. Illumina Cancer Panel™ and custom prostate cancer panel DASL array analyses were then performed on all RNA specimens. The Experimental Procedures section and Supplemental Tables 1 & 2 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008, describe the composition of the Cancer Panel and the design of the Custom Panel.
  • Table 1B summarizes the final block availability, the RNA isolation success rate, and the success rates of the expression array analyses. Of the 639 eligible patients, paraffin blocks were available on 623 (97.5%). Similarly, RNA was successfully isolated and the DASL assays successfully performed on a very high proportion of patients/specimens: Usable RNA was prepared from all 623 blocks, and the Cancer Panel and custom prostate cancer panel DASL arrays were both successful (after repeating some specimens—see below) on 596 RNA specimens (95.7% of RNAs; 93.3% of design patients). Only 9 (1.4%) RNA specimens failed both expression panels. The primary reason for these failures was poor RNA quality—as measured by qRT-PCR of the RPL13A gene expression (Bibikova et al., Genomics, 89(6):666-72 (2007)). Of the 1246 initial samples run on both panels, 87 (7.0%) specimens failed. Those specimens for which there was residual RNA were repeated with a success rate of 77.2% (61 of 79 samples).
  • TABLE 1B
    Availability of blocks, RNA isolation success and DASL assay success
    Pregression Case/
    Control Group
    None PSA Systemic Total
    Design Number
    213 213 213 639
    Blocks Available 205 211 207 623 (97.5%)
    Usable RNA 205 211 207 623
    Evaluable Data, Both DASL 195 201 200 596 (95.7%)
    Panels
    Evaluable Data, Cancer Panel 3 5 2 10
    Evaluable Data, Custom Panel 2 3 3 8
    Failed Both Panels 5 2 2  9 (1.4%)
  • Expression Analysis Reproducibility
  • Replicate analysis results, RT-PCR comparisons, and inter- and intra-panel gene expression comparisons are as follows.
  • Replicate analyses: The study design included several intra- and inter-run array replicates. To determine inter-run array variability, two specimens were run on each of 8 Cancer Panel v1 array runs. The median (range) inter-run correlation coefficients (r2) comparing these two specimen replicates were 0.94 (0.89-0.95) and 0.98 (0.90-0.98), respectively. The same two specimens were run on each of 8 custom prostate cancer panel array runs. The median (range) inter-run correlation coefficients (r2) comparing these specimen replicates were 0.97 (0.95-0.98) and 0.98 (0.96-0.99), respectively. FIG. 6A summarizes the inter-run replicates for one of the specimens on the custom panel. Twelve specimens were evaluated as intra-run array replicates. The median (range) intra-run r2 values comparing these paired specimens on the Cancer Panel v1 was 0.98 (0.93-0.99). The median (range) intra-run r2 values comparing these paired specimens on the custom panel was 0.98 (0.88-0.99). Two specimens were serially diluted, and the expression results of the diluted RNA specimens compared to that of the standard 200 ng of the parental RNA specimen. The r2 for RNA specimens of 25, 50, and 100 ng ranged from 0.98-0.99 (FIG. 6B) with slopes near 1.0.
  • Comparison with RT-PCR: RT-PCR analyses were performed for 9 genes (CDH1, VEGF, MUC1, IGFBP3, ERG, TPD52, YWHAZ, FAM13C1, and PAGE4) on 210 samples. Example results are illustrated in FIG. 7. Comparison of the quantitative RT-PCR and the DASL results gave r2 values of 0.72-0.94 for genes with dynamic range of at least 7 ΔCTs. Genes with a smaller dynamic range of ΔCT gave r2 values of 0.15-0.79 (FIG. 7). Thus, both the DASL and RT-PCR measurements appear to be highly correlated with each other when there is a broad range of RNA expression values.
  • Inter- and Intra-Panel Gene Expression Comparisons: By design several genes were evaluated twice on the custom and/or cancer panels. As an example of a specific inter-panel gene expression comparison, probe sets for ERG were present on both the custom (two 3 probe sets) and cancer (one 3 probe set) panels. The r2 comparing the 2 custom probe sets with the commercial probe set for all 596 patients was 0.96 in both cases (FIG. 8A). As an example of a specific intra-custom panel gene expression comparison are the probe sets for SRD5A2 and terparbo. Terparbo is a “novel” gene which is likely a variant of the SRD5A2 transcript (UCSC browser, http://genome.ucsc.edu). The r2 comparing the two custom probe sets for SRD5A2 and terparbo was 0.91 (FIG. 8B).
  • Specific Gene Expression Results Comparing the Systemic Progression Cohorts with the PSA Progression and No Evidence of Progression Cohorts:
  • Univariate Analyses by gene: Because the DASL assay appeared to generate precise and reproducible results, the array data was examined for genes whose expression was significantly altered when the SYS cases were compared with the PSA Controls. For this initial analysis, the DASL gene expression value was determined to be the average of the up-to-three probes for each gene on each array. Upon univariate analysis (two-sided t-test) of the probe-averaged and total fluorescence fast-lo normalized data, 68 genes were highly significantly over- or under-expressed in the SYS cases versus PSA controls (p<9.73×10−7, Bonferroni correction for p<0.001) (Table 2). One hundred twenty-six genes were significantly over- or under-expressed in the SYS cases versus the PSA controls (p<4.86×105, Bonferroni correction for p<0.05). Supplemental Table 3 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008, provides the complete gene list ordered by p-value. FIG. 1 illustrates nine genes with significantly different expression in the SYS cases and PSA controls.
  • TABLE 2
    Top 68 genes highly significantly correlated with prostate cancer systemic
    progression (p < 0.001; with Bonferroni correction p < 9.73E−07).
    DASL fast-lo Normalized
    Expression Value Systemic Systemic
    Systemic PSA to PSA to PSA
    Rank Gene Name Gene ID* Progression Progression Fold change p-value**
    1 RAD21*** NM_006265 7587 6409 1.18 8.57E−14
    2 YWHAZ NM_145690 15625 13417 1.16 1.92E−13
    3 TAF2*** NM_003184 3144 2681 1.17 6.99E−13
    4 SLC44A1 NM_080546 4669 4022 1.16 2.74E−12
    5 IGFBP3 NM_000598 4815 3782 1.27 3.75E−12
    6 RHOA NM_001664 15859 14542 1.09 1.22E−11
    7 MTPN NM_145808 7646 6840 1.12 1.69E−11
    8 BUB1 NM_001211 1257 957 1.31 2.07E−11
    9 TUBB NM_178014 17412 15659 1.11 6.52E−11
    10 CHRAC1*** NM_017444 3905 3233 1.21 6.74E−11
    11 HPRT1 NM_000194 3613 3179 1.14 8.19E−11
    12 SEC14L1 NM_003003 7248 6185 1.17 8.20E−11
    13 SOD1 NM_000454 17412 16043 1.09 1.30E−10
    14 ENY2 NM_020189 7597 6493 1.17 2.04E−10
    15 CCNB1 NM_031966 1871 1342 1.39 3.65E−10
    16 INHBA NM_002192 4859 3732 1.30 5.18E−10
    17 TOP2A NM_001067 5550 4123 1.35 7.42E−10
    18 ATP5J NM_001003703 13145 11517 1.14 1.75E−09
    19 C8orf53*** NM_032334 7373 6444 1.14 1.88E−09
    20 EIF3S3*** NM_003756 11946 10798 1.11 1.98E−09
    21 EIF2C2*** NM_012154 5908 5338 1.11 2.12E−09
    22 CDKN3 NM_005192 1562 1229 1.27 2.32E−09
    23 TPX2 NM_012112 1193 861 1.39 2.64E−09
    24 GLRX2 NM_197962 4154 3319 1.25 3.13E−09
    25 CTHRC1 NM_138455 3136 2480 1.26 3.83E−09
    26 KIAA0196*** NM_014846 5530 4945 1.12 4.12E−09
    27 DHX9 NM_030588 7067 6607 1.07 5.02E−09
    28 FAM13C1 NM_001001971 4448 5416 0.82 9.07E−09
    29 CSTB NM_000100 16424 15379 1.07 1.57E−08
    30 SESN3.a SESN3.a 8467 6811 1.24 1.99E−08
    31 SQLE*** NM_003129 2282 1832 1.25 2.43E−08
    32 IMMT NM_006839 4683 4190 1.12 2.43E−08
    33 MKI67 NM_002417 4204 3261 1.29 2.91E−08
    34 MRPL13*** NM_014078 5051 4158 1.21 3.80E−08
    35 SRD5A2 NM_000348 2318 2795 0.83 4.63E−08
    36 EZH2 NM_004456 3806 3257 1.17 4.76E−08
    37 F2R NM_001992 3856 3203 1.20 5.61E−08
    38 SH3KF2.a SH3RF2.a 1394 1705 0.82 6.48E−08
    39 ZNF313 NM_018683 9542 8766 1.09 7.14E−08
    40 SDHC NM_001035511 2363 2082 1.14 7.35E−08
    41 PGK1 NM_000291 2313 2001 1.16 7.84E−08
    42 GNPTAB NM_024312 5427 4587 1.18 9.04E−08
    43 meelar.d meelar.d 2566 3478 0.74 9.59E−08
    44 THBS2 NM_003247 3047 2458 1.24 9.72E−08
    45 BIRC5 NM_001168 2451 1802 1.36 1.00E−07
    46 POSTN NM_006475 7210 5812 1.24 1.02E−07
    47 GNB1 NM_002074 12350 11206 1.10 1.20E−07
    48 FAM49B*** NM_016623 6291 5661 1.11 1.21E−07
    49 WDR67*** NM_145647 1655 1423 1.16 1.67E−07
    50 TMEM65.a*** TMEM65.a 4117 3540 1.16 1.96E−07
    51 GMNN NM_015895 7458 5945 1.25 1.99E−07
    52 PAGE4 NM_007003 6419 8065 0.80 2.00E−07
    53 MYBPC1 NM_206821 8768 11120 0.79 2.61E−07
    54 GPR137B NM_003272 3997 3447 1.16 2.96E−07
    55 ALAS1 NM_000688 5380 5035 1.07 3.55E−07
    56 MSR1 NM_002445 3663 3025 1.21 3.65E−07
    57 CDC2 NM_033379 1420 1130 1.26 3.90E−07
    58 240093_x_at 240093_x_at 1789 1469 1.22 4.71E−07
    59 IGFBP3 NM_000598 10673 9433 1.13 4.85E−07
    60 RAP2B NM_002886 3270 2922 1.12 5.00E−07
    61 MGC14595.a*** MGC14595.a 2252 1995 1.13 5.46E−07
    62 AZGP1 NM_001185 17252 20133 0.86 6.55E−07
    63 NOX4 NM_016931 2321 1942 1.19 6.67E−07
    64 STIP1 NM_006819 7630 7123 1.07 7.23E−07
    65 PTPRN2 NM_130843 4471 5398 0.83 7.36E−07
    66 CTNNB1 NM_001904 9989 9354 1.07 7.50E−07
    67 C8orf76*** NM_032847 4088 3652 1.12 7.88E−07
    68 YY1 NM_003403 9529 8635 1.10 8.08E−07
    *The Gene ID is the accession number when available. Other Gene IDs can be found by searching the May 2004 assembly of the human genome at http://genome.ucsc.edu/cgi-bin/hgGateway.
    **t-test
    ***Genes mapped to 8q24
  • Systemic Progression Prediction Model Development and Testing on Training Set:
  • The training data were analyzed by panel (cancer, custom and merged), by gene (the average expression for all gene-specific probes), and by individual probes. A statistical model to predict systemic progression (with and without clinical variables) was developed using random forests (Breiman, Machine Learning. 45, 5-32 (2001)) and logistic regression as described herein. Table 3 lists the 15 genes and 2 individual probes selected for the final model.
  • TABLE 3
    Final random forest 17 gene/probe model to predict prostate cancer systemic
    progression after a rising PSA following radical prostatectomy
    Mean DASL Expression Values
    t-test Mean Gini p-value Systemic PSA Systemic:PSA
    Rank Symbol Decrease* (t-test) Progression Progression Fold Change
    1 RAD21** 2.15 8.57E−14 7587 6409 1.18
    22 CDKN3 1.28 2.32E−09 1562 1229 1.27
    15 CCNB1 1.25 3.65E−10 1871 1342 1.39
    12 SEC14L1 1.14 8.20E−11 7248 6185 1.17
    8 BUB1 1.06 2.07E−11 1257 957 1.31
    55 ALAS1 1.04 3.55E−07 5380 5035 1.07
    26 KIAA0196** 1.02 4.12E−09 5530 4945 1.12
    3 TAF2** 1.02 6.99E−13 3144 2681 1.17
    78 SFRP4 0.99 1.89E−06 15176 13059 1.16
    64 STIP1 0.95 7.23E−07 7630 7123 1.07
    25 CTHRC1 0.90 3.83E−09 3136 2480 1.26
    4 SLC44A1 0.90 2.74E−12 4669 4022 1.17
    5 IGFBP3 0.85 3.75E−12 4815 3782 1.27
    307 EDG7 0.82 7.07E−03 5962 6757 0.88
    48 FAM49B** 0.82 1.21E−07 6291 5661 1.11
    19 C8orf53** 0.97*** 1.88E−09 7373 6444 1.14
    275 CDK10 0.53*** 4.12E−03 12254 12868 0.95
    *Mean Gini Decrease for a variable is the average (over all random forest trees) decrease in node impurities from recursive partitioning splits on that variable. For classification, the node impurity is measured by the Gini index. The Gini index is the weighted average of the impurity in each branch, with impurity being the proportion of incorrectly classified samples in that branch. The larger the Gini decrease, the fewer the misclassification impurities.
    **Genes mapped to 8q24
    ***Single probes for C8orf53 and CDK10 were selected. The Mean Gini Decrease for these probes are derived from an independent random forest analysis of the all probes separately.
  • Table 4 and FIG. 2A summarize the areas under the curve (AUCs) for three clinical models, the final 17 gene/probe model and the combined clinical probe models. The variables in the clinical models were those items of clinical information that would be available at specific times in a patient's course. Clinical model A included revised Gleason score and pathologic stage—information available immediately after RRP. The addition of diagnostic PSA and age at surgery did not significantly add to the AUC and was left out of this model. Clinical model B added age at surgery, preoperative PSA value, and any adjuvant or hormonal therapy within 90 days after RRP—information available at RRP after RRP but before PSA recurrence. Clinical model C added age at PSA recurrence, the second PSA level at time of PSA recurrence, and the PSA slope—information available at the time of PSA recurrence.
  • TABLE 4
    Prediction of systemic progression - training set AUCs
    Probes Clinical model*
    alone A B C
    Clinical model alone NA 0.736 0.757 0.783
    Final 17 gene/probe 0.852 0.857 0.873 0.883
    Glinsky et al. 2004 Signature 1 0.665 0.762 0.776 0.798
    Glinsky et al. 2004 Signature 2 0.638 0.764 0.781 0.798
    Glinsky et al. 2004 Signature 3 0.669 0.770 0.788 0.810
    Glinsky et al. 2005 0.729 0.780 0.800 0.811
    Lapointe et al. 2004 Tumor 0.789 0.825 0.838 0.855
    Recurrence Sig.
    Lapointe et al. 2004 (MUC1 and AZGP1) 0.660 0.767 0.777 0.793
    Singh et al. 2002 0.783 0.824 0.838 0.851
    Yu et al. 2004 0.725 0.797 0.815 0.830
    *Clinical model
    Clinical variable A B C
    Revised Gleason score X X X
    pStage X X X
    Age at surgery X X
    Initial PSA at recurrence X X
    Hormone or radiation therapy after RRP X X
    Age at PSA recurrence X
    Second PSA X
    PSA slope X
  • A pStage or TNM staging system can be used as described elsewhere (e.g., on the World Wide Web at “upmccancercenters.com/cancer/prostate/TNMsystem.html”).
  • Using the training set, clinical models A, B and C alone had AUCs of 0.74 (95% CI 0.68-0.80), 0.76 (95% CI 0.70-0.82) and 0.78 (95% CI 0.73-0.84), respectively. The 17 gene/probe model alone had an AUC of 0.85 (95% CI 0.81-0.90). Together with the 17 gene/probe model, clinical models A, B, and C had AUCs of 0.86 (95% CI 0.81-0.90), 0.87 (95% CI 0.83-0.91) and 0.88 (95% CI 0.84-0.92), respectively. A 19 gene model that included the 17 gene/probe model as well as the averaged probe sets for TOP2A and survivin (BIRC5) was tested. Expression alterations have previously been reported to be associated with prostate cancer progression for both genes, and they were included in the top 68 gene list (see Table 2). The addition of these two genes did not improve the prediction of systemic progression in the training set.
  • The arrays were designed to contain probe sets for several previously published prostate aggressiveness models (Singh et al., 2002, Glinsky et al., 2004, Lapointe et al., 2004, Yu et al., 2004, Glinsky et al., 2005). Table 4 also summarizes the AUCs for array expression results for these models, with and without the inclusion of the three clinical models. FIG. 2C illustrates the AUCs for four of these models with the appropriate comparison with the clinical model C alone and with the 17 gene/probe model. With the clinical data, each of these models generated AUCs that were less than the developed model. However several of the models generated AUCs (e.g. Lapointe et al. 2004 recurrence model, Yu et al. 2004 model, and Singh et al. 2002 model) that were within or close to the 95% confidence limits of our AUC training set estimates.
  • Testing of Models on the Validation Set:
  • The 17 gene/probe model and the other previously published models were then applied to the reserved 205 patient validation set (FIGS. 2B and 2D). FIG. 2E compares the training and validation set AUCs of the each of the gene/probe models alone. With the exception of the Glinsky et al. 2004 Signature 1, all of the gene/probe models had significantly lower AUCs in the validation set compared to the training set. FIG. 2F compares the training and validation set AUCs of each of the gene/probe models including clinical model C. While the 17 gene/probe model and three of the previously published models (LaPointe et al. 2004 Recurrence model, Yu et al. 2004 model and Glinsky et al. 2005 model) outperformed the clinical model alone, the AUCs were significantly lower in the validation set compared to the training set.
  • The models were compared for their classification of patients into the known PSA progression control and SYS progression case groups. To compare models, the Cramér's V-statistic (Cramér, 1999) was used. Cramér's V-statistic measures how well two models agree. It is calculated by creating a contingency table (2×2 in this case) and computing a statistic from that table. Supplemental Table 4 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008, summarizes the Cramér's V-statistic of the various models, and includes a perfect predictor (“truth”) model for direct evaluation of the models. Briefly, the Cramér's V-statistic ranged from 0.38 to 0.70. The lowest Cramér's V value was between the true state (perfect prediction) and the Glinsky et al. 2005 model with clinical data. The highest Cramér's V value was between our 17 gene/probe model and Singh et al. 2002 model, both with clinical data. Most of the models classified the same patients into the known groups (e.g. classifying a patient in the PSA control group as a PSA progression and a patient in the SYS case group as a systemic progression). They also tended to incorrectly classify the same patients (e.g., classifying a patient in the PSA control group as a systemic progression and vice versa). The 17 gene/probe model correctly classified 5-15 more patients into their known category (PSA controls or SYS cases) compared to the other models.
  • Secondary Analyses
  • Exploratory Survival Studies:
  • As noted above, the 17 gene/probe model and the previously reported models each classified some of the SYS cases in the good outcome category (e.g. to be PSA recurrences, not systemic progressors) and some of the PSA controls in the poor outcome category (e.g. to go on to systemic progression). There was a curiosity to see if these apparently false classifications had any biologic or clinical relevance.
  • Seventeen men in the PSA control group (who had both array and clinical model C data) went on to have systemic progression beyond 5 years at the time of last follow-up. Of these 17 patients, 9 were predicted to have a poor outcome by the 17 gene/probe model. Of the 179 patients who did not have any systemic progression, 38 were classified in the poor outcome category by the model (p value=0.0066, Fisher exact test). FIG. 3A illustrates the systemic progression-free survival for the good and poor outcome groups in the PSA controls. PSA controls whose tumor classified as having a poor outcome had significantly increased hazard of developing systemic progression beyond 5 years (log rank p-value=0.00050) (HR=4.7, 95% CI: 1.8-12.1).
  • Ninety-three men in the SYS case group (who also had array and clinical model C data) went on to prostate cancer death at the time of last follow-up. Of these 93 patients, 78 were predicted to have a poor outcome by the 17 gene/probe model. Of the 98 patients who did not suffer a prostate cancer death, 61 were classified in the poor outcome category by the model (p value=0.0008, chi-square test). FIG. 3B illustrates the prostate cancer-specific overall survival for the good and poor outcome groups in the SYS cases. SYS cases whose tumor classified as having a poor outcome had significantly increased hazard of suffering a prostate cancer-specific death (HR=2.5, 95% CI: 1.5-4.4). The median survival from first positive bone scan or CT was 2.8 years (95% CI: 2.4-4.2) in the group classified as having a poor outcome and 8.6 years (95% CI: 7.4-∞) in the group classified as having a good outcome (log rank p-value=0.00068).
  • Similar associations were observed when 3 of the previously published models with high AUCs (Lapointe et al. 2004 recurrence model and the Glinsky et al. 2005 and Yu et al. 2004 models) were evaluated. The following describes the results for the LaPointe et al. 2004 recurrence model (data for the other two models were similar and not shown). Of the 98 patients who did not suffer a prostate cancer death, 60 were predicted to have a poor outcome by the Lapointe et al. 2004 recurrence model (p value=0.0001, chi-square test). FIG. 3C illustrates the prostate cancer-specific overall survival for the good and poor outcome groups in the SYS cases. SYS cases whose tumor classified as having a poor outcome had significantly increased hazard of suffering a prostate cancer-specific death (HR=2.3, 95% CI: 1.3-4.2). The median survival from first positive bone scan or CT was 3.1 years (95% CI: 2.5-4.3) in the group classified as having a poor outcome and 8.6 years (95% CI: 8.3-∞) in the group classified as having a good outcome (log rank p-value=0.0033).
  • Exploratory 8q24 Studies:
  • Because of recent tumor chromosome dosage and germ line association studies, the custom array included 82 8q genes on the custom array. Fourteen 8q genes were within the top 68 genes upon univariate analysis (Table 2). Compared to the proportion of 8q gene on both arrays the prevalence of 8q genes is non random (p=0.003, Fisher exact test). Twelve additional 8q genes were within the top 126 genes. The prevalence of 26 8q genes in the top 126 is statistically significant (p=1.56×10-5, Fisher exact test). Chromosome band 8q24.1 has the greatest over-representation of genes in the top 68 gene and 126 gene lists (11 genes, p=6.35×10-7 and 19 genes, p=9.34×10-12, Fisher exact test). Of the 17 genes/probes in our final model, 5 map to 8q24 (p=0.0043, Fisher exact test)(see Table 3).
  • Exploratory ETS Transcription Factor Studies:
  • Alterations of several ETS-family oncogenes are associated with the development of prostate cancer (Tomilins et al., Science. 310, 644-648 (2005); Tomlins et al., Cancer Res. 66, 3396-3400 (2006); and Demichelis et al., Oncogene. 26:4596-4599 (2007)). Oligonucleotide probe sets for the three major members of the ETS family involved in prostate cancer were included: ERG, ETV1, and ETV4, as well as their translocation partner TMPRSS2. FIG. 4 summarizes the expression results for these genes for the SYS cases and the PSA and NED controls. Several observations can be made: 1) With only 8 exceptions ERG, ETV1 and ETV4 overexpression are mutually exclusive; e.g. the overexpression of each generally occurs in different tumors. 2) Different probe sets for ERG give nearly identical expression results (FIG. 8A). 3) The prevalence of ERG overexpression was 50.0%, 52.2% and 53.8% in the SYS cases, PSA controls and NED controls, respectively (using a cutoff of 3200 normalized fluorescence units—see FIG. 4). There is no significant difference in the mean expression and the prevalence of ERG overexpression between the three cohorts. 4) The prevalence of ETV1 overexpression was 11.5%, 6.5% and 5.1% in the SYS cases, PSA controls and NED controls, respectively (using the cutoff of 6000 normalized fluorescence units—see FIG. 4). The prevalence of ETV1 overexpression was significantly higher in SYS Cases (p=0.043, chi-square test). 5) The prevalence of ETV4 overexpression ranged from 2.5%-5.5% among the three groups and was not significantly different. 6) None of the genes were selected by the formal statistical modeling (see Table 3). In fact, the 17 gene/probe model predicted similar rates of progression in ERG+ and ERG− patients.
  • Exploratory Pathway Analysis:
  • The 461 genes from both cancer and custom panels that are potentially differentially expressed between SYS cases and PSA controls (p<0.05) were used as the focus genes for Ingenuity Pathway Analysis (IPA, Ingenuity Systems Inc., Redwood City, Calif.). IPA identified 101 canonical pathways that are associated with the focus genes, 51 of which are over-represented with p<0.05 (see Supplemental Table 5 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008). However, because a limited number of genes on both DASL panels was measured, the p values from IPA analysis may not accurately quantify the degree of over-representation of focus genes in each pathway.
  • Gene Set Enrichment Analysis (GSEA) (Subramanian et al., Proc Natl Acad Sci USA. 102, 15545-15550 (2005)) was then performed on chromosome 8 genes grouped by map location. Genes mapped to 8q24.1 had a significant p value (p=0.0002) with a FDR q value=0.001 (see Supplemental Table 6 of U.S. Provisional Patent Application No. 61/057,698, filed May 30, 2008).
  • It was concluded that the measurement of gene expression patterns may be useful for determining which men may benefit from additional therapy after PSA recurrence. These measurements should be included in prospective evaluation of various therapeutic interventions in this setting.
  • Other Embodiments
  • It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims (17)

What is claimed is:
1. A method for predicting whether or not a human, at the time of PSA reoccurrence or retropubic radial prostatectomy, will later develop systemic disease, wherein said method comprises:
(a) determining an expression profile score for cancer tissue from said human, wherein said expression profile score is based on at least the expression levels of RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 nucleic acid, and
(b) prognosing said human as later developing systemic disease or as not later developing systemic disease based on at least said expression profile score.
2. The method of claim 1, wherein said method is performed at the time of said PSA reoccurrence.
3. The method of claim 1, wherein said method is performed at the time of said retropubic radial prostatectomy.
4. The method of claim 1, wherein said expression levels are mRNA expression levels.
5. The method of claim 1, wherein said prognosing step (b) comprises prognosing said human as later developing systemic disease or as not later developing systemic disease based on at least said expression profile score and a clinical variable.
6. The method of claim 5, wherein said clinical variable is selected from the group consisting of a Gleason score and a revised Gleason score.
7. The method of claim 5, wherein said clinical variable is selected from the group consisting of a Gleason score, a revised Gleason score, age at surgery, initial PSA at recurrence, use of hormone or radiation therapy after radical retropubic prostatectomy, age at PSA recurrence, the second PSA level at time of PSA recurrence, and PSA slope.
8. The method of claim 1, wherein said method comprises prognosing said human as later developing systemic disease based on at least said expression profile score.
9. The method of claim 1, wherein said method comprises prognosing said human as not later developing systemic disease based on at least said expression profile score.
10. A method for predicting whether or not a human, at the time of systemic disease, will later die from prostate cancer, wherein said method comprises:
(a) determining an expression profile score for cancer tissue from said human, wherein said expression profile score is based on at least the expression levels of RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 nucleic acid, and
(b) prognosing said human as later dying of said prostate cancer or as not later dying of said prostate cancer based on at least said expression profile score.
11. The method of claim 10, wherein said expression levels are mRNA expression levels.
12. The method of claim 10, wherein said prognosing step (b) comprises prognosing said human as later developing systemic disease or as not later developing systemic disease based on at least said expression profile score and a clinical variable.
13. The method of claim 12, wherein said clinical variable is selected from the group consisting of a Gleason score and a revised Gleason score.
14. The method of claim 12, wherein said clinical variable is selected from the group consisting of a Gleason score, a revised Gleason score, age at surgery, initial PSA at recurrence, use of hormone or radiation therapy after radical retropubic prostatectomy, age at PSA recurrence, the second PSA level at time of PSA recurrence, and PSA slope.
15. The method of claim 10, wherein said method comprises prognosing said human as later dying of said prostate cancer based on at least said expression profile score.
16. The method of claim 10, wherein said method comprises prognosing said human as not later dying of said prostate cancer based on at least said expression profile score.
17. A method for (1) predicting whether or not a patient, at the time of PSA reoccurrence, will later develop systemic disease, (2) predicting whether or not a patient, at the time of retropubic radial prostatectomy, will later develop systemic disease, or (3) predicting whether or not a patient, at the time of systemic disease, will later die from prostate cancer, wherein said method comprises determining whether or not cancer tissue from said patient contains an RAD21, CDKN3, CCNB1, SEC14L1, BUB1, ALAS1, KIAA0196, TAF2, SFRP4, STIP1, CTHRC1, SLC44A1, IGFBP3, EDG7, FAM49B, C8orf53, and CDK10 expression profile indicative of a later development of said systemic disease or said death.
US14/857,658 2008-05-30 2015-09-17 Biomarker panels for predicting prostate cancer outcomes Active 2030-11-30 US10407731B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/857,658 US10407731B2 (en) 2008-05-30 2015-09-17 Biomarker panels for predicting prostate cancer outcomes

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US5769808P 2008-05-30 2008-05-30
US74787909A 2009-05-29 2009-05-29
US14/857,658 US10407731B2 (en) 2008-05-30 2015-09-17 Biomarker panels for predicting prostate cancer outcomes

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US12/474,879 Continuation US20090298082A1 (en) 2008-05-30 2009-05-29 Biomarker panels for predicting prostate cancer outcomes
US12747879 Continuation 2010-06-22

Publications (3)

Publication Number Publication Date
US20160010162A1 US20160010162A1 (en) 2016-01-14
US20180312926A9 true US20180312926A9 (en) 2018-11-01
US10407731B2 US10407731B2 (en) 2019-09-10

Family

ID=55067135

Family Applications (1)

Application Number Title Priority Date Filing Date
US14/857,658 Active 2030-11-30 US10407731B2 (en) 2008-05-30 2015-09-17 Biomarker panels for predicting prostate cancer outcomes

Country Status (1)

Country Link
US (1) US10407731B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11216742B2 (en) 2019-03-04 2022-01-04 Iocurrents, Inc. Data compression and communication using machine learning

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009143603A1 (en) 2008-05-28 2009-12-03 Genomedx Biosciences, Inc. Systems and methods for expression-based discrimination of distinct clinical disease states in prostate cancer
DK3435084T3 (en) 2012-08-16 2023-05-30 Mayo Found Medical Education & Res PROSTATE CANCER PROGNOSIS USING BIOMARKERS
CN110506127B (en) 2016-08-24 2024-01-12 维拉科特Sd公司 Use of genomic tags to predict responsiveness of prostate cancer patients to post-operative radiation therapy
US11208697B2 (en) 2017-01-20 2021-12-28 Decipher Biosciences, Inc. Molecular subtyping, prognosis, and treatment of bladder cancer
EP3593140A4 (en) 2017-03-09 2021-01-06 Decipher Biosciences, Inc. Subtyping prostate cancer to predict response to hormone therapy
CA3062716A1 (en) 2017-05-12 2018-11-15 Decipher Biosciences, Inc. Genetic signatures to predict prostate cancer metastasis and identify tumor agressiveness
KR20220008175A (en) * 2020-07-13 2022-01-20 가톨릭대학교 산학협력단 Stage-specific markers for diagnosing prognosis and determining treatment stratagies of patient of Prostate adenocarcinoma

Family Cites Families (240)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AT277936B (en) 1968-02-27 1970-01-12 Guenter Dipl Ing Knapp Method for the quantitative determination of iodine and thyroid hormones and device for carrying out the method
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US5130238A (en) 1988-06-24 1992-07-14 Cangene Corporation Enhanced nucleic acid amplification process
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5744101A (en) 1989-06-07 1998-04-28 Affymax Technologies N.V. Photolabile nucleoside protecting groups
CA2020958C (en) 1989-07-11 2005-01-11 Daniel L. Kacian Nucleic acid sequence amplification methods
US5494810A (en) 1990-05-03 1996-02-27 Cornell Research Foundation, Inc. Thermostable ligase-mediated DNA amplifications system for the detection of genetic disease
US5798257A (en) 1990-07-09 1998-08-25 Research Corporation Technologies, Inc. Nucleic acid encoding human MTS-1 protein
ATE244065T1 (en) 1990-12-06 2003-07-15 Affymetrix Inc METHODS AND REAGENTS FOR VERY LARGE SCALE IMMOBILIZED POLYMER SYNTHESIS
US5455166A (en) 1991-01-31 1995-10-03 Becton, Dickinson And Company Strand displacement amplification
US5994069A (en) 1996-01-24 1999-11-30 Third Wave Technologies, Inc. Detection of nucleic acids by multiple sequential invasive cleavages
US5846717A (en) 1996-01-24 1998-12-08 Third Wave Technologies, Inc. Detection of nucleic acid sequences by invader-directed cleavage
CA2124087C (en) 1991-11-22 2002-10-01 James L. Winkler Combinatorial strategies for polymer synthesis
US5384261A (en) 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US6027887A (en) 1992-03-11 2000-02-22 Institute Of Virology, Solvak Academy Of Sciences MN gene and protein
US6204370B1 (en) 1992-03-11 2001-03-20 Institute Of Virology, Slovak Academy Of Sciences MN gene and protein
US5541061A (en) 1992-04-29 1996-07-30 Affymax Technologies N.V. Methods for screening factorial chemical libraries
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5538848A (en) 1994-11-16 1996-07-23 Applied Biosystems Division, Perkin-Elmer Corp. Method for detecting nucleic acid amplification using self-quenching fluorescence probe
US5989815A (en) 1994-03-18 1999-11-23 University Of Utah Research Foundation Methods for detecting predisposition to cancer at the MTS gene
US7625697B2 (en) 1994-06-17 2009-12-01 The Board Of Trustees Of The Leland Stanford Junior University Methods for constructing subarrays and subarrays made thereby
US5830753A (en) 1994-09-30 1998-11-03 Ludwig Institute For Cancer Research Isolated nucleic acid molecules coding for tumor rejection antigen precursor dage and uses thereof.
US5710029A (en) 1995-06-07 1998-01-20 Gen-Probe Incorporated Methods for determining pre-amplification levels of a nucleic acid target sequence from post-amplification levels of product
US5705365A (en) 1995-06-07 1998-01-06 Gen-Probe Incorporated Kits for determining pre-amplification levels of a nucleic acid target sequence from post-amplification levels of product
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US5985557A (en) 1996-01-24 1999-11-16 Third Wave Technologies, Inc. Invasive cleavage of nucleic acids
DE69733629T2 (en) 1996-04-16 2006-05-18 Kishimoto, Tadamitsu, Tondabayashi METHOD FOR DETECTING SOLID CANCER CELLS AND HISTOLOGICAL HETEROTYPES AND METHOD FOR TESTING TISSUES FOR BONE MARROW TRANSPLANTATION AND TRANSPLANTATION OF PERIPHERAL BLOOD TEMM CELLS
US6136182A (en) 1996-06-07 2000-10-24 Immunivest Corporation Magnetic devices and sample chambers for examination and manipulation of cells
ES2331388T3 (en) 1997-03-07 2009-12-30 Siemens Healthcare Diagnostics Inc. SPECIFIC MARKER FOR PROSTATE CANCER.
JP3981416B2 (en) 1997-04-10 2007-09-26 ユニバーシティ メディカル センター ナイメーヘン PCA3 protein, PCA3 gene, and uses thereof
GB9711040D0 (en) 1997-05-29 1997-07-23 Duff Gordon W Prediction of inflammatory disease
US20020076735A1 (en) 1998-09-25 2002-06-20 Williams Lewis T. Diagnostic and therapeutic methods using molecules differentially expressed in cancer cells
US8101349B2 (en) 1997-12-23 2012-01-24 Novartis Vaccines And Diagnostics, Inc. Gene products differentially expressed in cancerous cells and their methods of use II
US6846912B1 (en) 1998-06-08 2005-01-25 Fuso Pharmaceutical Industries, Ltd. Antibody against LAR phosphatase subunit
US6406921B1 (en) 1998-07-14 2002-06-18 Zyomyx, Incorporated Protein arrays for high-throughput screening
US20060204989A1 (en) 1998-09-22 2006-09-14 Kopreski Michael S Comparative analysis of extracellular RNA species
US6468476B1 (en) 1998-10-27 2002-10-22 Rosetta Inpharmatics, Inc. Methods for using-co-regulated genesets to enhance detection and classification of gene expression patterns
ES2393056T3 (en) 1998-11-09 2012-12-18 Eiken Kagaku Kabushiki Kaisha Nucleic Acid Synthesis Procedure
US6436642B1 (en) 1999-04-20 2002-08-20 Curagen Corporation Method of classifying a thyroid carcinoma using differential gene expression
KR100527265B1 (en) 1999-09-13 2005-11-09 뉴젠 테크놀로지스 인코포레이티드 Methods and compositions for linear isothermal amplification of polynucleotide sequences
US7211390B2 (en) 1999-09-16 2007-05-01 454 Life Sciences Corporation Method of sequencing a nucleic acid
US7244559B2 (en) 1999-09-16 2007-07-17 454 Life Sciences Corporation Method of sequencing a nucleic acid
US6723506B2 (en) 2000-01-20 2004-04-20 Brigham And Women's Hospital Method of identifying PAX8-PPAR gamma-nucleic acid molecules
WO2001053836A2 (en) 2000-01-24 2001-07-26 Millennium Pharmaceuticals, Inc. Identification, assessment, prevention, and therapy of prostate cancer
US7700341B2 (en) 2000-02-03 2010-04-20 Dendreon Corporation Nucleic acid molecules encoding transmembrane serine proteases, the encoded proteins and methods based thereon
US7361488B2 (en) 2000-02-07 2008-04-22 Illumina, Inc. Nucleic acid detection methods using universal priming
WO2001060860A2 (en) 2000-02-17 2001-08-23 Millennium Predictive Medicine, Inc. Genes differentially expressed in human prostate cancer and their use
WO2001066753A2 (en) 2000-03-09 2001-09-13 Chiron Corporation Human genes and gene expression products
FR2810677B1 (en) 2000-06-27 2004-10-01 Urogene PROCESS FOR IN VITRO DIAGNOSIS OF PROSTATE CANCER AND IMPLEMENTATION KIT
US6673545B2 (en) 2000-07-28 2004-01-06 Incyte Corporation Prostate cancer markers
US20070037165A1 (en) 2000-09-08 2007-02-15 Applera Corporation Polymorphisms in known genes associated with human disease, methods of detection and uses thereof
US7892730B2 (en) 2000-12-22 2011-02-22 Sagres Discovery, Inc. Compositions and methods for cancer
US6988040B2 (en) 2001-01-11 2006-01-17 Affymetrix, Inc. System, method, and computer software for genotyping analysis and identification of allelic imbalance
US20040029114A1 (en) 2001-01-24 2004-02-12 Eos Technology, Inc. Methods of diagnosis of breast cancer, compositions and methods of screening for modulators of breast cancer
US20020169137A1 (en) 2001-02-09 2002-11-14 Active Pass Pharmaceuticals, Inc. Regulation of amyloid precursor protein expression by modification of ABC transporter expression or activity
US7026163B1 (en) 2001-02-23 2006-04-11 Mayo Foundation For Medical Education And Research Sulfotransferase sequence variants
AU2002252144A1 (en) 2001-02-27 2002-09-12 Case Western Reserve University Novel methods of diagnosis of metastatic colorectal cancer, compositions and methods of screening for modulators of metastatic colorectal cancer
US20030190602A1 (en) 2001-03-12 2003-10-09 Monogen, Inc. Cell-based detection and differentiation of disease states
US20040009481A1 (en) 2001-06-11 2004-01-15 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of human prostate cancer
US7229774B2 (en) 2001-08-02 2007-06-12 Regents Of The University Of Michigan Expression profile of prostate cancer
ATE420351T1 (en) 2001-08-31 2009-01-15 Gen Probe Inc PROBE WITH AFFINITY SHIFT FOR QUANTIFICATION OF ANALYTE POLYNUCLEOTIDES
US20040142325A1 (en) 2001-09-14 2004-07-22 Liat Mintz Methods and systems for annotating biomolecular sequences
US20050202442A1 (en) 2003-12-15 2005-09-15 Morris David W. Novel therapeutic targets in cancer
ES2339433T3 (en) 2001-12-07 2010-05-20 Novartis Vaccines And Diagnostics, Inc. INCREASE IN THE EXPRESSION OF RETROVIRUS ENDOGENO IN CANCER DE PROSTATA.
US6949342B2 (en) 2001-12-21 2005-09-27 Whitehead Institute For Biomedical Research Prostate cancer diagnosis and outcome prediction by expression analysis
US7704700B2 (en) 2002-02-12 2010-04-27 Burnham Institute For Medical Research Methods for determining the prognosis for patients with a prostate neoplastic condition using inhibitor of apoptosis polypeptides
US20030186248A1 (en) 2002-03-29 2003-10-02 Erlander Mark G. Interpreting cytological specimens via molecular histological signatures
US20030194734A1 (en) 2002-03-29 2003-10-16 Tim Jatkoe Selection of markers
US20050143933A1 (en) 2002-04-23 2005-06-30 James Minor Analyzing and correcting biological assay data using a signal allocation model
US20050032065A1 (en) 2002-06-24 2005-02-10 Afar Daniel E. H. Methods of prognosis of prostate cancer
US20040018493A1 (en) 2002-07-12 2004-01-29 Anastasio Alison E. Haplotypes of the CD3E gene
WO2004011625A2 (en) 2002-07-31 2004-02-05 University Of Southern California Polymorphisms for predicting disease and treatment outcome
JP4632788B2 (en) 2002-09-20 2011-02-16 ニュー・イングランド・バイオラブズ・インコーポレイティッド Helicase-dependent amplification of nucleic acids
US7300788B2 (en) 2002-10-08 2007-11-27 Affymetrix, Inc. Method for genotyping polymorphisms in humans
US20050266443A1 (en) 2002-10-11 2005-12-01 Thomas Jefferson University Novel tumor suppressor gene and compositions and methods for making and using the same
AU2003277612A1 (en) 2002-11-07 2004-06-07 Shigeo Kure Method of detecting gene mutation
ES2342665T3 (en) 2003-01-29 2010-07-12 454 Corporation SEQUENCING FROM TWO EXTREME.
DE602004017426D1 (en) 2003-02-20 2008-12-11 Genomic Health Inc USE OF INTRONIC RNA SEQUENCES FOR THE QUANTIFICATION OF GENE EXPRESSION
US7378233B2 (en) 2003-04-12 2008-05-27 The Johns Hopkins University BRAF mutation T1796A in thyroid cancers
WO2004092397A2 (en) 2003-04-15 2004-10-28 Novartis Ag Tmprss2 regulatory sequences and uses thereof
AU2004248120B2 (en) 2003-05-28 2009-04-23 Genomic Health, Inc. Gene expression markers for predicting response to chemotherapy
AU2004251256B2 (en) 2003-05-30 2009-05-28 The Board Of Trustees Of The University Of Illinois Gene expression profiles that identify genetically elite ungulate mammals
US20060019256A1 (en) 2003-06-09 2006-01-26 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
EP1644858B1 (en) 2003-07-10 2017-12-06 Genomic Health, Inc. Expression profile algorithm and test for cancer prognosis
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
US20050118625A1 (en) 2003-10-02 2005-06-02 Mounts William M. Nucleic acid arrays for detecting gene expression associated with human osteoarthritis and human proteases
WO2005040396A2 (en) 2003-10-16 2005-05-06 Genomic Health, Inc. qRT-PCR ASSAY SYSTEM FOR GENE EXPRESSION PROFILING
US20050130177A1 (en) 2003-12-12 2005-06-16 3M Innovative Properties Company Variable valve apparatus and methods
WO2005059108A2 (en) 2003-12-12 2005-06-30 Bayer Pharmaceuticals Corporation Gene expression profiles and methods of use
WO2005062770A2 (en) 2003-12-19 2005-07-14 Novakoff James L Method for conducting pharmacogenomics-based studies
US20090020433A1 (en) 2003-12-31 2009-01-22 Microfabrica Inc. Electrochemical Fabrication Methods for Producing Multilayer Structures Including the use of Diamond Machining in the Planarization of Deposits of Material
US7407755B2 (en) 2004-01-15 2008-08-05 Lubinski Jan Determining a predisposition to cancer
WO2005078139A2 (en) 2004-02-09 2005-08-25 Thomas Jefferson University DIAGNOSIS AND TREATMENT OF CANCERS WITH MicroRNA LOCATED IN OR NEAR CANCER-ASSOCIATED CHROMOSOMAL FEATURES
WO2005085471A2 (en) 2004-03-03 2005-09-15 Universität Leipzig Method and means for differential diagnosis of thyroid tumours
US8426126B2 (en) 2004-03-18 2013-04-23 Applied Biosystems, Llc Modified surfaces as solid supports for nucleic acid purification
WO2005095654A1 (en) 2004-03-25 2005-10-13 Biohelix Corporation Helicase-dependent amplification of circular nucleic acids
US7319011B2 (en) 2004-04-08 2008-01-15 Duke University Method for distinguishing follicular thyroid adenoma (FTA) from follicular thyroid carcinoma (FTC)
EP1756303B1 (en) 2004-04-09 2010-12-08 GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Diagnostic tool for diagnosing benign versus malignant thyroid lesions
EP2163650B1 (en) 2004-04-09 2015-08-05 Genomic Health, Inc. Gene expression markers for predicting response to chemotherapy
US20050240357A1 (en) 2004-04-26 2005-10-27 Minor James M Methods and systems for differential clustering
EP2471922A1 (en) 2004-05-28 2012-07-04 Asuragen, Inc. Methods and compositions involving microRNA
AU2005260113A1 (en) 2004-06-04 2006-01-12 Aviaradx, Inc. The importance of the gene HoxB13 for cancer
US20060019615A1 (en) 2004-07-24 2006-01-26 Ditmer Larry R Wireless keying for a continuous wave (CW) amateur radio transmitter
WO2006014013A1 (en) 2004-08-04 2006-02-09 Riken Bone/joint disease sensitivity gene and use thereof
CA2576912A1 (en) 2004-08-11 2006-02-23 The Cleveland Clinic Foundation Method of detecting thyroid cancer
US20060046253A1 (en) 2004-09-02 2006-03-02 Suntory Limited Method for analyzing genes of industrial yeasts
US7642348B2 (en) 2004-10-04 2010-01-05 Rosetta Genomics Ltd Prostate cancer-related nucleic acids
ITBZ20040048A1 (en) 2004-10-06 2005-01-06 Genprofiler METHOD FOR THE IDENTIFICATION OF NEOPLASTIC TRANSFORMATION, WITH PARTICULAR REFERENCE TO PROSTATIC CANCER
HUE039237T2 (en) 2004-10-06 2018-12-28 Mayo Found Medical Education & Res B7-h1 and pd-1 in treatment of renal cell carcinoma
US7629325B2 (en) 2004-10-11 2009-12-08 Technion Research & Development Foundation Ltd. Human Sef isoforms and methods of using same for cancer diagnosis and gene therapy
US7485468B2 (en) 2004-10-15 2009-02-03 Galapagos Bv Molecular targets and compounds, and methods to identify the same, useful in the treatment of joint degenerative and inflammatory diseases
EP1812601A2 (en) 2004-10-22 2007-08-01 Redpath Integrated Pathology, Inc. Dynamic genomic deletion expansion and formulation of molecular marker panels for integrated molecular pathology diagnosis and characterization of tissue, cellular fluid, and pure fluid specimens
CA2586201A1 (en) 2004-11-03 2006-05-11 Almac Diagnostics Limited Transcriptome microarray technology and methods of using the same
WO2006091776A2 (en) 2005-02-25 2006-08-31 The Brigham And Women's Hospital, Inc. Biomarkers for predicting prostate cancer progression
US20120122718A1 (en) 2005-03-01 2012-05-17 Reisman David N BRM Expression and Related Diagnostics
WO2006110264A2 (en) 2005-03-16 2006-10-19 Sidney Kimmel Cancer Center Methods and compositions for predicting death from cancer and prostate cancer survival using gene expression signatures
US20070037186A1 (en) 2005-05-20 2007-02-15 Yuqiu Jiang Thyroid fine needle aspiration molecular assay
US20070020657A1 (en) 2005-05-20 2007-01-25 Grebe Stefan K Methods for detecting circulating tumor cells
WO2006135596A2 (en) 2005-06-06 2006-12-21 The Regents Of The University Of Michigan Prognostic meta signatures and uses thereof
CA2612021A1 (en) 2005-06-13 2006-12-28 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
WO2006135886A2 (en) 2005-06-13 2006-12-21 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
IL177006A0 (en) 2005-08-02 2006-12-10 Veridex Llc Predicting bone relapse of breast cancer
AU2006280321A1 (en) 2005-08-15 2007-02-22 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
US9957569B2 (en) 2005-09-12 2018-05-01 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
CA2814598A1 (en) 2005-09-12 2007-03-22 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
KR20080063343A (en) 2005-09-22 2008-07-03 차이나 신테틱 러버 코포레이션 Gene expression profiling for identification of prognostic subclasses in nasopharyngeal carcinomas
US20070172844A1 (en) 2005-09-28 2007-07-26 University Of South Florida Individualized cancer treatments
US7962291B2 (en) 2005-09-30 2011-06-14 Affymetrix, Inc. Methods and computer software for detecting splice variants
EP1943356B1 (en) 2005-10-03 2014-05-14 Epigenomics AG Methods and nucleic acids for the analysis of gene expression associated with the prognosis of cell proliferative disorders
US7598052B2 (en) 2005-10-11 2009-10-06 The Regents Of The University Of Michigan Expression profile of thyroid cancer
US20070220621A1 (en) 2005-10-31 2007-09-20 Clarke Michael F Genetic characterization and prognostic significance of cancer stem cells in cancer
CA2628390A1 (en) 2005-11-02 2007-05-18 The Regents Of The University Of Michigan Molecular profiling of cancer
US20090215054A1 (en) 2005-12-13 2009-08-27 Carter Scott L Prognosis indicators for solid human tumors
ES2429404T3 (en) 2006-01-05 2013-11-14 The Ohio State University Research Foundation MicroRNA-based procedures for the diagnosis and prognosis of lung cancer
US20070172841A1 (en) 2006-01-25 2007-07-26 Hui Wang Probe/target stabilization with add-in oligo
FR2896881B1 (en) 2006-01-31 2008-04-18 Biomerieux Sa METHOD FOR DETERMINING PRONGF FOR IN VITRO DIAGNOSIS OF BREAST CANCER AND USE OF PRONGF IN THERAPY
US7670775B2 (en) 2006-02-15 2010-03-02 The Ohio State University Research Foundation Method for differentiating malignant from benign thyroid tissue
JP4867018B2 (en) 2006-03-22 2012-02-01 富士フイルム株式会社 Cancer detection method and suppression method
US7914988B1 (en) 2006-03-31 2011-03-29 Illumina, Inc. Gene expression profiles to predict relapse of prostate cancer
US20100279327A1 (en) 2006-06-12 2010-11-04 Bipar Sciences, Inc. Method of treating diseases with parp inhibitors
US20080076674A1 (en) 2006-07-06 2008-03-27 Thomas Litman Novel oligonucleotide compositions and probe sequences useful for detection and analysis of non coding RNAs associated with cancer
US20080028302A1 (en) 2006-07-31 2008-01-31 Steffen Meschkat Method and apparatus for incrementally updating a web page
FI20060751A0 (en) 2006-08-23 2006-08-23 Valtion Teknillinen Method of treating prostate cancer and screening of patients who benefit from said method
US8273539B2 (en) 2006-09-25 2012-09-25 Mayo Foundation For Medical Education And Research Extracellular and membrane-associated prostate cancer markers
EP2090665A2 (en) 2006-10-20 2009-08-19 Exiqon A/S Novel human microRNAs associated with cancer
US8338109B2 (en) 2006-11-02 2012-12-25 Mayo Foundation For Medical Education And Research Predicting cancer outcome
WO2008069906A2 (en) 2006-11-14 2008-06-12 The Regents Of The University Of California Digital expression of gene analysis
US8470534B2 (en) 2006-12-01 2013-06-25 Erik S. Knudsen Methods of predicting resistance or sensitivity to therapies for cancer
WO2008086478A2 (en) 2007-01-10 2008-07-17 Henry Ford Health System Methods and compositions for identification of prostate cancer markers
WO2008112283A2 (en) 2007-03-12 2008-09-18 Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Microrna profiling of androgen responsiveness for predicting the appropriate prostate cancer treatment
US8802599B2 (en) 2007-03-27 2014-08-12 Rosetta Genomics, Ltd. Gene expression signature for classification of tissue of origin of tumor samples
CA2678919A1 (en) 2007-03-27 2008-10-02 Ranit Aharonov Gene expression signature for classification of cancers
US20100273172A1 (en) 2007-03-27 2010-10-28 Rosetta Genomics Ltd. Micrornas expression signature for determination of tumors origin
US9096906B2 (en) 2007-03-27 2015-08-04 Rosetta Genomics Ltd. Gene expression signature for classification of tissue of origin of tumor samples
EP1975252A1 (en) 2007-03-29 2008-10-01 INSERM (Institut National de la Santé et de la Recherche Medicale) Methods for the prognosis or for the diagnosis of a thyroid disease
US20090062144A1 (en) 2007-04-03 2009-03-05 Nancy Lan Guo Gene signature for prognosis and diagnosis of lung cancer
US7901888B2 (en) 2007-05-09 2011-03-08 The Regents Of The University Of California Multigene diagnostic assay for malignant thyroid neoplasm
WO2009009432A2 (en) 2007-07-06 2009-01-15 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
WO2009020521A2 (en) 2007-08-03 2009-02-12 The Brigham And Women's Hospital, Inc. Identification and treatment of estrogen responsive prostate tumors
EP2650383A1 (en) 2007-08-03 2013-10-16 The Ohio State University Research Foundation Ultraconserved regions encoding ncRNAs
US9234244B2 (en) 2007-08-27 2016-01-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Diagnostic tool for diagnosing benign versus malignant thyroid lesions
AT505726A2 (en) 2007-08-30 2009-03-15 Arc Austrian Res Centers Gmbh SET OF TUMOR MARKERS
NZ562237A (en) 2007-10-05 2011-02-25 Pacific Edge Biotechnology Ltd Proliferation signature and prognosis for gastrointestinal cancer
US20090191535A1 (en) 2007-12-22 2009-07-30 Mark Carle Connelly Method of assessing metastatic carcinomas from circulating endothelial cells and disseminated tumor cells
CN101999000B (en) 2008-01-04 2014-04-09 国家科研中心 Molecular in vitro diagnosis of breast cancer
US8293880B2 (en) 2008-03-25 2012-10-23 University Of Southern California Prognostic panel for urinary bladder cancer
JP2011523049A (en) 2008-05-14 2011-08-04 ドナー, インコーポレイテッド Biomarkers for head and neck cancer identification, monitoring and treatment
WO2009143603A1 (en) 2008-05-28 2009-12-03 Genomedx Biosciences, Inc. Systems and methods for expression-based discrimination of distinct clinical disease states in prostate cancer
US20090298082A1 (en) 2008-05-30 2009-12-03 Klee George G Biomarker panels for predicting prostate cancer outcomes
US20110077168A1 (en) 2008-06-17 2011-03-31 Nitzan Rosenfeld Methods for distinguishing between specific types of lung cancers
AU2009269540A1 (en) 2008-07-07 2010-01-14 Decode Genetics Ehf Genetic variants predictive of cancer risk in humans
US20110212855A1 (en) 2008-08-15 2011-09-01 Decode Genetics Ehf. Genetic Variants Predictive of Cancer Risk
US20110251091A1 (en) 2008-09-12 2011-10-13 Cornell University Thyroid tumors identified
EP2347009A4 (en) 2008-10-14 2012-05-30 Caris Mpi Inc Gene and gene expressed protein targets depicting biomarker patterns and signature sets by tumor type
WO2010056931A1 (en) 2008-11-14 2010-05-20 Intelligent Oncotherapeutics, Inc. Methods for identification of tumor phenotype and treatment
US20110230372A1 (en) 2008-11-14 2011-09-22 Stc Unm Gene expression classifiers for relapse free survival and minimal residual disease improve risk classification and outcome prediction in pediatric b-precursor acute lymphoblastic leukemia
GB2477705B (en) 2008-11-17 2014-04-23 Veracyte Inc Methods and compositions of molecular profiling for disease diagnostics
US10236078B2 (en) 2008-11-17 2019-03-19 Veracyte, Inc. Methods for processing or analyzing a sample of thyroid tissue
US9495515B1 (en) 2009-12-09 2016-11-15 Veracyte, Inc. Algorithms for disease diagnostics
US8420329B2 (en) 2008-11-20 2013-04-16 Oncotherapy Science, Inc. Methods for diagnosing or treating prostate cancer
NZ593628A (en) 2008-11-26 2013-07-26 Decode Genetics Ehf Genetic variants useful for risk assessment of thyroid cancer using rs944289
WO2010061396A1 (en) 2008-11-30 2010-06-03 Rosetta Genomics Ltd. Methods for detecting an increased susceptibility to cancer
CA2745961A1 (en) 2008-12-04 2010-06-10 The Regents Of The University Of California Materials and methods for determining diagnosis and prognosis of prostate cancer
US20120015839A1 (en) 2009-01-09 2012-01-19 The Regents Of The University Of Michigan Recurrent gene fusions in cancer
US9040286B2 (en) 2009-02-03 2015-05-26 Children's Medical Center Corporation Diagnosis and treatment of cancer
US8697275B2 (en) 2009-03-04 2014-04-15 Samsung Sdi Co., Ltd. Rechargeable battery having an extendable case region
US9074258B2 (en) 2009-03-04 2015-07-07 Genomedx Biosciences Inc. Compositions and methods for classifying thyroid nodule disease
US9206482B2 (en) 2009-04-29 2015-12-08 Genomedx Biosciences Inc. Systems and methods for expression-based classification of thyroid tissue
EP2427575B1 (en) 2009-05-07 2018-01-24 Veracyte, Inc. Methods for diagnosis of thyroid conditions
WO2011023413A1 (en) 2009-08-31 2011-03-03 Alcedo Biotech Gmbh Microrna-based methods and compositions for reprogramming cells
US9110065B2 (en) 2009-09-21 2015-08-18 Paul Walfish Methods and compositions for the diagnosis and treatment of thyroid cancer
CA2776228A1 (en) 2009-10-01 2011-04-07 Chipdx Llc System and method for classification of patients
US8975019B2 (en) 2009-10-19 2015-03-10 University Of Massachusetts Deducing exon connectivity by RNA-templated DNA ligation/sequencing
US10446272B2 (en) 2009-12-09 2019-10-15 Veracyte, Inc. Methods and compositions for classification of samples
GB0922085D0 (en) 2009-12-17 2010-02-03 Cambridge Entpr Ltd Cancer diagnosis and treatment
US9642821B2 (en) 2010-01-14 2017-05-09 Dana-Farber Cancer Institute, Inc. MiR-182 in the diagnosis and treatment of cancer
EP2366800A1 (en) 2010-03-01 2011-09-21 Centrum Onkologii-Instytut im M. Sklodowskiej-Curie Oddzial w Gliwicach Kit, method and use for the diagnosis of papillary thyroid cancer using a gene expression profile
WO2011116380A2 (en) 2010-03-19 2011-09-22 H. Lee Moffitt Cancer Center And Research Institute, Inc. Hybrid model for the classification of carcinoma subtypes
WO2011133424A2 (en) 2010-04-20 2011-10-27 The Johns Hopkins University Genetic amplification of iqgap1 in cancer
CN103038635B (en) 2010-05-11 2016-12-28 威拉赛特公司 For diagnosing the method and composition of condition of illness
WO2011150453A1 (en) 2010-06-01 2011-12-08 The University Of Queensland Diagnostic, prognostic and therapeutic use of a long non-coding rna
US20130302808A1 (en) 2010-07-22 2013-11-14 Mayo Foundation For Medical Education And Research Predicting cancer outcome
WO2012031008A2 (en) 2010-08-31 2012-03-08 The General Hospital Corporation Cancer-related biological materials in microvesicles
WO2012068400A2 (en) 2010-11-17 2012-05-24 Asuragen, Inc. Mirnas as biomarkers for distinguishing benign from malignant thyroid neoplasms
EP3336200A1 (en) 2010-11-19 2018-06-20 The Regents Of The University Of Michigan Prostate cancer ncrna and uses thereof
US20130267443A1 (en) 2010-11-19 2013-10-10 The Regents Of The University Of Michigan ncRNA AND USES THEREOF
US20130273543A1 (en) 2010-12-21 2013-10-17 Decode Genetics Ehf. Genetic variants useful for risk assessment of thyroid cancer
US20130303826A1 (en) 2011-01-11 2013-11-14 University Health Network Prognostic signature for oral squamous cell carcinoma
EP2686443A4 (en) 2011-03-17 2014-12-17 Decode Genetics Ehf Genetic variants useful for risk assessment of thyroid cancer
US8945829B2 (en) 2011-03-22 2015-02-03 Cornell University Distinguishing benign and malignant indeterminate thyroid lesions
WO2012135008A1 (en) 2011-03-26 2012-10-04 Oregon Health And Science University Gene expression predictors of cancer prognosis
WO2013006495A2 (en) 2011-07-01 2013-01-10 Dana-Farber Cancer Institute, Inc. Methods of predicting prognosis in cancer
WO2013066678A1 (en) 2011-10-26 2013-05-10 Georgetown University Microrna expression profiling of thyroid cancer
WO2013063544A1 (en) 2011-10-27 2013-05-02 Asuragen, Inc. Mirnas as diagnostic biomarkers to distinguish benign from malignant thyroid tumors
WO2013086429A2 (en) 2011-12-09 2013-06-13 Veracyte, Inc. Methods and compositions for classification of samples
WO2013086524A1 (en) 2011-12-09 2013-06-13 The Johns Hopkins University Compositions and methods for characterizing thyroid neoplasia
US20130150257A1 (en) 2011-12-10 2013-06-13 Veracyte, Inc. Methods and compositions for sample identification
AU2012352153B2 (en) 2011-12-13 2018-07-26 Veracyte, Inc. Cancer diagnostics using non-coding transcripts
WO2013088457A1 (en) 2011-12-13 2013-06-20 Decode Genetics Ehf Genetic variants useful for risk assessment of thyroid cancer
US20130184999A1 (en) 2012-01-05 2013-07-18 Yan Ding Systems and methods for cancer-specific drug targets and biomarkers discovery
EP2825674A4 (en) 2012-03-15 2016-03-02 Qiagen Sciences Llc Thyroid cancer biomarker
WO2013169858A1 (en) 2012-05-08 2013-11-14 The Broad Institute, Inc. Diagnostic and treatment methods in patients having or at risk of developing resistance to cancer therapy
DK3435084T3 (en) 2012-08-16 2023-05-30 Mayo Found Medical Education & Res PROSTATE CANCER PROGNOSIS USING BIOMARKERS
EP2898125A4 (en) 2012-09-20 2016-05-25 Genomedx Biosciences Inc Thyroid cancer diagnostics
US20150275306A1 (en) 2012-10-10 2015-10-01 Stichting Het Nederlands Kanker Instituut Antoni van Leeuwenhoek Ziekenhuis Methods and means for predicting resistance to anti-cancer treatment
US20140121126A1 (en) 2012-10-25 2014-05-01 Memorial Sloan-Kettering Cancer Center Methods of detecting axl and gas6 in cancer patients
US20140143188A1 (en) 2012-11-16 2014-05-22 Genformatic, Llc Method of machine learning, employing bayesian latent class inference: combining multiple genomic feature detection algorithms to produce an integrated genomic feature set with specificity, sensitivity and accuracy
MX367366B (en) 2012-11-27 2019-08-16 Univ Pontificia Catolica Chile Compositions and methods for diagnosing thyroid tumors.
WO2014089241A2 (en) 2012-12-04 2014-06-12 Caris Mpi, Inc. Molecular profiling for cancer
EP2971084A1 (en) 2013-03-12 2016-01-20 Cepheid Methods of detecting cancer
BR112015022490A2 (en) 2013-03-15 2017-07-18 Veracyte Inc methods and compositions for sample classification
US20160032400A1 (en) 2013-03-15 2016-02-04 Fundació Institut De Recerca Biomèdica (Irb Barcelona) Method for the prognosis and treatment of cancer metastasis
CA2905620A1 (en) 2013-03-15 2014-10-02 Biotheranostics, Inc. Neuroendocrine tumors
JP2016518123A (en) 2013-04-17 2016-06-23 ライフ テクノロジーズ コーポレーション Gene fusions and gene variants associated with cancer
JP6533519B2 (en) 2013-05-10 2019-06-19 メモリアル スローン ケタリング キャンサー センター Lipid capture in RAS cancer
PL406033A1 (en) 2013-11-14 2015-05-25 Warszawski Uniwersytet Medyczny Method for diagnosing of thyroid papillous carcinoma, application of microRNA marker for diagnosing of the thyroid carcinoma, assessment of the disease progress and evaluation of patient and/or disease susceptibility to the proposed treatment and diagnostic set containing such markers
US9708667B2 (en) 2014-05-13 2017-07-18 Rosetta Genomics, Ltd. MiRNA expression signature in the classification of thyroid tumors
AU2016226253A1 (en) 2015-03-04 2017-09-21 Veracyte, Inc. Methods for assessing the risk of disease occurrence or recurrence using expression level and sequence variant information
US20180291459A1 (en) 2015-10-08 2018-10-11 Genomedx Biosciences, Inc. Use of a genetic signature diagnostically to evaluate treatment strategies for prostate cancer
EP3593140A4 (en) 2017-03-09 2021-01-06 Decipher Biosciences, Inc. Subtyping prostate cancer to predict response to hormone therapy

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11216742B2 (en) 2019-03-04 2022-01-04 Iocurrents, Inc. Data compression and communication using machine learning
US11468355B2 (en) 2019-03-04 2022-10-11 Iocurrents, Inc. Data compression and communication using machine learning

Also Published As

Publication number Publication date
US10407731B2 (en) 2019-09-10
US20160010162A1 (en) 2016-01-14

Similar Documents

Publication Publication Date Title
US20090298082A1 (en) Biomarker panels for predicting prostate cancer outcomes
US10407731B2 (en) Biomarker panels for predicting prostate cancer outcomes
US20200270707A1 (en) Methylation pattern analysis of haplotypes in tissues in a dna mixture
Nakagawa et al. A tissue biomarker panel predicting systemic progression after PSA recurrence post-definitive prostate cancer therapy
CN104781422B (en) Noninvasive measurement of fetal or tumor methylation patterns from plasma
Riester et al. Combination of a novel gene expression signature with a clinical nomogram improves the prediction of survival in high-risk bladder cancer
ES2741745T3 (en) Method to use gene expression to determine the prognosis of prostate cancer
JP2009528825A (en) Molecular analysis to predict recurrence of Dukes B colorectal cancer
JP2008521412A (en) Lung cancer prognosis judging means
KR20110111462A (en) Cancer biomarkers
US20160208333A1 (en) Colorectal cancer recurrence gene expression signature
US20110070582A1 (en) Gene Expression Profiling for Predicting the Response to Immunotherapy and/or the Survivability of Melanoma Subjects
US20090192045A1 (en) Molecular staging of stage ii and iii colon cancer and prognosis
Low et al. A formalin-fixed paraffin-embedded (FFPE)-based prognostic signature to predict metastasis in clinically low risk stage I/II microsatellite stable colorectal cancer
AU2017268510A1 (en) Method for using gene expression to determine prognosis of prostate cancer
US20150294062A1 (en) Method for Identifying a Target Molecular Profile Associated with a Target Cell Population
BR112020012280A2 (en) compositions and methods for diagnosing lung cancers using gene expression profiles
CN114507717A (en) Method for predicting bile duct cancer recurrence by combining multiple mRNAs and application thereof
JP2021515587A (en) Its use for molecular signature and identification of low-grade prostate cancer
US20170226592A1 (en) Methods and kits used in classifying adrenocortical carcinoma
US20220180974A1 (en) Colorectal cancer consensus molecular subtype classifier codesets and methods of use thereof
WO2017205963A1 (en) Mitochondrial pna prostate cancer marker and related systems and methods
Jonker et al. Epigenetic dysregulation in adrenocortical carcinoma, a systematic review of the literature
Da et al. Camila Lopes-Ramos MSc Ph. Da, Fernanda C. Koyama Ph. Da, b, Angelita Habr-Gama, MD, Ph. Dc, d, Anna Christina M. Salim Ph. Db, Fabiana Bettoni MSc Ph. Da, Paula F. Asprino Ph. Da, Gustavo S. França MSca, e, Joaquim Gama-Rodrigues, MD, Ph. Dc, d, Raphael B. Parmigiani Ph. Da, Rodrigo O. Perez MD

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: PETITION RELATED TO MAINTENANCE FEES GRANTED (ORIGINAL EVENT CODE: PTGR); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

AS Assignment

Owner name: CRG SERVICING LLC, TEXAS

Free format text: SECURITY INTEREST;ASSIGNOR:DECIPHER BIOSCIENCES, INC.;REEL/FRAME:048720/0129

Effective date: 20190327

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KLEE, GEORGE G.;JENKINS, ROBERT B.;KOLLMEYER, THOMAS M.;AND OTHERS;SIGNING DATES FROM 20090812 TO 20090814;REEL/FRAME:050999/0883

AS Assignment

Owner name: DECIPHER BIOSCIENCES, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:CRG SERVICING LLC;REEL/FRAME:055596/0345

Effective date: 20210312

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4