US20110251091A1 - Thyroid tumors identified - Google Patents

Thyroid tumors identified Download PDF

Info

Publication number
US20110251091A1
US20110251091A1 US13/063,429 US200913063429A US2011251091A1 US 20110251091 A1 US20110251091 A1 US 20110251091A1 US 200913063429 A US200913063429 A US 200913063429A US 2011251091 A1 US2011251091 A1 US 2011251091A1
Authority
US
United States
Prior art keywords
kit
differentially expressed
expression
genes
tumors
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.)
Abandoned
Application number
US13/063,429
Other languages
English (en)
Inventor
Thomas J. Fahey, III
Nimmi Arora
Theresa Seognamiglio
Raymond K. Blanchard
Guozhen Liu
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.)
Qiagen Gaithersburg LLC
Cornell University
Original Assignee
Cornell University
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 Cornell University filed Critical Cornell University
Priority to US13/063,429 priority Critical patent/US20110251091A1/en
Assigned to CORNELL UNIVERSITY reassignment CORNELL UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SEOGNAMIGLIO, THERESA, ARORA, NIMMI, FAHEY, THOMAS J.
Assigned to QIAGEN GAITHERSBURG, INC reassignment QIAGEN GAITHERSBURG, INC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BLANCHARD, RAYMOND K., LIU, GUOZHEN
Publication of US20110251091A1 publication Critical patent/US20110251091A1/en
Abandoned legal-status Critical Current

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
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/16Primer sets for multiplex assays

Definitions

  • the invention relates to detection and diagnosis of thyroid cancer.
  • the methods of the invention can be used to distinguish between benign thyroid cells or tissues, malignant thyroid cells or tissues, and follicular adenomas with nuclear atypia (FANA).
  • FANA nuclear atypia
  • Thyroid nodules are common in the United States, occurring in greater than 60% of individuals. Moreover, their incidence is steadily increasing, mainly because of the increased detection of smaller, asymptomatic nodules. Although the majority of these nodules are benign, a significant numbers of patients undergo surgical excision. Upon pathologic review of such thyroid tumors, clear-cut benign or malignant diagnoses often can be rendered. However, follicular lesions of the thyroid often pose a diagnostic challenge.
  • a particular diagnostic dilemma is presented in a subset of encapsulated follicular lesions with partial nuclear features of papillary thyroid carcinoma (PTC) (occasional nuclear grooves, focal nuclear clearing, and overlapping nuclei) and with histological features that fail to place them reliably in either the benign category or the malignant category.
  • PTC papillary thyroid carcinoma
  • these tumors represent approximately 10% of all follicular-patterned lesions observed at surgical pathology (see also, Arora et al. World J. Surg. 32:1237-1246 (2008)).
  • the invention relates to methods for improved diagnosis of thyroid cancer that can distinguish not only benign nodules from malignant thyroid tumors but can also identify borderline, pre-cancerous tumors (e.g., encapsulated follicular lesions that may have partial nuclear features of PTC) that may not need aggressive treatment.
  • pre-cancerous tumors e.g., encapsulated follicular lesions that may have partial nuclear features of PTC
  • the majority of histologically uncertain tumors (66.7%) were determined to be premalignant tumors, while a smaller number of tumors were determined to be benign tumors (26.7%) and only a even smaller number of tumors were actually malignant tumors (6.7%).
  • the malignant thyroid tumors can be identified with greater certainty, thereby avoiding unnecessary, expensive and invasive medical procedures that might otherwise have been used to treat histologically uncertain tumors.
  • one aspect of the invention is a method of detecting whether thyroid cancer cells are present in a test tissue or cell sample which comprises (a) observing test levels of RNA or protein expression in the test tissue or cell sample for any differentially expressed gene, and (b) comparing the test levels of expression to one or more standard or control levels of expression, to ascertain whether higher or lower levels of expression of any of the genes is present in the test tissue or cell sample, and thereby detecting whether thyroid cancer cells are present in the test tissue or cell sample; wherein the differentially expressed gene is selected from the group consisting of ANK2, ARHGAP6, C11orf17, CAPN3, CDH16, ChGn, CITED 1, CITED 2, CKB, COL9A3, CSRP2, DAPK2, DIO1, DPP4, DTX4, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GALNT7, GATM, HGD, HMGA2, IGFBP6, KIT, LRP4, MATN2, MET, MYH10, NA
  • the differentially expressed gene can be selected from the group consisting of DIO1, DTX4, GALNT7, HMGA2, IGFBP6, MET, PROS1, SDC4, SERPINA1, SLC4A4, TIAM1, TIMP1, UPP1 and a combination thereof.
  • the differentially expressed gene can be selected from the group consisting of ANK2, ARHGAP6, CDH16, CITED 1, CITED 2, COL9A3, ChGn, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GATM, KIT, LRP4, MATN2, SLIT1, SPTAN1, TFCP2L1, PIP3-E, PSD3, TNS3, TSPAN12, TIAM1 and a combination thereof.
  • the differentially expressed gene can be selected from the group consisting of C11orf17, CAPN3, CAPN3, CKB, CSRP2, DAPK2, DPP4, HGD, MYH10, NAUK2, PFAAP5, PGF, PKNOX2, PRKACB, QPCT, RAB27A, RXRG, and SLC25A15 and a combination thereof.
  • test levels of RNA expressed can be detected by microarray analysis or by nucleic acid amplification.
  • the test levels of RNA expressed are detected by microarray analysis that includes use of one or more probes on the microarray that can hybridize to one or more of the differentially expressed genes, or an RNA or DNA copy of the one or more differentially expressed genes.
  • such methods can employ one or more probes that can hybridize to any of SEQ ID NO:119-172.
  • the one or more probes hybridize to one or more of the differentially expressed genes, or an RNA or DNA copy of the one or more differentially expressed genes under moderate to highly stringent hybridization conditions.
  • the hybridization conditions can be highly stringent hybridization conditions.
  • nucleic acid amplification can be employed.
  • Such nucleic acid amplification can include reverse transcription polymerase chain reaction, real time polymerase chain reaction, or quantitative polymerase chain reaction.
  • the test levels of RNA expressed can be detected by nucleic acid amplification using one or more primers that hybridize to one or more of the differentially expressed genes, or an RNA or DNA copy of the one or more differentially expressed genes under moderate to highly stringent hybridization conditions.
  • the one or more primers employed can, for example, hybridize to any of SEQ ID NO:119-172.
  • Such hybridization conditions can in some instances be highly stringent hybridization conditions.
  • the one or more standard or control levels of expression can include: an expression level observed for a malignant thyroid cancer cell or tissue; an expression level observed for a benign thyroid cell or tissue; an expression level observed for a follicular adenoma with nuclear atypia; an expression level observed for a borderline thyroid cell or tissue; an expression level observed for a normal non-cancerous thyroid cell or tissue; or an expression level observed for a constitutively expressed gene.
  • these methods can distinguish between benign, malignant and borderline thyroid cells or tissues.
  • these methods can distinguish between benign thyroid cells or tissues, malignant thyroid cells or tissues, and follicular adenomas with nuclear atypia (FANA).
  • FANA nuclear atypia
  • the test tissue or cell sample is obtained from a patient with thyroid cancer or suspected of having thyroid cancer.
  • kits comprising: (a) at least one set of oligonucleotide primers, wherein a first primer in the set contains a sequence complementary to a region in one strand of a nucleic acid sequence template and primes the synthesis of a first extension product, and a second primer contains a sequence complementary to a region in said first extension product and primes the synthesis of a nucleic acid strand complementary to said first extension product, and wherein the template is a differentially expressed gene, or an RNA or DNA copy of the differentially expressed gene; and (b) instructions for using the at least one set of oligonucleotide primers; wherein the differentially expressed gene is selected from the group consisting of ANK2, ARHGAP6, C11orf17, CAPN3, CDH16, ChGn, CITED 1, CITED 2, CKB, COL9A3, CSRP2, DAPK2, DIO1, DPP4, DTX4, DUSP4, EFEMP1, ELMO1, FGFR
  • the differentially expressed gene can be selected from the group consisting of DIO1, DTX4, GALNT7, HMGA2, IGFBP6, MET, PROS1, SDC4, SERPINA1, SLC4A4, TIAM1, TIMP1, UPP1 and a combination thereof.
  • the differentially expressed gene can be selected from the group consisting of ANK2, ARHGAP6, CDH16, CITED 1, CITED 2, COL9A3, ChGn, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GATM, KIT, LRP4, MATN2, SLIT1, SPTAN1, TFCP2L1, PIP3-E, PSD3, TNS3, TSPAN12, TIAM1 and a combination thereof.
  • the differentially expressed gene can be selected from the group consisting of C11orf17, CAPN3, CAPN3, CKB, CSRP2, DAPK2, DPP4, HGD, MYH10, NAUK2, PFAAP5, PGF, PKNOX2, PRKACB, QPCT, RAB27A, RXRG, and SLC25A15 and a combination thereof.
  • the first primer and/or the second primer can include a label.
  • a container of nucleotides can also be included in the kit where the nucleotides are used as subunits in the synthesis of and amplified product.
  • the nucleotides can be ribonucleotides and/or deoxyribonucleotides.
  • One or more of such nucleotides can include a label.
  • the instructions can describe a method for amplifying an mRNA, cRNA or cDNA corresponding to the differentially expressed gene(s).
  • the first primer and/or the second primer may hybridize to an mRNA, cRNA or cDNA corresponding to the differentially expressed gene under moderate to highly stringent hybridization conditions.
  • the hybridization conditions can be highly stringent hybridization conditions in some instances.
  • kits that includes (a) a microarray with covalently attached probes that can hybridize to a differentially expressed gene selected from the group consisting of ANK2, ARHGAP6, C11orf17, CAPN3, CDH16, ChGn, CITED 1, CITED 2, CKB, COL9A3, CSRP2, DAPK2, DIO1, DPP4, DTX4, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GALNT7, GATM, HGD, HMGA2, IGFBP6, KIT, LRP4, MATN2, MET, MYH10, NAUK2, PFAAP5, PGF, PIP3-E, PKNOX2, PRKACB, PROS1, PSD3, QPCT, RAB27A, RXRG, SDC4, SERPINA1, SLC25A15, SLC4A4, SLIT1, SPTAN1, TFCP2L1, TIAM1, TIMP1, TNS3, TSPAN12, UPP1, and
  • the differentially expressed gene can be selected from the group consisting of DIO1, DTX4, GALNT7, HMGA2, IGFBP6, MET, PROS1, SDC4, SERPINA1, SLC4A4, TIAM1, TIMP1, UPP1 and a combination thereof.
  • the differentially expressed gene can be selected from the group consisting of ANK2, ARHGAP6, CDH16, CITED 1, CITED 2, COL9A3, ChGn, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GATM, KIT, LRP4, MATN2, SLIT1, SPTAN1, TFCP2L1, PIP3-E, PSD3, TNS3, TSPAN12, TIAM1 and a combination thereof.
  • the differentially expressed gene can be selected from the group consisting of C11orf17, CAPN3, CAPN3, CKB, CSRP2, DAPK2, DPP4, HGD, MYH10, NAUK2, PFAAP5, PGF, PKNOX2, PRKACB, QPCT, RAB27A, RXRG, and SLC25A15 and a combination thereof.
  • Such probes can, in some embodiments, hybridize to an mRNA, cRNA or cDNA corresponding to the differentially expressed gene, for example, under moderate to highly stringent hybridization conditions. In some embodiments, the hybridization conditions are highly stringent hybridization conditions.
  • Such a kit can also include one or more standard or control probes.
  • the kit can include one or more probes for a constitutively expressed gene.
  • Another aspect of the invention is a method of detecting a mutation in a human BRAF gene that includes: (a) obtaining a test sample of genomic DNA from a human; (b) amplifying a segment of BRAF DNA from the genomic DNA using primers with SEQ ID NO: 1 and SEQ ID NO:2; and (c) detecting whether the mutation exists in the segment amplified; wherein the mutation consists of a glutamate substituted for valine at codon 600.
  • Such a method can also include detecting or confirming whether the human has thyroid cancer by observing test levels of RNA or protein expression in the test tissue or cell sample for any of the differentially expressed genes described herein, using any of the methods and/or kits described herein.
  • FIG. 1 is an image of a histologically borderline lesion. Note the follicular architecture with focal nuclear clearing and occasional nuclear grooves.
  • FIG. 2 is a graphic generated by an unsupervised hierarchical cluster analysis.
  • FA indicates follicular adenoma
  • HYP hyperplastic lesion
  • BOR borderline tumor
  • FVPTC follicular variant of papillary thyroid carcinoma
  • PTC papillary thyroid carcinoma.
  • FIG. 3 is a graphic generated by a 2-group K-means cluster analysis.
  • FA indicates follicular adenoma
  • BOR borderline tumor
  • FVPTC follicular variant of papillary thyroid carcinoma
  • PTC papillary thyroid carcinoma
  • HYP hyperplastic lesion.
  • FIG. 4 is graphic generated by three-group K-means cluster analysis that identified 3 distinct groups of tumors based upon their gene expression patterns: malignant (left), benign (center), and intermediate (right).
  • BOR indicates borderline tumor; FVPTC, follicular variant of papillary thyroid carcinoma; PTC, papillary thyroid carcinoma; FA, follicular adenoma; HYP, hyperplastic lesion.
  • FIG. 5 shows a Venn diagram illustrating the differentially expressed genes relating 61 genes to benign, borderline, and malignant tumors.
  • FIG. 6 is a schematic diagram showing proposed gene expression changes during tumorigenesis of follicular-patterned lesions of the thyroid.
  • the invention relates to methods of detecting malignant thyroid tumors and/or distinguishing whether thyroid tumors are benign, malignant, and/or pre-cancerous borderline tumors. While currently available histological and/or cytological procedures can sometimes distinguish benign and malignant thyroid tumors, there are many thyroid tumors that cannot readily be classified as either malignant or benign by such histological procedures. Patients with such unclassified tumors are often aggressively treated as though their tumors were malignant. However, by employing the methods and kits described herein, these unclassified tumors can be properly identified as either benign, malignant, or pre-cancerous borderline tumors, thereby reducing the need for expensive, invasive and unpleasant medical treatment when it is unnecessary.
  • the application describes an analysis of fifty histologically-unequivocal benign and malignant tumors, which led to the identification of a list of sixty-one genes that are differentially expressed in benign and malignant thyroid tumors. These differentially expressed genes are listed in Table 1.
  • Twenty-seven genes were expressed differentially between the benign and borderline groups, including the cyclic AMP response element binding protein/p300-interactivator with glutamic acid/aspartic acid-rich carboxy-terminal domain 1 or CITED1 gene and the fibroblast growth factor receptor 2 or FGFR2 gene.
  • Fourteen genes were expressed differentially between the borderline group and malignant tumors, for example, the met proto-oncogene and of the high-mobility group adenine/thymine-hook 2 or HMGA2 gene in malignancies.
  • Mutations of the v-raf murine sarcoma viral oncogene homolog B1 or BRAF gene were identified in 4 of 14 malignant tumors but not in benign or borderline tumors.
  • the gene expression profiling methods described herein are more accurate than existing procedures for diagnosing problematic thyroid tumors.
  • the methods of the invention can identify malignant thyroid tumors with greater than 90% sensitivity and 80% specificity.
  • the methods of the invention can identify malignant thyroid tumors with greater than 93% sensitivity and greater than 82% specificity.
  • Genes are the units of heredity in living organisms. They are encoded in the organism's genetic material (DNA or RNA), and control the physical development and behavior of the organism. Genes encode the information necessary to construct the proteins (etc.) needed for the organism to function.
  • the term “genes” generally refers to the region of DNA (or RNA, in the case of some viruses) that determines the structure of a protein (the coding sequence), together with the region of DNA that controls when and where the protein will be produced (the regulatory sequences).
  • expression profiling refers to differential gene expression analysis/techniques. Examples of such techniques include microarray analyses, real time PCR and qPCR. Microarray technology allows for the comparison of gene expression between, for example, normal and diseased (e.g., cancerous) cells or cells which express different cell markers. There are several names for microarray technology including DNA microarrays, DNA arrays, DNA chips, gene chips, and others.
  • the expression levels of one or more of the genes listed in Table 1 can be detected using the methods and kits of the invention. In some embodiments, the expression levels of two or more, or three or more, or four or more, or five or more of the genes listed in Table 1 are detected to assess whether a thyroid nodule contains benign or malignant cancer cells. In other embodiments, the expression levels of seven or more, or eight or more, or ten or more, or twelve or more of the genes listed in Table 1 are detected to assess whether a thyroid nodule contains benign or malignant cancer cells. In further embodiments, the expression levels of no more than ten, no more than twelve, no more than fifteen, no more than twenty of the genes listed in Table 1 are detected to assess whether a thyroid nodule contains benign or malignant cancer cells.
  • Differential expression of these genes means that the mRNA or transcript levels produced by these genes increases or decreases in a test tissue or cell sample (e.g., a thyroid tissue biopsy) relative to a control, thereby indicating the presence of benign thyroid cells or tissues, malignant thyroid cells or tissues, and/or borderline tumors (e.g., encapsulated thyroid follicular lesions with partial nuclear features of PTC) in the test tissue or cell sample from which the RNA/transcripts were obtained.
  • a test tissue or cell sample e.g., a thyroid tissue biopsy
  • borderline tumors e.g., encapsulated thyroid follicular lesions with partial nuclear features of PTC
  • Genes whose expression changes in thyroid tumor cells include one or more of the following genes: ANK2, ARHGAP6, C11orf17, CAPN3, CDH16, ChGn, CITED 1, CITED 2, CKB, COL9A3, CSRP2, DAPK2, DIO1, DPP4, DTX4, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GALNT7, GATM, HGD, HMGA2, IGFBP6, KIT, LRP4, MATN2, MET, MYH10, NAUK2, PFAAP5, PGF, PIP3-E, PKNOX2, PRKACB, PROS1, PSD3, QPCT, RAB27A, RXRG, SDC4, SERPINA1, SLC4A4, SLC25A15, SLIT1, SPTAN1, TFCP2L1, TIAM1, TIMP1, TNS3, TSPAN12, UPP1, or a combination thereof.
  • the following genes were expressed at higher levels in malignant thyroid cancer tissues and cells than in benign thyroid lesions: CAPN3, CITED 1, DAPK2, DPP4, DUSP4, DTX4, GALNT7, HMGA2, IGFBP6, LRP4, MET, MYH10, PFAAP5, PROS1, PSD3, QPCT, RAB27A, RXRG, SERPINA1, SLIT1, SPTAN1, TIAM1, TIMP1, and UPP1.
  • detection of an increase in the expression of one or more of these genes in a tissue or cell sample, relative to a benign control tissue sample is indicative of thyroid cancer.
  • the following genes are expressed at lower levels in malignant thyroid cancer tissues than in benign thyroid lesions: ANK2, ARHGAP6, C11orf17, CDH16, CITED 2, COL9A3, ChGn, CKB, CSRP2, DIO1, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GATM, HGD, KIT, MATN2, NAUK2, PGF, PIP3-E, PKNOX2, PRKACB, SDC4, SLC4A4, SLC25A15, TFCP2L1, TNS3, and TSPAN12.
  • these borderline tumors can be distinguished from benign and malignant by their expression patterns.
  • the following genes are differentially expressed between malignant and borderline/benign tumors: DIO1, DTX4, GALNT7, HMGA2, IGFBP6, MET, PROS1, SDC4, SERPINA1, SLC4A4, TIAM1, TIMP1, and/or UPP1.
  • Each of these genes exhibit increased expression in malignant tumors relative to borderline and benign tumors, except DIO1, SDC4, and SLC4A4, which are expressed at lower levels in malignant thyroid tissues and cells when compared to their expression in benign and borderline tumors.
  • DIO1, SDC4, and SLC4A4 differential expression of one or more of these genes is detected in a thyroid test or cell sample, such differential expression is indicative of the presence of malignant tumor cells.
  • the following genes are differentially expressed between benign and borderline/malignant lesions: ANK2, ARHGAP6, CDH16, CITED 1, CITED 2, COL9A3, ChGn, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GATM, KIT, LRP4, MATN2, SLIT1, SPTAN1, TFCP2L1, PIP3-E, PSD3, TNS3, TSPAN12, and/or TIAM1.
  • Each of these genes exhibit decreased expression in malignant tumors relative to borderline/malignant tumors, except CITED 1, DUSP4, LRP4, PSD3, SLIT1, SPTAN1, and TIAM1, which are expressed at higher levels in malignant tissues and cells compared to borderline/malignant tissues and cells.
  • benign thyroid lesions can be identified and distinguished from borderline/malignant tumors by their differential expression patterns in a thyroid test tissue or cell sample.
  • the following genes are differentially expressed between benign and malignant lesions: C11orf17, CAPN3, CAPN3, CKB, CSRP2, DAPK2, DPP4, HGD, MYH10, NAUK2, PFAAP5, PGF, PKNOX2, PRKACB, QPCT, RAB27A, RXRG, and SLC25A15.
  • Each of these genes are expressed at higher levels in malignant thyroid tumors relative to their expression levels in benign thyroid lesions, except the following genes: C11orf17, CKB, CSRP2, HGD, PGF, PKNOX2, PRKACB, and SLC25A15, which are expressed at lower levels in malignant thyroid tissues relative to benign thyroid lesions.
  • the expression of these genes can be evaluated.
  • the difference in expression levels between a differentially expressed gene in malignant thyroid tissues relative to the expression levels for that gene in a control can be at least a 20% difference in expression levels, at least a 30% difference in expression levels, at least a 40% difference in expression levels, at least a 50% difference in expression levels, at least a 60% difference in expression levels, at least a 70% difference in expression levels, at least an 80% difference in expression levels, at least a 90% difference in expression levels, at least a 100% difference in expression levels, and/or a more than a 100% difference in expression levels.
  • the difference in expression levels between a differentially expressed gene in malignant thyroid tissues relative to the expression levels for that gene in a control can be at least 1.5 fold, at least 1.7 fold, at least 1.8 fold, at least 2-fold, at least 2.2 fold, at least at least 2.3 fold, at least 2.4 fold, at least 2.5 fold, or more than 2.5 fold.
  • Table 1 provides examples of the differences in expression levels that can readily be determined and observed.
  • Gene expression data may be gathered in any way that is available to one of skill in the art.
  • gene expression levels can be detected and quantified by employing an array of probes that hybridize to the different transcripts of one or more of the genes listed in Table 1, by using nucleic acid amplification (e.g., quantitative polymerase chain reaction) and through nucleic acid hybridization procedures.
  • nucleic acid amplification e.g., quantitative polymerase chain reaction
  • Other methods of determining expression of the genes include traditional Northern blotting, nuclease protection, RT-PCR and differential display methods can be used for detecting gene expression levels. Such methods are described in the following sections and in the Examples.
  • Probes and primers that can hybridize to an RNA, cDNA corresponding to any of the following genes can be used to detect differential gene expression: ANK2, ARHGAP6, C11orf17, CAPN3, CDH16, ChGn, CITED 1, CITED 2, CKB, COL9A3, CSRP2, DAPK2, DIO1, DPP4, DTX4, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GALNT7, GATM, HGD, HMGA2, IGFBP6, KIT, LRP4, MATN2, MET, MYH10, NAUK2, PFAAP5, PGF, PIP3-E, PKNOX2, PRKACB, PROS1, PSD3, QPCT, RAB27A, RXRG, SDC4, SERPINA1, SLC4A4, SLC25A15, SLIT1, SPTAN1, TFCP2L1, TIAM1, TIMP1, TNS3, TSPAN12, UPP1, or a combination thereof.
  • Sequences for these differentially expressed genes are available and can be used to make probes and primers for detecting expression levels. Examples of sequences that can be used to make probes and primers for these are provided hereinbelow. Any probe or primer that can hybridize to an RNA or cDNA of any of these genes can be used in the methods of the invention. In some embodiments, such a probe or primer hybridizes such to an RNA or cDNA of a differentially expressed gene under moderately stringent conditions. In other embodiments, such a probe or primer hybridizes such to an RNA or cDNA of a differentially expressed gene under highly stringent conditions. Such conditions are known to one of skill in the art and are described herein.
  • RNA sample or a nucleic acid sample derived from the mRNA transcript(s) refers to a nucleic acid ultimately synthesized from the mRNA transcript.
  • the original mRNA obtained from a test tissue or cell sample can serve as a template for generating a nucleic acid derived from an mRNA.
  • such a nucleic acid derived from an mRNA can be a cDNA reverse transcribed from an mRNA, an RNA transcribed from the cDNA, a DNA amplified from the cDNA, an RNA transcribed from the amplified DNA, and the like. Detection of such derived products is indicative of the presence and abundance of the original mRNA transcript in a test tissue or cell sample.
  • suitable samples include, but are not limited to, mRNA transcripts of the gene or genes, cDNA reverse transcribed from the mRNA, cRNA transcribed from the cDNA, and the like.
  • the concentration of the mRNA transcript(s) of the gene or genes is proportional to the transcription level of that gene.
  • the hybridization signal intensity can be proportional to the amount of hybridized nucleic acid.
  • the nucleic acid may be isolated from a test tissue or cell sample (and/or a control tissue sample) according to any of a number of methods well known to those of skill in the art.
  • RNA RNA
  • Methods of isolating total mRNA are well known to those of skill in the art. For example, methods of isolation and purification of nucleic acids are described in Sambrook et al. (1989) M OLECULAR C LONING : A L ABORATORY M ANUAL , Cold Spring Harbor Laboratory Press; and Sambrook et al. (2001).
  • Filter based methods for the isolation of mRNA are also available in the art and can be used for isolating mRNA from biological samples. Examples of commercially available filter-based RNA isolation systems include RNAqueousTM (Ambion) and RNeasyTM (Qiagen).
  • RNAqueousTM Ambion
  • RNeasyTM Qiagen
  • nucleic acid sample it is desirable to amplify the nucleic acid sample prior to evaluation. If a quantitative result is desired care can be taken to use an amplification method that maintains or controls for the relative frequencies of the amplified nucleic acids.
  • Quantitative amplification involves simultaneously co-amplifying a known quantity of an internal control nucleic acid. This provides an internal standard that may be used to calibrate the PCR reaction. Detection of the internal control sequence along with the mRNAs of interest (e.g., those from any of the genes in Table 1) allows one of skill in the art to monitor whether the mRNA isolation, purification and quantification procedures accurately reflect actual expression levels or whether there is a problem with any of these procedures (e.g., the mRNA has become degraded during one of the procedures).
  • Suitable amplification methods include, but are not limited to polymerase chain reaction (PCR) (Innis & Gelfand, Optimization of PCRs .
  • PCR PROTOCOLS A GUIDE TO METHODS AND APPLICATIONS (eds. M. A. Innis, et al.), pp. 3-12. Academic Press, San Diego (1990); ligase chain reaction (LCR) (see, Wu and Wallace, Genomics (1989)); Landegren, et al., Science 241: 1077-1080 (1988); Barringer, et al., Gene 89: 117-122 (1990); transcription amplification (Kwoh, et al., Proc. Natl. Acad. Sci. USA 86, 1173-1177 (1989)), and self-sustained sequence replication (Guatelli, et al., Proc. Natl. Acad. Sci. 87: 1874-1878 (1990)).
  • a nucleic acid sample is the total mRNA isolated from a biological sample (e.g., a test tissue or cell sample).
  • a biological sample refers to a sample obtained from an organism or from components (e.g., cells) of an organism, including normal tissue (e.g., as a control) and diseased tissue such as a tumor, a neoplasia or a hyperplasia.
  • the sample may be of any biological tissue or fluid or cells from any organism as well as cells raised in vitro, such as cell lines and tissue culture cells.
  • the biological sample may also be referred to as a “clinical sample” derived from a patient.
  • Such samples include, but are not limited to, tissue biopsy or fine needle aspiration biopsy samples, blood, blood cells (e.g., white cells), urine, peritoneal fluid, and pleural fluid, or cells therefrom.
  • Biological samples may also include sections of tissues such as frozen sections or formalin fixed sections taken for histological purposes.
  • the sample mRNA is reverse transcribed with a reverse transcriptase, such as SuperScript II (Invitrogen), and a primer consisting of an oligo-dT to generate first-strand cDNA.
  • a reverse transcriptase such as SuperScript II (Invitrogen)
  • a primer consisting of an oligo-dT to generate first-strand cDNA.
  • Other desirable sequences can be incorporated into the first-strand cDNA by linking those sequences onto the oligo-dT primer (e.g., a restriction site sequence, a sequence encoding a promoter such as a phage T7 promoter, etc.).
  • a second-strand DNA is polymerized in the presence of a DNA polymerase, DNA ligase, and RNase H.
  • the resulting double-stranded cDNA may be blunt-ended using T4 DNA polymerase and purified by phenol/chloroform extraction.
  • the double-stranded cDNA can then be then transcribed into cRNA or amplified to generate a pool of amplified cDNAs.
  • Methods for the in vitro transcription of RNA are known in the art and describe in, for example, Van Gelder, et al. (1990) and U.S. Pat. Nos. 5,545,522; 5,716,785; and 5,891,636, all of which are incorporated herein by reference.
  • a label may be incorporated into the cRNA or cDNA when it is transcribed.
  • Those of skill in the art are familiar with methods for labeling nucleic acids.
  • the cRNA may be transcribed in the presence of biotin-ribonucleotides or the cDNA may be synthesized in the presence of biotin-deoxyribonucleotides.
  • the BioArray High Yield RNA Transcript Labeling Kit (Enzo Diagnostics) is a commercially available kit for biotinylating cRNA.
  • the direct transcription method described above provides an antisense (aRNA) pool.
  • the antisense RNA can be the “target nucleic acid” that is hybridized to an array of the oligonucleotide probes provided in the microarray.
  • the oligonucleotide probes on the microarray are chosen to be complementary to subsequences of the antisense nucleic acids.
  • the target nucleic acid pool is a pool of sense nucleic acids
  • the oligonucleotide probes are selected to be complementary to subsequences of the sense nucleic acids.
  • the probes may be of either sense, or both senses, as the target nucleic acids include both sense and antisense strands.
  • nucleic acids may be advantageous to employ nucleic acids in combination with an appropriate detection means.
  • Recognition moieties incorporated into primers, incorporated into the amplified product during amplification, or attached to probes that can hybridize to the amplified product are useful in the identification of nucleic acid molecules.
  • a number of different labels may be used for this purpose including, but not limited to, fluorophores, chromophores, radiophores, enzymatic tags, antibodies, chemiluminescence, electroluminescence, and affinity labels.
  • fluorophores fluorophores, chromophores, radiophores, enzymatic tags, antibodies, chemiluminescence, electroluminescence, and affinity labels.
  • affinity labels include, but are not limited to the following: an antibody, an antibody fragment, a receptor protein, a hormone, biotin, Dinitrophenyl (DNP), or any polypeptide/protein molecule that binds to an affinity label.
  • enzyme tags include enzymes such as urease, alkaline phosphatase or peroxidase to mention a few.
  • Colorimetric indicator substrates can be employed to provide a detection means visible to the human eye or spectrophotometrically, to identify specific hybridization with complementary nucleic acid-containing samples.
  • fluorophores examples include, but are not limited to, Alexa 350, Alexa 430, AMCA, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, Cascade Blue, Cy2, Cy3, Cy5, 6-FAM, Fluoroscein, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rhodamine Green, Rhodamine Red, ROX, TAMRA, TET, Tetramethylrhodamine, and Texas Red.
  • radiolabels may be detected using photographic film or scintillation counters.
  • fluorescent markers may be detected using a photodetector to detect emitted light.
  • enzymatic labels are detected by providing the enzyme with a substrate and detecting the reaction product produced by the action of the enzyme on the substrate, and colorimetric labels are detected by simply visualizing the colored label or by use of spectrometer.
  • direct labels are detectable labels that are directly attached to or incorporated into the target (sample) nucleic acid prior to hybridization to a probe or microarray.
  • indirect labels are joined to the hybrid duplex after hybridization.
  • the indirect label is attached to a binding moiety that has been attached to the target nucleic acid prior to the hybridization.
  • the target nucleic acid may be biotinylated before the hybridization. After hybridization, an avidin-conjugated fluorophore will bind the biotin-bearing hybrid duplexes providing a label that is easily detected.
  • the present invention includes a method for detecting and/or quantifying expression of any combination of the genes listed in Table 1 (e.g., a target nucleic acid) in a biological sample.
  • Such detection and quantification methods can involve nucleic acid amplification (e.g., reverse transcription PCR, quantitative PCR and/or real-time PCR), wherein a sample containing a target nucleic acid that is to be amplified (e.g. a cDNA generated from an RNA sample by reverse transcription) is mixed with 1) primers that are complementary to sequences within the target sequence to be amplified, 2) a thermostable polymerase, and 3) four different nucleoside triphosphates. The normal steps of nucleic acid amplification are then followed—melting, annealing and synthesis—by thermal cycling of the mixture.
  • the primers employed can be linked to a label.
  • a fluorescent intercalating agent is used in the reaction. The labeled primers and/or fluorescent intercalating agents allow quantification of the amounts of amplified products generated in various test reactions.
  • RNA amplification When nucleic acid amplification is used to detect gene expression, any procedure that amplifies RNA can be used, for example, reverse transcription-polymerase chain reaction (RT-PCR) assays, strand displacement amplification and other amplification procedures. Strand displacement amplification can be used as described in Walker et al (1992) Nucl. Acids Res. 20, 1691-1696.
  • PCR polymerase chain reaction
  • the steps involved in PCR nucleic acid amplification method are described in more detail below.
  • the nucleic acid to be amplified is described as being double-stranded.
  • the process is readily adapted to amplify a single-stranded nucleic acid, such as an mRNA from any of the genes listed in Table 1.
  • the first step involves the synthesis of a complementary strand, for example, by reverse transcription so that two complementary target strands are available for amplification.
  • the PCR process for amplifying a target nucleic acid consists of introducing a large excess of the two primers to a mixture that may contain the mRNA (or cDNA generated therefrom) from any of the genes listed in Table 1, followed by a precise sequence of thermal cycling in the presence of a nucleic acid polymerase.
  • each of the two primers is complementary to a distinct region in one of the two strands of the double stranded target sequence. Primers are selected so that they hybridize just outside the region of interest to be amplified and so that, upon primer extension, one primer will be extended towards the hybridization site of a second primer hybridized on the opposite target strand.
  • the nucleic acid (RNA or cDNA) is denatured to open up double-stranded target sites and the temperature is lowered so that the primers anneal to their complementary sequences within the target molecule.
  • the primers are extended with a polymerase.
  • Such primer extension forms a new pair of complementary strands that likely have different ends than the original target.
  • Such complementary strands can hybridize together to form an “amplicon” that can also be a target for amplification.
  • the steps of denaturation, primer annealing and primer extension can be repeated many times. Each round of denaturation, annealing and extension constitutes one “cycle.” There can be numerous cycles, and the amount of amplified DNA produced increases with the number of cycles. Hence, to obtain a high concentration of an amplified target nucleic acid, many cycles are performed.
  • Each target nucleic acid strand is contacted with four different nucleoside triphosphates and one oligonucleotide primer, wherein each primer is selected to be substantially complementary to a portion the nucleic acid strand to be amplified (hmgn3), such that the extension product synthesized from one primer, when it is separated from its complement, can serve as a template for synthesis of the extension product of the other primer.
  • a selected primer-hybridization temperature is used that allows hybridization of each primer to a complementary nucleic acid strand.
  • the inhibitors of the invention can be added or included in this melting/annealing reaction.
  • a nucleic acid polymerase is used for primer extension.
  • the nucleic acid polymerase incorporates the nucleoside triphosphates into a growing nucleic acid strand to form a new strand that is complementary to the template strand hybridized by the primer.
  • this primer extension reaction is performed at a temperature and for a time effective to promote the activity of the nucleic acid enzyme and to synthesize a “full length” complementary nucleic acid strand that extends into and through a complete second primer binding site.
  • the temperature is not so high as to separate each extension product from its nucleic acid template strand.
  • the polymerase may be added after the first melting/annealing reaction.
  • step (c) The mixture from step (b) is then heated for a time and at a temperature sufficient to separate the primer extension products from their complementary templates.
  • the temperature chosen is not so high as to irreversibly denature the nucleic acid polymerase present in the mixture.
  • step (e) The mixture from step (d) is maintained at a temperature and for a time sufficient to promote primer extension by the polymerase to produce a “full length” extension product.
  • the temperature used is not so high as to separate each extension product from the complementary strand template. Steps (c)-(e) are repeated until the desired level of amplification is obtained.
  • real-time polymerase chain reaction (real time PCR; also called quantitative real time polymerase chain reaction (Q-PCR/qPCR) or kinetic polymerase chain reaction) is employed to quantify the expression of genes.
  • Real-time PCR amplifies and simultaneously quantifies a targeted nucleic acid (e.g., an RNA expressed by one of the genes listed in Table 1).
  • a targeted nucleic acid e.g., an RNA expressed by one of the genes listed in Table 1.
  • real-time PCR permits both detection and quantification (as absolute number of copies or relative amount when normalized to DNA input or additional normalizing genes) of a specific nucleic acid (e.g., RNA) in a sample.
  • Real-time PCR employs many of the same steps as polymerase chain reaction but the amplified DNA product is quantified as it accumulates in the reaction in real time after each amplification cycle.
  • Methods that are often used to quantify the amplified DNA include the use of fluorescent dyes intercalate with double-stranded DNA product, and the use of modified DNA primers that fluoresce when hybridized with a complementary nucleic acid template.
  • any of the SEQ ID NO:3-118 primers can be used in a real-time PCR assay for evaluating expression levels of the differentially expressed genes.
  • One type of real-time PCR assay that can be employed involves use of SYBRGreen dye.
  • SYBR Green is a dye that binds the minor groove of double stranded DNA. When SYBR Green dye binds to double stranded DNA, the intensity of the fluorescent emissions increases. As more double stranded amplicons are produced, SYBR Green dye signal will increase. During the PCR assay, such a fluorescent signal is directly proportional to the number of amplicons generated.
  • RNA expression levels To detect RNA expression levels, real-time polymerase chain reaction is combined with reverse transcription PCR, where the RNA in a sample is first treated with reverse transcriptase to generate a cDNA that can then be amplified.
  • Reverse transcription PCR and real-time PCR can be used to quantify relative levels of expression from any of the genes listed in Table 1.
  • the present invention therefore includes a method for detecting and/or quantifying expression of any of the genes listed in Table 1 (a target nucleic acid) that involves nucleic acid amplification (e.g., reverse transcription PCR and real-time PCR), wherein a sample containing a target nucleic acid that is to be amplified (e.g. a cDNA generated from an RNA sample by reverse transcription) is mixed with 1) primers that are complementary to sequences within the target sequence to be amplified, 2) a thermostable polymerase, and 3) four different nucleoside triphosphates.
  • the normal steps of nucleic acid amplification are then followed—melting, annealing and synthesis—by thermal cycling of the mixture.
  • the primers employed can be linked to a label.
  • a fluorescent intercalating agent is used in the reaction.
  • the labeled primers and/or fluorescent intercalating agents allow quantification of the amounts of amplified products generated in various test reactions.
  • Microarrays exploit the preferential binding of complementary nucleic acid sequences.
  • a microarray is typically a glass slide, on to which DNA molecules are attached at fixed locations (spots or features). There may be tens of thousands of spots on an array, each containing a huge number of identical DNA molecules (or fragments of identical molecules), of lengths from twenty to hundreds of nucleotides.
  • the spots on a microarray are either printed on the microarrays by a robot, or synthesized by photo-lithography (similar to computer chip productions) or by ink-jet printing. There are commercially available microarrays, however many labs produce their own microarrays.
  • Nucleic acid hybridization simply involves contacting a probe and target nucleic acid under conditions where the probe and its complementary target can form stable hybrid duplexes through complementary base pairing (see Lockhart et al., 1999, WO 99/32660, for example). The nucleic acids that do not form hybrid duplexes are then washed away leaving the hybridized nucleic acids to be detected, for example, through detection of an attached detectable label. It is generally recognized that nucleic acids are denatured by increasing the temperature and/or decreasing the salt concentration of the buffer containing the nucleic acids.
  • hybrid duplexes e.g., DNA-DNA, RNA-RNA or RNA-DNA
  • hybrid duplexes e.g., DNA-DNA, RNA-RNA or RNA-DNA
  • specificity of hybridization is reduced at lower stringency.
  • higher stringency e.g., higher temperature or lower salt
  • hybridization conditions may be selected to provide any degree of stringency. Stringency can also be increased by addition of agents such as formamide.
  • Hybridization specificity may be evaluated by comparison of hybridization to the test probes with hybridization to the various controls that can be present (e.g., expression level control, normalization control, mismatch controls, etc.).
  • the wash is performed at the highest stringency that produces consistent results and that provides a signal intensity greater than approximately 10% of the background intensity.
  • the hybridized sequences e.g., on a microarray
  • the hybridized sequences may be washed at successively higher stringency solutions and read between each wash. Analysis of the data sets thus produced will reveal a wash stringency above which the hybridization pattern is not appreciably altered and which provides adequate signal for the particular oligonucleotide probes of interest.
  • hybridize and “hybridization” refer to the annealing of a complementary sequence to the target nucleic acid, i.e., the ability of two polymers of nucleic acid (polynucleotides) containing complementary sequences to anneal through base pairing.
  • annealed and “hybridized” are used interchangeably throughout, and are intended to encompass any specific and reproducible interaction between a complementary sequence and a target nucleic acid, including binding of regions having only partial complementarity.
  • Certain bases not commonly found in natural nucleic acids may be included in the nucleic acids of the present invention and include, for example, inosine and 7-deazaguanine.
  • nucleic acid technology can determine duplex stability empirically considering a number of variables including, for example, the length of the complementary sequence, base composition and sequence of the oligonucleotide, ionic strength and incidence of mismatched base pairs.
  • the stability of a nucleic acid duplex is measured by the melting temperature, or “T m ”.
  • T m melting temperature
  • the T m of a particular nucleic acid duplex under specified conditions is the temperature at which on average half of the base pairs have disassociated.
  • stringency is used in reference to the conditions of temperature, ionic strength, and the presence of other compounds, under which nucleic acid hybridizations are conducted. With “high stringency” conditions, nucleic acid base pairing will occur only between nucleic acid fragments that have a high frequency of complementary base sequences. Thus, conditions of “medium” or “low” stringency are often required when it is desired that nucleic acids which are not completely complementary to one another be hybridized or annealed together. The art knows well that numerous equivalent conditions can be employed to comprise medium or low stringency conditions.
  • hybridization conditions are generally evident to one skilled in the art and is usually guided by the purpose of the hybridization, the type of hybridization (DNA-DNA or DNA-RNA), and the level of desired relatedness between the sequences (e.g., Sambrook et al. (1989); N UCLEIC A CID H YBRIDIZATION , A P RACTICAL A PPROACH , IRL Press, Washington D.C. 1985, for a general discussion of the methods).
  • hybridization stringency can be used to maximize or minimize stability of such duplexes.
  • Hybridization stringency can be altered by: adjusting the temperature of hybridization; adjusting the percentage of helix destabilizing agents, such as formamide, in the hybridization mix; and adjusting the temperature and/or salt concentration of the wash solutions.
  • the final stringency of hybridizations often is determined by the salt concentration and/or temperature used for the post-hybridization washes.
  • “High stringency conditions” when used in reference to nucleic acid hybridization include conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5 ⁇ SSPE (43.8 g/l NaCl, 6.9 g/l NaH 2 PO 4 H 2 O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5 ⁇ Denhardt's reagent and 100 ⁇ g/ml denatured salmon sperm DNA followed by washing in a solution comprising 0.1 ⁇ SSPE, 1.0% SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • “Medium stringency conditions” when used in reference to nucleic acid hybridization include conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5 ⁇ SSPE (43.8 g/l NaCl, 6.9 g/l NaH 2 PO 4 H 2 O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5 ⁇ Denhardt's reagent and 100 ⁇ g/ml denatured salmon sperm DNA followed by washing in a solution comprising 1.0 ⁇ SSPE, 1.0% SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • Low stringency conditions include conditions equivalent to binding or hybridization at 42EC in a solution consisting of 5 ⁇ SSPE (43.8 g/l NaCl, 6.9 g/l NaH 2 PO 4 H 2 O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5 ⁇ Denhardt's reagent [50 ⁇ Denhardt's contains per 500 ml: 5 g Ficoll (Type 400, Pharmacia), 5 g BSA (Fraction V; Sigma)] and 100 g/ml denatured salmon sperm DNA followed by washing in a solution comprising 5 ⁇ SSPE, 0.1% SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • 5 ⁇ SSPE 43.8 g/l NaCl, 6.9 g/l NaH 2 PO 4 H 2 O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH
  • 5 ⁇ Denhardt's reagent 50 ⁇ Denhard
  • homology refers to a degree of sequence identity. There may be partial homology or complete homology (i.e., identity). Homology is often measured using sequence analysis software (e.g., Sequence Analysis Software Package of the Genetics Computer Group. University of Wisconsin Biotechnology Center. 1710 University Avenue. Madison, Wis. 53705). Such software matches similar sequences by assigning degrees of homology to various substitutions, deletions, insertions, and other modifications.
  • sequence analysis software e.g., Sequence Analysis Software Package of the Genetics Computer Group. University of Wisconsin Biotechnology Center. 1710 University Avenue. Madison, Wis. 53705.
  • Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • low stringency or “low stringency conditions,” and non-limiting examples of low stringency include hybridization performed at about 0.15 M to about 0.9 M NaCl at a temperature range of about 20° C. to about 50° C.
  • hybridization performed at about 0.15 M to about 0.9 M NaCl at a temperature range of about 20° C. to about 50° C.
  • hybridization conditions selected also depend on the particular circumstances (depending, for example, on the G+C content, type of target nucleic acid, source of nucleic acid, and size of hybridization probe). Optimization of hybridization conditions for the particular application of interest is well known to those of skill in the art.
  • Representative solid phase hybridization methods are disclosed in U.S. Pat. Nos. 5,843,663, 5,900,481, and 5,919,626, which are incorporated herein by reference in their entireties.
  • Other methods of hybridization that may be used in the practice of the present invention are disclosed in U.S. Pat. Nos. 5,849,481, 5,849,486, and 5,851,772, which are also incorporated herein by reference in their entireties.
  • the hybridized nucleic acids are typically detected by detecting one or more labels attached to either the nucleic acids derived from a test sample (e.g., an amplified product) or to a probe that is hybridized to the mRNA or an amplified product of the mRNA.
  • the labels may be incorporated by any of a number of means well known to those of skill in the art (for example, see Affymetrix GeneChipTM Expression Analysis Technical Manual).
  • DNA arrays and gene chip technology provide a means of rapidly screening a large number of nucleic acid samples for their ability to hybridize to a variety of single stranded DNA probes immobilized on a solid substrate. These techniques involve quantitative methods for analyzing large numbers of genes rapidly and accurately.
  • the technology capitalizes on the complementary binding properties of single stranded DNA to screen nucleic acid samples by hybridization (Pease et al., 1994; Fodor et al., 1991).
  • a DNA array or gene chip consists of a solid substrate with an attached array of single-stranded DNA molecules.
  • the chip or array is contacted with a single stranded nucleic acid sample (e.g., cRNA or cDNA), which is allowed to hybridize under stringent conditions.
  • a single stranded nucleic acid sample e.g., cRNA or cDNA
  • the chip or array is then scanned to determine which probes have hybridized.
  • Exemplary methods include: the immobilization of biotinylated nucleic acid molecules to avidin/streptavidin coated supports (Holmstrom, 1993), the direct covalent attachment of short, 5′-phosphorylated primers to chemically modified polystyrene plates (Rasmussen et al., 1991), or the precoating of the polystyrene or glass solid phases with poly-L-Lys or poly L-Lys, Phe, followed by the covalent attachment of either amino- or sulfhydryl-modified oligonucleotides using bifunctional crosslinking reagents (Running et al., 1990; Newton et al., 1993). When immobilized onto a substrate, the probes are stabilized and therefore may be used repeatedly.
  • hybridization is performed on an immobilized nucleic acid target or a probe molecule that is attached to a solid surface such as nitrocellulose, nylon membrane or glass.
  • a solid surface such as nitrocellulose, nylon membrane or glass.
  • matrix materials including reinforced nitrocellulose membrane, activated quartz, activated glass, polyvinylidene difluoride (PVDF) membrane, polystyrene substrates, polyacrylamide-based substrate, other polymers such as poly(vinyl chloride), poly(methyl methacrylate), poly(dimethyl siloxane), photopolymers (which contain photoreactive species such as nitrenes, carbenes and ketyl radicals capable of forming covalent links with target molecules).
  • PVDF polyvinylidene difluoride
  • PVDF polystyrene substrates
  • polyacrylamide-based substrate other polymers such as poly(vinyl chloride), poly(methyl methacrylate), poly(dimethyl siloxane), photopolymers (which contain photoreactive
  • the Affymetrix GeneChip system may be used for hybridization and evaluation of the probe arrays, where the probes have been selected to hybridize to any combination of the genes listed in Table 1 (or a cRNA or cDNA obtained from an mRNA generated by any of those genes).
  • the Affymetrix U95A or U133A array is used in conjunction with Microarray Suite 5.0 for data acquisition and preliminary analysis of gene expression patterns and/or levels.
  • Normalization controls are oligonucleotide probes that are complementary to labeled reference oligonucleotides that are added to the nucleic acid sample.
  • the signals obtained from the normalization controls after hybridization provide a control for variations in hybridization conditions, label intensity, “reading” efficiency and other factors that may cause the hybridization signal to vary between arrays. For example, signals read from all other probes in the array can be divided by the signal from the control probes thereby normalizing the measurements.
  • Virtually any probe may serve as a normalization control.
  • Preferred normalization probes are selected to reflect the average length of the other probes present in the array, however, they can be selected to cover a range of lengths.
  • the normalization control(s) can also be selected to reflect the (average) base composition of the other probes in the array, however in a preferred embodiment, only one or a few normalization probes are used and they are selected such that they hybridize well (i.e. no secondary structure) and do not match any target-specific probes. Normalization probes can be localized at any position in the array or at multiple positions throughout the array to control for spatial variation in hybridization efficiently.
  • a standard probe cocktail supplied by Affymetrix is added to the hybridization to control for hybridization efficiency when using Affymetrix Gene Chip arrays.
  • Expression level controls are probes that hybridize specifically with constitutively expressed genes in the sample.
  • the expression level controls can be used to evaluate the efficiency of cRNA preparation.
  • Virtually any constitutively expressed gene provides a suitable target for expression level controls.
  • expression level control probes have sequences complementary to subsequences of constitutively expressed “housekeeping genes.”
  • the ratio of the signal obtained for a 3′ expression level control probe and a 5′ expression level control probe that specifically hybridize to a particular housekeeping gene is used as an indicator of the efficiency of cRNA preparation.
  • a ratio of 1-3 indicates an acceptable preparation.
  • Any appropriate computer platform may be used to perform the necessary comparisons between sequence information, gene expression information and any other information in a database or provided as an input.
  • a large number of computer workstations and programs are available from a variety of manufacturers, such as those available from Affymetrix.
  • SAM Significance Analysis of Microarrays
  • FDR false discovery rate
  • kits generally include probes and/or primers for detecting and/or quantifying expression of the differnentially expressed genes described herein, and instructions for performing the detection and/or quantification methods.
  • one aspect of the invention is a kit that includes, for example, (a) at least one set of oligonucleotide primers, wherein a first primer in the set contains a sequence complementary to a region in one strand of a nucleic acid sequence template and primes the synthesis of a first extension product, and a second primer contains a sequence complementary to a region in said first extension product and primes the synthesis of a nucleic acid strand complementary to said first extension product, and wherein the template is a differentially expressed gene, or an RNA or DNA copy of the differentially expressed gene; and (b) instructions for using the at least one set of oligonucleotide primers; wherein differentially expressed gene is selected from the group consisting of ANK2, ARHGAP6, C11orf17, CAPN3, CDH16, ChGn, CITED 1, CITED 2, CKB, COL9A3, CSRP2, DAPK2, DIO1, DPP4, DTX4, DUSP4, EFEMP1, ELMO
  • the first primer and/or the second primer provided in the kit can have a covalently attached label.
  • the first primer and/or the second primer can be selected from any of SEQ ID NO:3-118.
  • kits that can be made and/or used for detecting differential expression can include (a) a microarray with covalently attached probes that can hybridize to a differentially expressed gene selected from the group consisting of ANK2, ARHGAP6, C11orf17, CAPN3, CDH16, ChGn, CITED 1, CITED 2, CKB, COL9A3, CSRP2, DAPK2, DIO1, DPP4, DTX4, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GALNT7, GATM, HGD, HMGA2, IGFBP6, KIT, LRP4, MATN2, MET, MYH10, NAUK2, PFAAP5, PGF, PIP3-E, PKNOX2, PRKACB, PROS1, PSD3, PSD3, QPCT, RAB27A, RXRG, SDC4, SERPINA1, SLC25A15, SLC4A4, SLIT1, SPTAN1, TFCP2L1, TIAM1, TIMP1, TNS3, TSPA
  • Probes useful in the microarray of this kit can hybridize to any of ANK2, ARHGAP6, C11orf17, CAPN3, CDH16, ChGn, CITED 1, CITED 2, CKB, COL9A3, CSRP2, DAPK2, DIO1, DPP4, DTX4, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GALNT7, GATM, HGD, HMGA2, IGFBP6, KIT, LRP4, MATN2, MET, MYH10, NAUK2, PFAAP5, PGF, PIP3-E, PKNOX2, PRKACB, PROS1, PSD3, QPCT, RAB27A, RXRG, SDC4, SERPINA1, SLC25A15, SLC4A4, SLIT1, SPTAN1, TFCP2L1, TIAM1, TIMP1, TNS3, TSPAN12, UPP1, or a combination thereof
  • the kit can include other useful components.
  • the kit can include a container of nucleotides for use as subunits in the synthesis of and amplified product.
  • the one or more nucleotides provided can have a covalently attached label.
  • the nucleotides provided with the kit can be ribonucleotides or deoxyribonucleotides.
  • Other components provided by the kit include reagents or devices for isolating and/or purifying mRNA, enzymes such as reverse transcriptase, ligase, DNA polymerase (e.g., Taq polymerase), solutions and buffers for performing enzymatic reaction, and/or solutions for performing hybridization.
  • kits can include one or more buffers, such as a DNA isolation buffers, an amplification buffer or a hybridization buffer.
  • the kit may also contain compounds and reagents to prepare DNA templates and isolate RNA from a test sample.
  • the kit may also include various labeling reagents and compounds.
  • the kit of can also include one or more standard or control probes.
  • one or more of the standard or control probes can be a probe or probes for one or more constitutively expressed genes.
  • the instructions provided with the kit can describe a method for amplifying an mRNA, cRNA or cDNA corresponding to the differentially expressed gene(s).
  • One of skill in the art may choose to utilize the kit for detecting differential expression by hybridization of a first primer and/or a second primer to an mRNA, cRNA or cDNA corresponding to the differentially expressed gene under moderate to highly stringent hybridization conditions.
  • the kit with the microarray one of skill in the art may choose to utilize the kit for detecting differential expression by hybridization of a probe to an mRNA, cRNA or cDNA corresponding to the differentially expressed gene under moderate to highly stringent hybridization conditions.
  • the instructions provided in the kit can inform one of skill in the art to employ hybridization conditions that are moderately to highly stringent hybridization conditions.
  • the kit can include primers and/or probes for detecting some or all of the differentially expressed genes.
  • the kits can detect and/or quantify expression of a subset of differentially expressed genes such as any one of DIO1, DTX4, GALNT7, HMGA2, IGFBP6, MET, PROS1, SDC4, SERPINA1, SLC4A4, TIAM1, TIMP1, UPP1 or a combination thereof.
  • kits can detect and/or quantify expression of other subsets of differentially expressed genes, for example, any one of ANK2, ARHGAP6, CDH16, CITED 1, CITED 2, COL9A3, ChGn, DUSP4, EFEMP1, ELMO1, FGFR2, FLRT1, FMOD, GATM, KIT, LRP4, MATN2, SLIT1, SPTAN1, TFCP2L1, PIP3-E, PSD3, TNS3, TSPAN12, TIAM1 or a combination thereof.
  • kits can be used to detect another subset of differentially expressed genes such as one or more of the following genes C11orf17, CAPN3, CAPN3, CKB, CSRP2, DAPK2, DPP4, HGD, MYH10, NAUK2, PFAAP5, PGF, PKNOX2, PRKACB, QPCT, RAB27A, RXRG, and SLC25A15 or a combination thereof.
  • probes and/or primers for detecting mRNA expression of any of the genes listed in Table 1 may be included in a kit.
  • the kit may further include individual nucleic acids that can be amplified with the nucleic acids of interest.
  • the kit can also include probes and/or primers for detecting particular control nucleic acid sequences.
  • the control nucleic acids included in the kit can be mRNA(s) and/or control cDNA(s).
  • the probes, primers and/or control RNA and/or DNA sequences can be provided on a microarray. Alternatively, the probes, primers and/or control RNA and/or DNA sequences can be provided in separate vials or wells of an assay plate (e.g., a microtiter plate).
  • kits may be packaged either in aqueous media or in lyophilized form.
  • the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred.
  • the components of the kit may also be provided as dried powder(s).
  • the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container or by the user.
  • the containers for the kits can include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and/or suitably aliquoted.
  • a labeling reagent and label may be included and packaged separately or together.
  • the kit can also contain a second, third or other additional container into which the additional components may be separately placed.
  • various combinations of components may be included together in a vial.
  • the kits of the present invention can also include a means for containing the nucleic acids, and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained.
  • kits will also include instructions for employing the kit components as well the use of any other reagent not included in the kit. Instructions may include variations that can be implemented.
  • Tissue samples were collected at time of surgery, snap-frozen in liquid nitrogen, and stored at ⁇ 80° C. Representative slides for all tumors were reviewed by two dedicated pathologists. A total of 90 thyroid tumor samples, including 16 papillary thyroid carcinoma (PTC), 22 follicular variants of papillary thyroid carcinoma (FVPTC), 15 hyperplastic nodules, 22 follicular adenomas, and 15 histologically borderline tumors were analyzed in this study. This study was approved by our Institutional Review Board.
  • PTC papillary thyroid carcinoma
  • FVPTC papillary thyroid carcinoma
  • hyperplastic nodules 22 follicular adenomas
  • histologically borderline tumors were analyzed in this study. This study was approved by our Institutional Review Board.
  • Borderline tumors were defined as encapsulated lesions with follicular architecture in which the morphologic features of papillary thyroid carcinoma were qualitatively incomplete and the lesions did not demonstrate evidence of capsular and/or vascular invasion.
  • the incomplete features of papillary thyroid carcinoma were widespread in the lesions that were analyzed in this study and did not represent focal findings in an otherwise benign nodule.
  • An example of such a borderline tumor sample is shown in FIG. 1 .
  • these cases could be classified as well differentiated tumors of uncertain malignant potential (WDT-UMP) as proposed by Williams et al. ( Int J Surg Pathol. 8:181-183 (2000)).
  • WDT-UMP uncertain malignant potential
  • ArrayAssist 5.2.2 (Stratagene, Inc., La Jolla, Calif.) was used for gene chip analysis. Interchip and intrachip normalization was performed via robust multichip analysis (RMA). After RMA, hybridization signals underwent variance stabilization, log transformation, and baseline transformation. Advanced significance analysis was performed on 50 U95A GeneChips including 10 hyperplastic nodules, 16 follicular adenomas, 13 follicular variants of papillary thyroid carcinomas (FVPTCs), and 11 papillary thyroid carcinomas. This formed the training set. Gene expression of benign tumors was compared to that of malignant tumors. After Benjamini-Hochberg correction for false-discovery, gene probe sets with significant differential expression (2-fold or more with p ⁇ 0.05) were identified.
  • RMA robust multichip analysis
  • This probe list was then converted to correspond to probes on the U133A Gene Chips (array comparison software; available from Affymetrix.com).
  • the test set was then assessed using unsupervised hierarchical cluster analysis and K-means cluster analysis with both 2- and 3-group cluster algorithms. Genes that were differentially expressed between borderline tumors and classic benign and malignant tumors were further identified with advanced significance analysis.
  • Amplification was carried out for 35 cycles (at 94° C. for 15 seconds, at 60° C. for 1 minute, and at 72° C. for 1 minute). All PCR products were visualized by electrophoresis on a 2% agarose gel and purified using a PCR purification kit (Qiagen Inc). BRAF mutations were detected by direct sequencing of PCR products. All sequencing was performed bidirectionally using the Big Dye Terminator cycle-sequencing kit and the Applied Biosystems Automated 3730 DNA Analyzer (Applied Biosystems, Foster City, Calif.).
  • This Example illustrates that gene expression analysis can be used to identify whether tumors of uncertain malignancy are benign or malignant. Based on their benign clinical behavior, it is proposed that these encapsulated thyroid follicular lesions with partial nuclear features of papillary thyroid carcinoma be called ‘follicular adenomas with nuclear atypia’ and the data indicate that these lesions may not need to be treated as cancers.
  • the training set consisted of 50 tumors including 26 unequivocal benign tumors (16 follicular adenoma and 10 hyperplastic nodules) and 24 unequivocal malignancies (11 PTC and 13 FVPTC).
  • a total of 66 probe sets corresponding to 56 genes showed significant differential expression between benign and malignant tumors. Thirty-one genes had up-regulated expression in malignancies compared to benign tumors, and 30 genes were down-regulated (Table 1).
  • This tumor group involved 15 tumors, where the vast majority (10 cases) were histologically borderline tumors. Three FVPTCs and 2 follicular adenomas were also identified in this borderline group of tumor types. Of the 5 remaining borderline tumors, 4 clustered with the benign group and 1 with the malignant group. It is noteworthy that these tumors were the most peripheral nodes in these two groups, indicating an expression profile closer to the intermediate group than other benign and malignant tumors ( FIG. 2 ).
  • the test set was also subjected to K-means cluster analysis using both 2- and 3-groups.
  • the 2-group cluster algorithm tumors were separated into two groups based on their gene expression of the genes of interest. This algorithm distinguished benign and malignant tumors with 93% sensitivity and 82% specificity ( FIG. 3 ). Borderline tumors were divided, with four tumors (27%) grouped with benign tumors while eleven (73%) were grouped with the malignant tumors.
  • tumors were separated into three designated groups based on their expression profile.
  • malignant tumors primarily formed one group (with 1 borderline tumors)
  • benign tumors formed a second group (with 4 borderline tumors)
  • a third group was composed of 10 borderline tumors, 2 follicular adenoma, and 3 FVPTC ( FIG. 4 ).
  • These 2 follicular adenomas were previously grouped with malignancies in the 2-group algorithm (FA-3 and FA-4) and one of the three FVPTCs that was grouped with the borderline tumors (FVPTC-3) had previously clustered with the benign tumors in the 2-group algorithm.
  • MET met proto-oncogene
  • HMGA2 high-mobility group adenine/thymine-hook 2 gene
  • DIO1 deiodinase-1
  • BRAF mutational analysis was performed on all tumors in the test set. BRAF mutations were identified in 4 of 14 of the malignant tumors (29%) (Table 3). No borderline tumors or benign tumors had BRAF mutations.
  • Encapsulated follicular lesions with cytologic atypia remain a diagnostic challenge for pathologists.
  • the foregoing experiments employed molecular profiling to identify a third category of thyroid tumors that, based on gene expression data, is likely to be premalignant.
  • This third category of encapsulated follicular tumors with cytologic atypia typically did not fit into previously proposed benign or malignant classification schemes using standard histology, immunohistochemistry, or mutation analysis.
  • the majority of histologically borderline tumors (66.7%) fell into an intermediate group and only a small number share gene expression similarities with benign tumors (26.7%) or malignant tumors (6.7%; Kmeans cluster analysis) ( FIG. 4 ).
  • borderline tumors like malignant tumors, exhibited up-regulated gene expression of CITED 1 and pleckstrin and Sec7 domain 3 (PSD3) and down-regulated gene expression of FGFR2 relative to benign tumors (Table 1). These genes and others listed in Table 1 are potential markers of early tumorigenesis.
  • BRAF mutation analysis also were in agreement with other studies (see, Nikiforova et al., J Clin Endocrinol Metab. 2003; 88:5399-5404 (2003)), with mutations identified in 29% of malignancies.
  • BRAF mutations have not been identified in benign lesions or in borderline encapsulated follicular tumors (see, Arora et al., World J. Surg. 32:1237-1246 (2008); Fontaine et al., Oncogene 27:2228-2236 (2008)).
  • Some studies indicate that BRAF mutations are associated with more aggressive tumors (Frasca et al., Endocr Relat Cancer. 15:191-205 (2008); Kebebew et al., Ann Surg. 246:466-471 (2007)) indicating that borderline tumors are more likely to be indolent tumors.
  • BRAF mutation is more frequent in classic PTC than in FVPTC also indicates that, for FVPTCs derived from FAs, BRAF either is uninvolved in carcinogenesis or is involved only as a late event. In addition, because of its higher frequency in classic PTC versus FVPTC, BRAF mutational analysis remains of limited usefulness in the diagnostic evaluation of these lesions.
  • Immunohistochemical markers have been studied in a few well differentiated tumors of uncertain malignant potential (WDT-UMP) with variable results.
  • Papotti et al. (Mod Pathol. 18:541-546 (2005)) studied the expression of galectin-3 and HBME1 in 13 WDT-UMPs and noted some degree of staining with either antibody in 12 of 13 tumors.
  • Immunohistochemical staining for HBME1, Galectin-3, and CK19 revealed heterogeneous staining patterns. This variability, again, may reflect the biologically borderline nature of these tumors.
  • Liu et al. reviewed the outcome data from 42 patients with encapsulated, noninvasive FVPTCs who had a median 10-year follow-up and reported that no patients had recurrences and that none had lymph node metastasis.
  • borderline tumors represent a molecularly distinct group of tumors that may not need aggressive treatment.
  • This example describes procedures for performing reverse transcription, real-time, quantitative PCR (RT-qPCR).
  • RNA from human cells is isolated by a standard mini-column method, RNAeasy® Mini Kit (Qiagen, Valencia, Calif.). RNA sample quality is evaluated based on electrophoretic integrity of 18S and 28S rRNA bands on a 2100 Bioanalyzer instrument (Agilent, Santa Clara, Calif.) and by standard spectrophotometric absorbance methods at 230, 260 and 280 nm wavelengths on a NanoDrop 1000 (NanoDrop/Thermo Scientific, Wilmington, Del.).
  • Preparation of cDNA from the RNA samples is carried out using 1.0 ⁇ g of total RNA into a standard 20 ⁇ l MMLV reverse transcriptase (Promega, Madison, Wis.) reaction according to the manufacturer's instructions using Promega buffers with a combination of 50 ⁇ g/ml random hexamers (Integrated DNA Technologies, Coralville, Iowa) and 2.5 ng/ ⁇ l oligo d(T16) (Integrated DNA Technologies, Coralville, Iowa) to prime the first strand synthesis.
  • the cDNA sample is diluted with 91 ⁇ l nuclease-free water ( ⁇ 5 fold) so that 1 ⁇ l ( ⁇ 1/100) is used as the template for individual 25 ⁇ l PCR reactions.
  • SYBR® Green real-time PCR is set up by combining 12.5 ⁇ l 2 ⁇ SYBR® Green PCR Master Mix (Applied Biosystems, Foster City, Calif.) with 1 ⁇ l cDNA sample, 1 ⁇ l PCR primer mix (10 ⁇ M each forward and reverse primers from Tables 4 and 5) and 10 ⁇ l nuclease-free water in an appropriate reaction tube or plate.
  • Real-time PCR thermal cycling and detection is performed on either an ABI 7500 (Applied Biosystems, Foster City, Calif.) or Stratagene Mx3005P (Agilent, Santa Clara, Calif.) instrument for 1 cycle of 10 minutes at 95° C., then 40 cycles of 15 seconds at 95° C. and 60 seconds at 60° C., followed by the instrument specific dissociation analysis steps.
  • Ct values are determined and relative expression measurements obtained by the ⁇ Ct calculation method (Livak, K J, Schmittgen, T D. 2001 , Methods 25, 402-408).
  • This Example describes primers and probes for detecting expression of the differentially expressed genes described herein.
  • sequences of primers with SEQ ID NO:3-118 are shown below in Tables 4 and 5.
  • nucleic acid sequence for human ANK2 is available as NCBI accession number NM — 001148 (gi: 188595661). This sequence is recited below for easy reference as SEQ ID NO:119.
  • nucleic acid sequence for human ARHGAP6 is available as NCBI accession number NM — 013427 (gi: 95091874). This sequence is recited below for easy reference as SEQ ID NO:120.
  • nucleic acid sequence for human C11orf17 is available as NCBI accession number NM — 182901 (gi: 116174739). This sequence is recited below for easy reference as SEQ ID NO:121.
  • nucleic acid sequence for human CAPN3 is available as NCBI accession number NM — 000070 (gi: 27765081). This sequence is recited below for easy reference as SEQ ID NO:122.
  • nucleic acid sequence for human CDH16 is available as NCBI accession number NM — 004062 (gi: 16507958). This sequence is recited below for easy reference as SEQ ID NO:123.
  • nucleic acid sequence for human ChGn is available as NCBI accession number BC060772 (gi: 38174239). This sequence is recited below for easy reference as SEQ ID NO:124.
  • nucleic acid sequence for human CITED1 is available as NCBI accession number NM — 004143 (gi: 222136685). This sequence is recited below for easy reference as SEQ ID NO:125.
  • nucleic acid sequence for human CITED2 is available as NCBI accession number NM — 006079 (gi: 51807294). This sequence is recited below for easy reference as SEQ ID NO:126.
  • nucleic acid sequence for human CKB is available as NCBI accession number M16451 (gi: 180571). This sequence is recited below for easy reference as SEQ ID NO:127.
  • nucleic acid sequence for human COL9A3 is available as NCBI accession number NM — 001853 (gi: 119508425). This sequence is recited below for easy reference as SEQ ID NO:128.
  • nucleic acid sequence for human CSRP2 is available as NCBI accession number NM — 001321 (gi: 4503100). This sequence is recited below for easy reference as SEQ ID NO:129.
  • nucleic acid sequence for human DAPK2 is available as NCBI accession number NM — 0014326 (gi: 71774012). This sequence is recited below for easy reference as SEQ ID NO:130.
  • nucleic acid sequence for human DIO1 is available as NCBI accession number NM — 00972 (gi: 89357933). This sequence is recited below for easy reference as SEQ ID NO:131.
  • nucleic acid sequence for human DPP4 is available as NCBI accession number NM — 001935 (gi: 47078262). This sequence is recited below for easy reference as SEQ ID NO:132.
  • nucleic acid sequence for human DTX4 is available as NCBI accession number NM — 015177 (gi: 148237497). This sequence is recited below for easy reference as SEQ ID NO:133.
  • nucleic acid sequence for human DUSP4 is available as NCBI accession number NM — 001394 (gi: 58331238). This sequence is recited below for easy reference as SEQ ID NO:134.
  • nucleic acid sequence for human EFEMP1 is available as NCBI accession number NM — 004105 (gi: 86787911). This sequence is recited below for easy reference as SEQ ID NO:135.
  • nucleic acid sequence for human ELMO1 is available as NCBI accession number NM — 014800 (gi: 86787650). This sequence is recited below for easy reference as SEQ ID NO:136.
  • nucleic acid sequence for human FGFR2 is available as NCBI accession number NM — 000141 (gi: 189083823). This sequence is recited below for easy reference as SEQ ID NO:137.
  • nucleic acid sequence for human FLRT1 is available as NCBI accession number NM — 013280 (gi: 48762940). This sequence is recited below for easy reference as SEQ ID NO:138.
  • nucleic acid sequence for human FMOD is available as NCBI accession number NM — 002023 (gi: 71040110). This sequence is recited below for easy reference as SEQ ID NO:139.
  • nucleic acid sequence for human GALNT7 is available as NCBI accession number NM — 017423 (gi: 157502211). This sequence is recited below for easy reference as SEQ ID NO:140.
  • nucleic acid sequence for human GATM is available as NCBI accession number NM — 001482 (gi: 126090880). This sequence is recited below for easy reference as SEQ ID NO:141.
  • nucleic acid sequence for human HGD is available as NCBI accession number NM — 000187 (gi: 115527116). This sequence is recited below for easy reference as SEQ ID NO:142.
  • nucleic acid sequence for human HMGA2 is available as NCBI accession number NM — 003483 (gi: 62912480). This sequence is recited below for easy reference as SEQ ID NO:143.
  • nucleic acid sequence for human IGFBP6 is available as NCBI accession number NM — 002178 (gi: 49574524). This sequence is recited below for easy reference as SEQ ID NO:144.
  • nucleic acid sequence for human KIT is available as NCBI accession number NM — 000222 (gi: 148005048). This sequence is recited below for easy reference as SEQ ID NO:145.
  • nucleic acid sequence for human LRP4 is available as NCBI accession number NM — 002334 (gi: 157384997). This sequence is recited below for easy reference as SEQ ID NO:146.
  • nucleic acid sequence for human MATN2 is available as NCBI accession number NM — 002380 (gi: 62548859). This sequence is recited below for easy reference as SEQ ID NO:147.
  • nucleic acid sequence for human MET is available as NCBI accession number NM — 001127500 (gi: 188595715). This sequence is recited below for easy reference as SEQ ID NO:148.
  • nucleic acid sequence for human MYH10 is available as NCBI accession number NM — 005964 (gi: 41406063). This sequence is recited below for easy reference as SEQ ID NO:149.
  • nucleic acid sequence for human PFAAP5 is available as NCBI accession number AF530063 (gi: 33329092). This sequence is recited below for easy reference as SEQ ID NO:150.
  • nucleic acid sequence for human PGF is available as NCBI accession number NM — 002632 (gi: 56676307). This sequence is recited below for easy reference as SEQ ID NO:151.
  • nucleic acid sequence for human PIP3-E is available as NCBI accession number AJ310566 (gi: 18307480). This sequence is recited below for easy reference as SEQ ID NO:152.
  • nucleic acid sequence for human PKNOX2 is available as NCBI accession number NM — 022062 (gi: 116812643). This sequence is recited below for easy reference as SEQ ID NO:153.
  • nucleic acid sequence for human PRKACB is available as NCBI accession number NM — 182948 (gi: 46909585). This sequence is recited below for easy reference as SEQ ID NO:154.
  • nucleic acid sequence for human PROS1 is available as NCBI accession number NM — 000313 (gi: 223671900). This sequence is recited below for easy reference as SEQ ID NO:155.
  • nucleic acid sequence for human PSD3 is available as NCBI accession number NM — 015310 (gi: 117606359). This sequence is recited below for easy reference as SEQ ID NO:156.
  • nucleic acid sequence for human QPCT is available as NCBI accession number NM — 012413 (gi: 68216098). This sequence is recited below for easy reference as SEQ ID NO:157.
  • nucleic acid sequence for human RAB27A is available as NCBI accession number NM — 004580 (gi: 34485707). This sequence is recited below for easy reference as SEQ ID NO:158.
  • nucleic acid sequence for human RXRG is available as NCBI accession number NM — 006917 (gi: 58331205). This sequence is recited below for easy reference as SEQ ID NO:159.
  • nucleic acid sequence for human SDC4 is available as NCBI accession number NM — 002999 (gi: 38201674). This sequence is recited below for easy reference as SEQ ID NO:160.
  • nucleic acid sequence for human SERPINA1 is available as NCBI accession number NM — 001127707 (gi: 189163541). This sequence is recited below for easy reference as SEQ ID NO:161.
  • nucleic acid sequence for human SLC25A15 is available as NCBI accession number NM — 014252 (gi: 237649033). This sequence is recited below for easy reference as SEQ ID NO:162.
  • nucleic acid sequence for human SLC4A4 is available as NCBI accession number NM — 001098484 (gi: 197927157). This sequence is recited below for easy reference as SEQ ID NO:163.
  • nucleic acid sequence for human SLIT1 is available as NCBI accession number NM — 003061 (gi: 188528674). This sequence is recited below for easy reference as SEQ ID NO:164.
  • nucleic acid sequence for human SPTAN1 is available as NCBI accession number NM — 001130438 (gi: 194595508). This sequence is recited below for easy reference as SEQ ID NO:165.
  • nucleic acid sequence for human TFCP2L1 is available as NCBI accession number NM — 014553 (gi: 212276201). This sequence is recited below for easy reference as SEQ ID NO:166.
  • nucleic acid sequence for human TIAM1 is available as NCBI accession number NM — 003253 (gi: 115583669). This sequence is recited below for easy reference as SEQ ID NO:167.
  • nucleic acid sequence for human TIMP1 is available as NCBI accession number NM — 003254 (gi: 73858576). This sequence is recited below for easy reference as SEQ ID NO:168.
  • nucleic acid sequence for human TNS3 is available as NCBI accession number NM — 022748 (gi: 134152712). This sequence is recited below for easy reference as SEQ ID NO:169.
  • nucleic acid sequence for human TSPAN12 is available as NCBI accession number NM — 012338 (gi: 48255911). This sequence is recited below for easy reference as SEQ ID NO:170.
  • nucleic acid sequence for human UPP1 is available as NCBI accession number NM — 003364 (gi: 31742506). This sequence is recited below for easy reference as SEQ ID NO:171.
  • nucleic acid sequence for human NAUK2 is available as NCBI accession number NM — 030952 (gi: 13569921). This sequence is recited below for easy reference as SEQ ID NO:172.
  • a host cell includes a plurality (for example, a culture or population) of such host cells, and so forth.
  • a reference to “a host cell” includes a plurality (for example, a culture or population) of such host cells, and so forth.
  • the patent be interpreted to be limited to the specific examples or embodiments or methods specifically disclosed herein.
  • the patent be interpreted to be limited by any statement made by any Examiner or any other official or employee of the Patent and Trademark Office unless such statement is specifically and without qualification or reservation expressly adopted in a responsive writing by Applicants.
US13/063,429 2008-09-12 2009-09-11 Thyroid tumors identified Abandoned US20110251091A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/063,429 US20110251091A1 (en) 2008-09-12 2009-09-11 Thyroid tumors identified

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US19184508P 2008-09-12 2008-09-12
US20781209P 2009-02-17 2009-02-17
US13/063,429 US20110251091A1 (en) 2008-09-12 2009-09-11 Thyroid tumors identified
PCT/US2009/005091 WO2010030365A2 (fr) 2008-09-12 2009-09-11 Identification de tumeurs de la tyroïde

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/005091 A-371-Of-International WO2010030365A2 (fr) 2008-09-12 2009-09-11 Identification de tumeurs de la tyroïde

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/087,812 Continuation US9587279B2 (en) 2008-09-12 2013-11-22 Thyroid tumors identified

Publications (1)

Publication Number Publication Date
US20110251091A1 true US20110251091A1 (en) 2011-10-13

Family

ID=41343416

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/063,429 Abandoned US20110251091A1 (en) 2008-09-12 2009-09-11 Thyroid tumors identified
US14/087,812 Active US9587279B2 (en) 2008-09-12 2013-11-22 Thyroid tumors identified
US15/440,489 Active US10889865B2 (en) 2008-09-12 2017-02-23 Thyroid tumors identified

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/087,812 Active US9587279B2 (en) 2008-09-12 2013-11-22 Thyroid tumors identified
US15/440,489 Active US10889865B2 (en) 2008-09-12 2017-02-23 Thyroid tumors identified

Country Status (2)

Country Link
US (3) US20110251091A1 (fr)
WO (1) WO2010030365A2 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9587279B2 (en) 2008-09-12 2017-03-07 Cornell University Thyroid tumors identified
CN110257515A (zh) * 2019-06-14 2019-09-20 清华-伯克利深圳学院筹备办公室 一种用于乳腺癌诊断的分子标志物及其应用
US20210164045A1 (en) * 2019-08-30 2021-06-03 Janssen Biotech, Inc. B-cell maturation complex car t construct and primers
CN115792247A (zh) * 2023-02-09 2023-03-14 杭州市第一人民医院 蛋白组合在制备甲状腺乳头状癌风险辅助分层系统中的应用

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8338109B2 (en) 2006-11-02 2012-12-25 Mayo Foundation For Medical Education And Research Predicting cancer outcome
AU2009253675A1 (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
US10407731B2 (en) 2008-05-30 2019-09-10 Mayo Foundation For Medical Education And Research Biomarker panels for predicting prostate cancer outcomes
US9495515B1 (en) 2009-12-09 2016-11-15 Veracyte, Inc. Algorithms for disease diagnostics
US10236078B2 (en) 2008-11-17 2019-03-19 Veracyte, Inc. Methods for processing or analyzing a sample of thyroid tissue
US9074258B2 (en) 2009-03-04 2015-07-07 Genomedx Biosciences Inc. Compositions and methods for classifying thyroid nodule disease
JP6078339B2 (ja) 2009-05-07 2017-02-08 ベラサイト インコーポレイテッド 甲状腺状態の診断のための方法および組成物
US10446272B2 (en) 2009-12-09 2019-10-15 Veracyte, Inc. Methods and compositions for classification of samples
US20190100809A1 (en) * 2010-05-11 2019-04-04 Veracyte, Inc. Algorithms for disease diagnostics
AU2012352153B2 (en) 2011-12-13 2018-07-26 Veracyte, Inc. Cancer diagnostics using non-coding transcripts
EP4219765A3 (fr) 2012-08-16 2023-09-20 Decipher Biosciences, Inc. Pronostic du cancer de la prostate à l'aide de biomarqueurs
JP6200281B2 (ja) * 2012-11-29 2017-09-20 シスメックス株式会社 甲状腺腫瘍の性状の判別を補助する方法およびその方法に用いるマーカーセット
US10526655B2 (en) 2013-03-14 2020-01-07 Veracyte, Inc. Methods for evaluating COPD status
US11976329B2 (en) 2013-03-15 2024-05-07 Veracyte, Inc. Methods and systems for detecting usual interstitial pneumonia
EP3215170A4 (fr) 2014-11-05 2018-04-25 Veracyte, Inc. Systèmes et procédés de diagnostic de la fibrose pulmonaire idiopathique sur des biopsies transbronchiques à l'aide de l'apprentissage automatique et de données de transcription dimensionnelle élevée
EP3034620A1 (fr) * 2014-12-17 2016-06-22 Diaxonhit Compositions et procédés permettant de diagnostiquer un cancer de la thyroïde
CN104450927B (zh) * 2014-12-18 2016-09-07 北京大学人民医院 定量检测csrp2基因表达的引物和探针及其应用
WO2018039490A1 (fr) 2016-08-24 2018-03-01 Genomedx Biosciences, Inc. Utilisation de signatures génomiques en vue d'une prédiction de la réactivité de patients atteints d'un cancer de la prostate à une radiothérapie postopératoire
US11208697B2 (en) 2017-01-20 2021-12-28 Decipher Biosciences, Inc. Molecular subtyping, prognosis, and treatment of bladder cancer
EP3593140A4 (fr) 2017-03-09 2021-01-06 Decipher Biosciences, Inc. Sous-typage du cancer de la prostate pour prédire la réponse à une thérapie hormonale
EP3596469A1 (fr) 2017-03-12 2020-01-22 Yeda Research and Development Co., Ltd. Procédés de diagnostic et de pronostic du cancer
WO2018167780A1 (fr) 2017-03-12 2018-09-20 Yeda Research And Development Co. Ltd. Méthodes de diagnostic et de traitement du cancer
CA3062716A1 (fr) 2017-05-12 2018-11-15 Decipher Biosciences, Inc. Signatures genetiques pour predire une metastase du cancer de la prostate et identifier la virulence d'une tumeur
US11217329B1 (en) 2017-06-23 2022-01-04 Veracyte, Inc. Methods and systems for determining biological sample integrity
CN109457032B (zh) * 2018-12-20 2022-04-12 北京优迅医学检验实验室有限公司 甲状腺癌分子诊断试剂盒
KR102399033B1 (ko) * 2019-10-30 2022-05-17 재단법인 아산사회복지재단 갑상선 여포 종양의 감별진단용 조성물 및 이를 이용한 감별진단 방법

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070099209A1 (en) * 2005-06-13 2007-05-03 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US20090298061A1 (en) * 2005-07-29 2009-12-03 Siemens Healthcare Diagnostics Inc. Diagnostic Methods for the Prediction of Therapeutic Success, Recurrence Free and Overall Survival in Cancer Therapy
US20120172243A1 (en) * 2009-04-29 2012-07-05 Elai Davicioni Systems and methods for expression-based classification of thyroid tissue

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5698410A (en) * 1995-06-07 1997-12-16 Mount Sinai School Of Medicine Of The City University Of New York Highly sensitive immunocytochemical method for diagnosis of malignant effusions
US6436642B1 (en) * 1999-04-20 2002-08-20 Curagen Corporation Method of classifying a thyroid carcinoma using differential gene expression
JP2007516693A (ja) 2003-06-09 2007-06-28 ザ・リージェンツ・オブ・ザ・ユニバーシティ・オブ・ミシガン 癌の治療および診断のための組成物および方法
WO2005085471A2 (fr) * 2004-03-03 2005-09-15 Universität Leipzig Procede et agent pour diagnostiquer de maniere differentielle des tumeurs de la glande thyroide
US7319011B2 (en) * 2004-04-08 2008-01-15 Duke University Method for distinguishing follicular thyroid adenoma (FTA) from follicular thyroid carcinoma (FTC)
WO2005100608A2 (fr) * 2004-04-09 2005-10-27 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services, National Institutes Of Health Outil diagnostique permettant de diagnostiquer des lesions thyroidiennes benignes contre des lesions thyroidiennes malignes
CN101137903A (zh) * 2004-07-19 2008-03-05 罗彻斯特大学 神经变性疾病的生物标记
US20070037186A1 (en) * 2005-05-20 2007-02-15 Yuqiu Jiang Thyroid fine needle aspiration molecular assay
US7598052B2 (en) * 2005-10-11 2009-10-06 The Regents Of The University Of Michigan Expression profile of thyroid cancer
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
WO2010030365A2 (fr) 2008-09-12 2010-03-18 Cornell Research Foundation, Inc. Wmc Identification de tumeurs de la tyroïde
CA3153682A1 (en) * 2008-11-17 2010-05-20 Veracyte, Inc. Methods and compositions of molecular profiling for disease diagnostics

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070099209A1 (en) * 2005-06-13 2007-05-03 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US20090298061A1 (en) * 2005-07-29 2009-12-03 Siemens Healthcare Diagnostics Inc. Diagnostic Methods for the Prediction of Therapeutic Success, Recurrence Free and Overall Survival in Cancer Therapy
US20120172243A1 (en) * 2009-04-29 2012-07-05 Elai Davicioni Systems and methods for expression-based classification of thyroid tissue

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9587279B2 (en) 2008-09-12 2017-03-07 Cornell University Thyroid tumors identified
US10889865B2 (en) 2008-09-12 2021-01-12 Cornell University Thyroid tumors identified
CN110257515A (zh) * 2019-06-14 2019-09-20 清华-伯克利深圳学院筹备办公室 一种用于乳腺癌诊断的分子标志物及其应用
US20210164045A1 (en) * 2019-08-30 2021-06-03 Janssen Biotech, Inc. B-cell maturation complex car t construct and primers
CN115792247A (zh) * 2023-02-09 2023-03-14 杭州市第一人民医院 蛋白组合在制备甲状腺乳头状癌风险辅助分层系统中的应用

Also Published As

Publication number Publication date
WO2010030365A2 (fr) 2010-03-18
US9587279B2 (en) 2017-03-07
US10889865B2 (en) 2021-01-12
US20170166980A1 (en) 2017-06-15
WO2010030365A3 (fr) 2010-06-03
US20150167094A1 (en) 2015-06-18

Similar Documents

Publication Publication Date Title
US10889865B2 (en) Thyroid tumors identified
KR102114412B1 (ko) 위장관췌장 신경내분비 신생물 (GEP-NENs)의 예측 방법
KR101421326B1 (ko) 유방암 예후 예측을 위한 조성물 및 이를 포함하는 키트
AU2018210695A1 (en) Molecular subtyping, prognosis, and treatment of bladder cancer
KR100964193B1 (ko) 간암 예후 마커
CA2430981A1 (fr) Caracterisation de l'expression genique des carcinomes primaires du sein a l'aide de reseaux de genes d'interet
CN101258249A (zh) 检测黑素瘤的方法和试剂
CA2442820A1 (fr) Etablissement de profils d'expression genique dans un microreseau dans un adenocarcinome a cellules claires, pronostic et identification de cible medicamenteuse
AU2016331663A1 (en) Pathogen biomarkers and uses therefor
MXPA05005653A (es) Determinacion y seleccion terapeutica de genes de insuficiencia cardiaca.
CA2403946A1 (fr) Genes exprimes lors de la differenciation de cellules spumeuses
KR20060045950A (ko) 혈액학적 악성종양에 대한 예후
AU2008203226A1 (en) Colorectal cancer prognostics
US20230022417A1 (en) Chemical compositions and methods of use
WO2007135174A1 (fr) Modèle d'expression génique prédictif pour le cancer colorectal
AU2004205270B2 (en) Colorectal cancer prognostics
US20020137077A1 (en) Genes regulated in activated T cells
US20230022236A1 (en) Chemical compositions and methods of use
JP2003235573A (ja) 糖尿病性腎症マーカーおよびその利用
CN100516876C (zh) 用于诊断肾细胞癌(rcc)和其他实体瘤的方法
CN1856573A (zh) 用于神经母细胞瘤预后诊断的微阵列和神经母细胞瘤预后诊断方法
KR102480128B1 (ko) 면역력 강화 소 African Humped Cattle (AFH) 품종 특이적 단일염기다형성 및 그의 용도
KR20100115283A (ko) 간암 예후 마커
MXPA04008401A (en) Colorectal cancer prognostics

Legal Events

Date Code Title Description
AS Assignment

Owner name: QIAGEN GAITHERSBURG, INC, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BLANCHARD, RAYMOND K.;LIU, GUOZHEN;REEL/FRAME:026603/0355

Effective date: 20110513

Owner name: CORNELL UNIVERSITY, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FAHEY, THOMAS J.;ARORA, NIMMI;SEOGNAMIGLIO, THERESA;SIGNING DATES FROM 20110512 TO 20110525;REEL/FRAME:026603/0278

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION