US20140031250A1 - Biomarkers of Cancer - Google Patents

Biomarkers of Cancer Download PDF

Info

Publication number
US20140031250A1
US20140031250A1 US13/877,373 US201113877373A US2014031250A1 US 20140031250 A1 US20140031250 A1 US 20140031250A1 US 201113877373 A US201113877373 A US 201113877373A US 2014031250 A1 US2014031250 A1 US 2014031250A1
Authority
US
United States
Prior art keywords
subject
cancer
satellite
level
value
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
US13/877,373
Inventor
David Tsai Ting
Daniel A. Haber
Shyamala Maheswaran
Doron Lipson
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.)
General Hospital Corp
Original Assignee
General Hospital Corp
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 General Hospital Corp filed Critical General Hospital Corp
Priority to US13/877,373 priority Critical patent/US20140031250A1/en
Publication of US20140031250A1 publication Critical patent/US20140031250A1/en
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MAHESWARAN, SHYAMALA, TING, David Tsai
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HABER, DANIEL A.
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
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • This invention relates to methods of diagnosing cancer, based on detecting the presence of increased levels of expression of satellite repeats and/or Line-1.
  • heterochromatin is comprised of centric (minor) and pericentric (major) satellite repeats that are required for formation of the mitotic spindle complex and faithful chromosome segregation (M. Guenatri, D. Bailly, C. Maison, G. Almouzni, J Cell Biol 166, 493 (Aug.
  • Kanellopoulou et al. Genes Dev 19, 489 (Feb. 15, 2005); T. Fukagawa et al., Nat Cell Biol 6, 784 (August, 2004)) and from DNA demethylation, heat shock, or the induction of apoptosis (H. Bouzinba-Segard, A. Guais, C. Francastel, Proc Natl Acad Sci USA 103, 8709 (Jun. 6, 2006); R. Valgardsdottir et al., Nucleic Acids Res 36, 423 (February, 2008)).
  • the present invention is based, at least in part, on the identification of massive expression of satellite repeats in tumor cells, and of increased levels of Line-1, e.g., in tumor cells including circulating tumor cells (CTCs).
  • CTCs circulating tumor cells
  • Described herein are methods for diagnosing cancer, e.g., solid malignancies of epithelial origin such as pancreatic, lung, breast, prostate, renal, ovarian or colon cancer, based on the presence of increased levels of expression of satellite repeats and/or Line-1.
  • the invention provides methods, e.g., in vitro methods, for detecting the presence of cancer in a subject, including determining a level of LINE-1 in a sample from the subject to obtain a test value; and comparing the test value to a reference value, wherein a test value compared to the reference value indicates whether the subject has cancer.
  • the reference value represents a threshold level of LINE-1, wherein the presence of a level of LINE-1 in the subject that is above the reference value indicates that the subject has cancer, and the presence of a level of LINE-1 in the subject that is below the reference value indicates that the subject is unlikely to have cancer.
  • the invention provides methods, e.g., in vitro methods, for detecting the presence of cancer in a subject, including determining a level of satellite transcripts in a sample from the subject to obtain a test value; and comparing the test value to a reference value, wherein a test value compared to the reference value indicates whether the subject has cancer.
  • the satellite transcripts comprise one or more of ALR, HSATII, GSATII, TAR1, and SST1.
  • the satellite transcript is ALR and/or HSATII, and the presence of a level of ALR and/or HSATII satellite transcripts above the reference level indicates that the subject has a tumor.
  • the satellite transcript is GSATII, TAR1 and/or SST1, and the presence of a level of GSATII, TAR1 and/or SST1 satellite transcripts below the reference level indicates that subject has a tumor.
  • the invention provides methods, e.g., in vitro methods, for evaluating the efficacy of a treatment for cancer in a subject.
  • the methods include determining a level of LINE-1 in a first sample from the subject to obtain a first value; administering a treatment for cancer to the subject; determining a level of LINE-1 in a subsequent sample obtained from the subject at a later time, to obtain a treatment value; and comparing the first value to the treatment value.
  • a treatment value that is below the first value indicates that the treatment is effective.
  • the invention provides methods, e.g., in vitro methods, for evaluating the efficacy of a treatment for cancer in a subject.
  • the methods include determining a level of satellite transcripts in a first sample from the subject to obtain a first value; administering a treatment for cancer to the subject; determining a level of satellite transcripts in a subsequent sample obtained from the subject at a later time, to obtain a treatment value; and comparing the first value to the treatment value, wherein a treatment value that is below the first value indicates that the treatment is effective.
  • the satellite transcripts comprise one or more of ALR, HSATII, GSATII, TAR1, and SST1.
  • the first and second samples are known or suspected to comprise tumor cells, e.g., blood samples known or suspected of comprising circulating tumor cells (CTCs), or biopsy samples known or suspected of comprising tumor cells.
  • the sample comprises free RNA in serum or RNA within exosomes in blood.
  • the treatment includes administration of a surgical intervention, chemotherapy, radiation therapy, or a combination thereof.
  • the subject is a mammal, e.g., a human or veterinary subject, e.g., experimental animal.
  • the cancer is a solid tumor of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • the methods described herein include measuring a level of LINE-1 transcript.
  • the level of a LINE-1 transcript or satellite is determined using a branched DNA assay.
  • FIG. 1B is a graphical representation of sequence read contributions from major satellite among all primary tumors, cancer cell lines, and normal tissues.
  • FIG. 2A shows the results of Northern blot analysis of three KrasG12D, Tp53lox/+ pancreatic primary tumors (Tumors 1-3) and a stable cell line (CL3) derived from Tumor 3.
  • FIG. 2B shows the results of Northern blot analysis of CL3 before (0) and after (+) treatment with the DNA hypomethylating agent 5-azacitadine (AZA).
  • FIG. 2C shows the results of Northern blot analysis of total RNA from multiple adult and fetal mouse tissues. All Northern blots exposed for approximately 30 minutes.
  • FIG. 2D is a pair of photomicrographs showing the results of RNA in-situ hybridization (ISH) of normal pancreas (left) and primary pancreatic ductal adenocarcinoma (right), hybridized with a 1 kb major satellite repeat probe.
  • ISH RNA in-situ hybridization
  • FIG. 2E is a set of three photomicrographs showing the results of ISH analysis of preneoplastic PanIN (P) lesion, adjacent to PDAC (T) and normal pancreas (N), showing positive staining in PanIN, with increased expression in full carcinoma. Higher magnification (40 ⁇ ) of PanIN (left) and PDAC (right) lesions.
  • FIG. 3A is a bar graph showing the Total satellite expression in human pancreatic ductal adenocarcinoma (PDAC), normal pancreas, other cancers (L—lung, K—kidney, O—ovary, P—prostate), and other normal human tissues (1—fetal brain, 2—brain, 3—colon, 4—fetal liver, 5—liver, 6—lung, 7—kidney, 8—placenta, 9—prostate, and 10—uterus) quantitated by DGE. Satellite expression is shown as transcripts per million aligned to human genome.
  • PDAC pancreatic ductal adenocarcinoma
  • FIG. 4A shows the results of multiple linear correlation analysis of major satellite to other cellular transcripts among all mouse tumors and normal tissues as depicted by a heat map.
  • X-axis is samples ordered by expression of major satellite and y-axis is genes ordered by linear correlation to major satellite expression.
  • Light grey (High) and dark grey (Low) color is log 2 (reads per million).
  • FIG. 4B is a dot graph showing the Median distance of transcriptional start sites of all genes to Line-1 elements ordered by linearity to satellite expression (Dark gray; highest linearity to the left) or by random (Light gray). Plotted by genes binned in 100 s.
  • FIG. 4C is a dot graph showing Top genes with highest linearity (R>0.85) defining satellite correlated genes or SCGs plotted by frequency against distance of transcriptional start site to LINE-1 elements (Dark gray) compared to the expected frequency of these genes (Light gray).
  • FIG. 4D is a set of four photomicrographs showing the results of immunohistochemistry of mouse PDAC (KrasG12D, Tp53 lox/+) for the neuroendocrine marker chromogranin A. Tumors are depicted as a function of increasing chromogranin A staining (dark grey), with the relative level of major satellite expression noted for each tumor at the bottom of each image (percentage of all transcripts)
  • FIG. 5 is a bar graph indicating fold change expression of the indicated genes in CTC Device vs. control device.
  • the subjects were newly diagnosed metastatic pancreatic adenocarcinoma patients. LINE-1 expression was seen in all patients at some point.
  • FIG. 6B is an image of RNA in situ hybridization of HSATII using Affymetrix ViewRNA of a potential human pancreatic circulating tumor cell captured on the HB-chip.
  • HSATII lightest areas; yellow in original
  • DAPI nuclear stain medium grey areas, blue in original.
  • Scale bar 20 ⁇ m.
  • the present invention is based, at least in part, on the identification of a massive generation of LINE-1 protein and bidirectional ncRNAs from the major satellite repeat in mouse tumor models and from ALR and HSATII satellite repeats in human cancers.
  • the exceptional magnitude of satellite levels in these cancers is unprecedented. This is likely to result from a general derepression of chromosomal marks affecting both satellites and LINE-1 retrotransposons, with proximity to LINE-1 activation potentially affecting the expression of selected cellular mRNAs. Together, the very high expression of satellites may affect chromosomal integrity and genetic stability, while the co-deregulated coding sequences may affect cell fates and biological behavior of cancer cells.
  • the methods described herein can be used to diagnose the presence of, and monitor the efficacy of a treatment for, cancer, e.g., solid tumors of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer, in a subject.
  • cancer e.g., solid tumors of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer
  • hyperproliferative refer to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth.
  • Hyperproliferative disease states may be categorized as pathologic, i.e., characterizing or constituting a disease state, or may be categorized as non-pathologic, i.e., a deviation from normal but not associated with a disease state.
  • pathologic i.e., characterizing or constituting a disease state
  • non-pathologic i.e., a deviation from normal but not associated with a disease state.
  • the term is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness.
  • a “tumor” is an abnormal growth of hyperproliferative cells.
  • Cancer refers to pathologic disease states, e.g., characterized by malignant tumor growth.
  • cancer e.g., solid tumors of epithelial origin, e.g., as defined by the ICD-O (International Classification of Diseases-Oncology) code (revision 3), section (8010-8790), e.g., early stage cancer
  • ICD-O International Classification of Diseases-Oncology
  • section (8010-8790) e.g., early stage cancer
  • the methods can include the detection of expression levels of satellite repeats in a sample comprising cells known or suspected of being tumor cells, e.g., cells from solid tumors of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer cells.
  • the methods can include the detection of increased levels of LINE-1 in a sample, e.g., a sample known or suspected of including tumor cells, e.g., circulating tumor cells (CTCs), e.g., using a microfluidic device as described herein.
  • CTCs circulating tumor cells
  • Cancers of epithelial origin can include pancreatic cancer (e.g., pancreatic adenocarcinoma or intraductal papillary mucinous carcinoma (IPMN, pancreatic mass)), lung cancer (e.g., non-small cell lung cancer), prostate cancer, breast cancer, renal cancer, ovarian cancer, or colon cancer.
  • pancreatic cancer e.g., pancreatic adenocarcinoma or intraductal papillary mucinous carcinoma (IPMN, pancreatic mass)
  • lung cancer e.g., non-small cell lung cancer
  • prostate cancer e.g., breast cancer, renal cancer, ovarian cancer, or colon cancer.
  • the present methods can be used to distinguish between benign IPMN, for which surveillance is the standard treatment, and malignant IPMN, which require resection, a procedure associated with significant morbidity and a small but significant possibility of death.
  • the methods described herein can be used for surveillance/monitoring of the subject, e.g., the methods can be repeated at selected intervals (e.g., every 3, 6, 12, or 24 months) to determine whether a benign IPMN has become a malignant IPMN warranting surgical intervention.
  • the methods can be used to distinguish bronchioloalveolar carcinomas from reactive processes (e.g., postpneumonic reactive processes) in samples from subjects suspected of having non-small cell lung cancer.
  • the methods in a sample from a subject who is suspected of having breast cancer, can be used to distinguish ductal hyperplasia from atypical ductal hyperplasia and ductal carcinoma in situ (DCIS).
  • DCIS ductal carcinoma in situ
  • the methods can be used to distinguish between atypical small acinar proliferation and malignant cancer.
  • the methods in subjects suspected of having bladder cancer, can be used to detect, e.g., transitional cell carcinoma (TCC), e.g., in urine specimens.
  • TCC transitional cell carcinoma
  • subjects diagnosed with Barrett's Esophagus in subjects diagnosed with Barrett's Esophagus (Sharma, N Engl J Med.
  • the methods can be used for distinguishing dysplasia in Barrett's esophagus from a reactive process.
  • the clinical implications are significant, as a diagnosis of dysplasia demands a therapeutic intervention.
  • Other embodiments include, but are not limited to, diagnosis of well differentiated hepatocellular carcinoma, ampullary and bile duct carcinoma, glioma vs. reactive gliosis, melanoma vs. dermal nevus, low grade sarcoma, and pancreatic endocrine tumors, inter alia.
  • the methods include obtaining a sample from a subject, and evaluating the presence and/or level of LINE-1 or satellites in the sample, and comparing the presence and/or level with one or more references, e.g., a control reference that represents a normal level of LINE-1 or satellites, e.g., a level in an unaffected subject or a normal cell from the same subject, and/or a disease reference that represents a level of LINE-1 or satellites associated with cancer, e.g., a level in a subject having pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • a control reference that represents a normal level of LINE-1 or satellites, e.g., a level in an unaffected subject or a normal cell from the same subject
  • a disease reference that represents a level of LINE-1 or satellites associated with cancer, e.g., a level in a subject having pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • the present methods can also be used to determine the stage of a cancer, e.g., whether a sample includes cells that are from a precancerous lesion, an early stage tumor, or an advanced tumor. For example, the present methods can be used to determine whether a subject has a precancerous pancreatic, breast, or prostate lesion.
  • the markers used are LINE-1, or satellite transcript ALR and/or HSATII
  • increasing levels are correlated with advancing stage.
  • satellite transcripts GSATII, TAR1 and/or SST1 decreasing levels are correlated with increasing stage.
  • levels of LINE-1 and satellite ALR and/or HSATII may be prognostic and predictive to clinical outcomes.
  • the sample is or includes blood, serum, and/or plasma, or a portion or subfraction thereof, e.g., free RNA in serum or RNA within exosomes in blood.
  • the sample comprises (or is suspected of comprising) CTCs.
  • the sample is or includes urine or a portion or subfraction thereof.
  • the sample includes known or suspected tumor cells, e.g., is a biopsy sample, e.g., a fine needle aspirate (FNA), endoscopic biopsy, or core needle biopsy; in some embodiments the sample comprises cells from the pancreatic, lung, breast, prostate, renal, ovarian or colon of the subject.
  • FNA fine needle aspirate
  • the sample comprises lung cells obtained from a sputum sample or from the lung of the subject by brushing, washing, bronchoscopic biopsy, transbronchial biopsy, or FNA, e.g., bronchoscopic, fluoroscopic, or CT-guided FNA (such methods can also be used to obtain samples from other tissues as well).
  • FNA fluoroscopic, or CT-guided FNA
  • the sample is frozen, fixed and/or permeabilized, e.g., is an formalin-fixed paraffin-embedded (FFPE) sample.
  • FFPE formalin-fixed paraffin-embedded
  • the level of satellite transcripts is detected, e.g., in a sample known or suspected to include tumor cells. In some embodiments, the level of satellite transcripts in a known or suspected tumor cell, e.g., a test cell, is compared to a reference level.
  • the methods include detecting levels of alpha (ALR) satellite transcripts (D. Lipson et al., Nat Biotechnol 27, 652 (July, 2009)) or HSATII satellite transcripts (J. Jurka et al., Cytogenet Genome Res 110, 462 (2005)); in some embodiments, those levels are compared to a reference.
  • ALR alpha
  • the reference level is a level of ALR and/or HSATII satellite transcripts in a normal (non-cancerous) cell, e.g., a normal cell from the same subject, or a reference level determined from a cohort of normal cells; the presence of levels of ALR and/or HSATII in the test cell above those in the normal cell indicate that the test cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • a normal (non-cancerous) cell e.g., a normal cell from the same subject, or a reference level determined from a cohort of normal cells
  • the presence of levels of ALR and/or HSATII in the test cell above those in the normal cell indicate that the test cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • the reference level of ALR and/or HSATII transcripts is a threshold level, and the presence of a level of ALR and/or HSATII satellite transcripts above the threshold level indicates that the cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • the methods include detecting levels of GSATII, TAR1 and/or SST1 transcripts; in some embodiments, those levels are compared to a reference.
  • the reference level is a level of GSATII, TAR1 and/or SST1 satellite transcripts in a normal (non-cancerous) cell, e.g., a normal cell from the same subject, or a reference level determined from a cohort of normal cells; the presence of levels of GSATII, TAR1 and/or SST1 in the test cell below those in the normal cell indicate that the test cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • the reference level of GSATII, TAR1 and/or SST1 transcripts is a threshold level, and the presence of a level of GSATII, TAR1 and/or SST1 satellite transcripts below the threshold level indicates that the cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • the levels of the satellite transcripts are normalized to a relatively non-variant transcript such as GAPDH, actin, or tubulin, e.g., the level of expression of the satellite is compared to the non-variant transcript.
  • a ratio of expression levels can be calculated, and the ratio can be compared to the ratio in a normal (non-cancerous) cell.
  • the presence of a ratio of ALR:GAPDH of over 10:1, e.g., over 50:1, over 100:1, or over 150:1, indicates that the test cell is a cancer cell; in some embodiments the presence of a ratio of ALR:GAPDH of about 3:1 or 5:1 indicates that the test cell is a normal cell.
  • the presence of a ratio of HSATII satellites:GAPDH transcripts of over 10:1, e.g., 20:1, 30:1, 40:1, or 45:1, indicates that the test cell is a cancer cell.
  • the presence of significant (e.g., more than about 100 transcripts per million aligned) levels of HSATII indicates that the test cell is a cancer cell.
  • the absence or presence of very low levels (e.g., less than about 20 transcripts per million aligned) of HSATII indicates that the test cell is a normal cell.
  • LINE Long interspersed nucleotide element
  • Singer, Cell 28 (3): 433-4 (1982) are a group of genetic elements that are found in large numbers in eukaryotic genomes, and generate insertion mutations, contribute to genomic instability and innovation, and can alter gene expression.
  • the canonical, full-length LINE-1 element is about 6 kilobases (kb) in length and includes a 5′ untranslated region (UTR) with an internal RNA polymerase II promoter (Swergold, Mol Cell Biol. 10(12):6718-29 (1990)), two open reading frames (designated ORF1 and ORF2) and a 3′ UTR containing a polyadenylation signal ending with an oligo dA-rich tail of variable length (Babushok and Kazazian, Hum Mutat. 28(6):527-39 (2007)).
  • UTR 5′ untranslated region
  • ORF1 and ORF2 two open reading frames
  • 3′ UTR containing a polyadenylation signal ending with an oligo dA-rich tail of variable length
  • Exemplary LINE-1 sequences include GenBank Ref. No. NM — 001164835.1 (nucleic acid) and NP — 001158307.1 (protein) for variant (1); and GenBank Ref. No. NM — 019079.4 (nucleic acid) and NP — 061952.3 (protein) for variant 2, which is the shorter transcript.
  • Variant 2 differs in the 5′ UTR compared to variant 1, but both variants 1 and 2 encode the same protein. See also Gene ID: 54596.
  • the methods for diagnosing cancer described herein include determining a level of LINE-1 mRNA in a cell, e.g., in CTCs present in blood of a subject to obtain a LINE-1 value, and comparing the value to an appropriate reference value, e.g., a value that represents a threshold level, above which the subject can be diagnosed with cancer.
  • the reference can also be a range of values, e.g., that indicate severity or stage of the cancer in the subject.
  • a suitable reference value can be determined by methods known in the art.
  • the reference level is a level of LINE-1 transcripts in a normal (non-cancerous) cell, e.g., a normal cell from the same subject, or a reference level determined from a cohort of normal cells; the presence of levels of LINE-1 in the test cell above those in the normal cell indicate that the test cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • the reference level of LINE-1 transcripts is a threshold level, and the presence of a level of LINE-1 transcripts above the threshold level indicates that the cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • RNA expression assays e.g., microarray analysis, RT-PCR, deep sequencing, cloning, Northern blot, and quantitative real time polymerase chain reaction (qRT-PCR).
  • qRT-PCR quantitative real time polymerase chain reaction
  • the level of the LINE-1 protein is detected.
  • the presence and/or level of a protein can be evaluated using methods known in the art, e.g., using quantitative immunoassay methods such as enzyme linked immunosorbent assays (ELISAs), immunoprecipitations, immunofluorescence, immunohistochemistry, enzyme immunoassay (EIA), radioimmunoassay (RIA), and Western blot analysis.
  • ELISAs enzyme linked immunosorbent assays
  • IA enzyme immunoassay
  • RIA radioimmunoassay
  • the methods include contacting an agent that selectively binds to a biomarker, e.g., to a satellite transcript or LINE-1 mRNA or protein (such as an oligonucleotide probe, an antibody or antigen-binding portion thereof) with a sample, to evaluate the level of the biomarker in the sample.
  • a biomarker e.g., to a satellite transcript or LINE-1 mRNA or protein (such as an oligonucleotide probe, an antibody or antigen-binding portion thereof)
  • the agent bears a detectable label.
  • the term “labeled,” with regard to an agent encompasses direct labeling of the agent by coupling (i.e., physically linking) a detectable substance to the agent, as well as indirect labeling of the agent by reactivity with a detectable substance.
  • detectable substances examples include chemiluminescent, fluorescent, radioactive, or colorimetric labels.
  • detectable substances can include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride, quantum dots, or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125 I, 131 I, 35 S or 3 H.
  • antibodies can be used.
  • Antibodies can be polyclonal, or more preferably, monoclonal.
  • An intact antibody, or an antigen-binding fragment thereof (e.g., Fab or F(ab′) 2 ) can be used.
  • high throughput methods e.g., protein or gene chips as are known in the art (see, e.g., Ch. 12, “Genomics,” in Griffiths et al., Eds. Modern genetic Analysis, 1999, W. H. Freeman and Company; Ekins and Chu, Trends in Biotechnology, 1999; 17:217-218; MacBeath and Schreiber, Science 2000, 289(5485):1760-1763; Simpson, Proteins and Proteomics: A Laboratory Manual, Cold Spring Harbor Laboratory Press; 2002; Hardiman, Microarrays Methods and Applications: Nuts & Bolts, DNA Press, 2003), can be used to detect the presence and/or level of satellites or LINE-1.
  • a kit for performing this assay is commercially-available from Affymetrix (ViewRNA).
  • microfluidic (e.g., “lab-on-a-chip”) devices can be used in the present methods. Such devices have been successfully used for microfluidic flow cytometry, continuous size-based separation, and chromatographic separation.
  • methods in which expression of satellites or LINE-1 is detected in circulating tumor cells (CTCs) can be used for the early detection of cancer, e.g., early detection of tumors of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • the devices can be used for separating CTCs from a mixture of cells, or preparing an enriched population of CTCs.
  • such devices can be used for the isolation of CTCs from complex mixtures such as whole blood.
  • a device can include an array of multiple posts arranged in a hexagonal packing pattern in a microfluidic channel upstream of a block barrier.
  • the posts and the block barrier can be functionalized with different binding moieties.
  • the posts can be functionalized with anti-EPCAM antibody to capture circulating tumor cells (CTCs); see, e.g., Nagrath et al., Nature 450:1235-1239 (2007), optionally with downstream block barriers functionalized with to capture LINE-1 nucleic acids or proteins, or satellites. See, e.g., (S. Maheswaran et al., N Engl J Med. 359, 366 (Jul.
  • Processes for enriching specific particles from a sample are generally based on sequential processing steps, each of which reduces the number of undesired cells/particles in the mixture, but one processing step may suffice in some embodiments.
  • Devices for carrying out various processing steps can be separate or integrated into one microfluidic system.
  • the devices include devices for cell/particle binding, devices for cell lysis, devices for arraying cells, and devices for particle separation, e.g., based on size, shape, and/or deformability or other criteria.
  • processing steps are used to reduce the number of cells prior to introducing them into the device or system.
  • the devices retain at least 75%, e.g., 80%, 90%, 95%, 98%, or 99% of the desired cells compared to the initial sample mixture, while enriching the population of desired cells by a factor of at least 100, e.g., by 1000, 10,000, 100,000, or even 1,000,000 relative to one or more non-desired cell types.
  • Some devices for the separation of particles rely on size-based separation with or without simultaneous cell binding.
  • Some size-based separation devices include one or more arrays of obstacles that cause lateral displacement of CTCs and other components of fluids, thereby offering mechanisms of enriching or otherwise processing such components.
  • the array(s) of obstacles for separating particles according to size typically define a network of gaps, wherein a fluid passing through a gap is divided unequally into subsequent gaps.
  • Both sieve and array sized-based separation devices can incorporate selectively permeable obstacles as described above with respect to cell-binding devices.
  • Devices including an array of obstacles that form a network of gaps can include, for example, a staggered two-dimensional array of obstacles, e.g., such that each successive row is offset by less than half of the period of the previous row.
  • the obstacles can also be arranged in different patterns. Examples of possible obstacle shapes and patterns are discussed in more detail in WO 2004/029221.
  • the device can provide separation and/or enrichment of CTCs using array-based size separation methods, e.g., as described in U.S. Pat. Pub. No. 2007/0026413.
  • the devices include one or more arrays of selectively permeable obstacles that cause lateral displacement of large particles such as CTCs and other components suspended in fluid samples, thereby offering mechanisms of enriching or otherwise processing such components, while also offering the possibility of selectively binding other, smaller particles that can penetrate into the voids in the dense matrices of nanotubes that make up the obstacles.
  • Devices that employ such selectively permeable obstacles for size, shape, or deformability based enrichment of particles, including filters, sieves, and enrichment or separation devices, are described in International Publication Nos.
  • 60/668,415 devices useful for arraying cells, e.g., those described in International Publication No. 2004/029221, U.S. Pat. No. 6,692,952, and U.S. application Ser. Nos. 10/778,831 and 11/146,581; and devices useful for fluid delivery, e.g., those described in U.S. application Ser. Nos. 11/071,270 and 11/227,469.
  • Two or more devices can be combined in series, e.g., as described in International Publication No. WO 2004/029221. All of the foregoing are incorporated by reference herein.
  • a device can contain obstacles that include binding moieties, e.g., monoclonal anti-EpCAM antibodies or fragments thereof, that selectively bind to particular cell types, e.g., cells of epithelial origin, e.g., tumor cells. All of the obstacles of the device can include these binding moieties; alternatively, only a subset of the obstacles include them.
  • Devices can also include additional modules, e.g., a cell counting module or a detection module, which are in fluid communication with the microfluidic channel device. For example, the detection module can be configured to visualize an output sample of the device.
  • a detection module can be in fluid communication with a separation or enrichment device.
  • the detection module can operate using any method of detection disclosed herein, or other methods known in the art.
  • the detection module includes a microscope, a cell counter, a magnet, a biocavity laser (see, e.g., Gourley et al., J. Phys. D: Appl. Phys., 36: R228-R239 (2003)), a mass spectrometer, a PCR device, an RT-PCR device, a microarray, RNA in situ hybridization system, or a hyperspectral imaging system (see, e.g., Vo-Dinh et al., IEEE Eng. Med. Biol.
  • a computer terminal can be connected to the detection module.
  • the detection module can detect a label that selectively binds to cells, proteins, or nucleic acids of interest, e.g., LINE-1 DNA, mRNA, or proteins, or satellite DNA or mRNA.
  • the microfluidic system includes (i) a device for separation or enrichment of CTCs; (ii) a device for lysis of the enriched CTCs; and (iii) a device for detection of LINE-1 DNA, mRNA, or proteins, or satellite DNA or mRNA.
  • a population of CTCs prepared using a microfluidic device as described herein is used for analysis of expression of LINE-1 and/or satellites using known molecular biological techniques, e.g., as described above and in Sambrook, Molecular Cloning: A Laboratory Manual, Third Edition (Cold Spring Harbor Laboratory Press; 3rd edition (Jan. 15, 2001)); and Short Protocols in Molecular Biology, Ausubel et al., eds. (Current Protocols; 52 edition (Nov. 5, 2002)).
  • devices for detection and/or quantification of expression of satellites or LINE-1 in an enriched population of CTCs are described herein and can be used for the early detection of cancer, e.g., tumors of epithelial origin, e.g., early detection of pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • cancer e.g., tumors of epithelial origin, e.g., early detection of pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • a treatment e.g., as known in the art, can be administered.
  • the efficacy of the treatment can be monitored using the methods described herein; an additional sample can be evaluated after (or during) treatment, e.g., after one or more doses of the treatment are administered, and a decrease in the level of LINE-1, and/or ALR and/or HSATII satellites expression, or in the number of LINE-1-, and/or ALR and/or HSATII satellite-expressing cells in a sample, would indicate that the treatment was effective, while no change or an increase in the level of LINE-1, and/or ALR and/or HSATII satellite expression or LINE-1-, and/or ALR and/or HSATII satellite-expressing cells would indicate that the treatment was not effective (the converse would of course be true for levels of GSATII, TAR1 and/or SST1 satellites).
  • the methods can be repeated multiple
  • the methods can be repeated at selected intervals, e.g., at 3, 6, 12, or 24 month intervals, to monitor the disease in the subject for early detection of progression to malignancy or development of cancer in the subject.
  • DGE digital gene expression
  • mice with pancreatic cancer of different genotypes were bred as previously described in the Bardeesy laboratory (Bardeesy et al., Proc Natl Acad Sci USA 103, 5947 (2006)). Normal wild type mice were purchased from Jackson laboratories. Animals were euthanized as per animal protocol guidelines. Pancreatic tumors and normal tissue were extracted sterilely and then flash frozen with liquid nitrogen. Tissues were stored at ⁇ 80° C. Cell lines were generated fresh for animals AH367 and AH368 as previously described (Aguirre et al., Genes Dev 17, 3112 (2003)) and established cell lines were cultured in RPMI-1640+10% FBS+1% Pen/Strep (Gibco/Invitrogen). Additional mouse tumors from colon and lung were generously provided by Kevin Haigis (Massachusetts General Hospital) and Kwok-Kin Wong (Dana Farber Cancer Institute).
  • Fresh frozen tissue was pulverized with a sterile pestle in a microfuge tube on dry ice.
  • Cell lines were cultured and fresh frozen in liquid nitrogen prior to nucleic acid extraction.
  • RNA and DNA from cell lines and fresh frozen tumor and normal tissues were all processed in the same manner.
  • RNA was extracted using the TRIzol® Reagent (Invitrogen) per manufacturer's specifications.
  • DNA from tissue and cell lines was extracted using the QIAamp Mini Kit (QIAGEN) per manufacturer's protocol.
  • DGE Digital Gene Expression
  • DNA sequencing sample prepping protocol from Helicos that has been previously described (Pushkarev, N. F. Neff, S. R. Quake, Nat Biotech 27, 847 (2009)). Briefly, genomic DNA was sheared with a Covaris S2 acoustic sonicator producing fragments averaging 200 bps and ranging from 100-500 bps. Sheared DNA was then cleaned with SPRI. DNA was then denatured and a poly-A tail was added to the 3′ end using terminal transferase.
  • the levels of satellite transcripts in tumor tissues were about 8,000-fold higher than the abundant mRNA Gapdh.
  • a second independent pancreatic tumor nodule from the same mouse showed a lower, albeit still greatly elevated, level of satellite transcript (4.5% of total cellular transcripts).
  • the composite distribution of all RNA reads among coding, ribosomal and other non-coding transcripts showed significant variation between primary tumors and normal tissues ( FIG. 1A ), suggesting that the global cellular transcriptional machinery is affected by the massive expression of satellite transcripts in primary tumors.
  • Immortalized cell lines established from 3 primary pancreatic tumors displayed minimal expression of satellite repeats, suggesting either negative selection pressure during in vitro proliferation or reestablishment of stable satellite silencing mechanisms under in vitro culture conditions ( FIG. 1A ).
  • the composite distribution of all RNA reads among coding, ribosomal and other non-coding transcripts shows significant variation with that of normal tissues ( FIG. 1B ), suggesting that the cellular transcriptional machinery is affected by the massive expression of satellite transcripts in these tumors.
  • Satellite Transcripts are of Various Sizes Depending on Tissue Type and Expression Levels are Linked to Genomic Methylation and Amplification
  • Northern Blot analysis of mouse primary pancreatic tumors was carried out as follows. Northern Blot was performed using the NorthernMax-Gly Kit (Ambion). Total RNA (10 ug) was mixed with equal volume of Glyoxal Load Dye (Ambion) and incubated at 50° C. for 30 min. After electrophoresis in a 1% agarose gel, RNA was transferred onto BrightStar-Plus membranes (Ambion) and crosslinked with ultraviolet light. The membrane was prehybridized in ULTRAhyb buffer (Ambion) at 68° C. for 30 min.
  • the mouse RNA probe (1100 bp) was prepared using the MAXIscript Kit (Ambion) and was nonisotopically labeled using the BrightStar Psoralen-Biotin Kit (Ambion) according to the manufacturer's instructions.
  • the membrane was hybridized in ULTRAhyb buffer (Ambion) at 68° C. for 2 hours. The membrane was washed with a Low Stringency wash at room temperature for 10 min, followed by two High Stringency washes at 68° C. for 15 min.
  • the BrightStar BioDetect Kit was used according to the manufacturer's instructions.
  • the single molecule sequencing platform was exceptionally sensitive for quantitation of small repetitive ncRNA fragments, each of which is scored as a unique read.
  • High level expression of the mouse major satellite was evident in all cells within the primary tumor ( FIG. 2D ), as shown by RNA in situ hybridization (ISH).
  • ISH RNA in situ hybridization
  • FIG. 2E RNA in situ hybridization
  • Clearly defined metastatic lesions to the liver ware strongly positive by RNA ISH, as were individual PDAC cells within the liver parenchyma that otherwise would not have been detected by histopathological analysis ( FIG. 2F ).
  • Low level diffuse expression was evident in liver and lung, as shown by whole mount embryo analysis, but no normal adult or embryonic tissues demonstrated satellite expression comparable to that evident in tumor cells.
  • the index AH284 tumor was analyzed using next generation DNA digital copy number variation (CNV) analysis as described above for genomic DNA sequencing.
  • CNV next generation DNA digital copy number variation
  • ALR alpha RI
  • normal pancreatic tissue has a much higher representation of GSATII, TAR1 and SST1 classes (26.4%, 10.6%, and 8.6% of all satellite reads), while these were a small minority of satellite reads in pancreatic cancers.
  • cancers express high levels of HSATII satellites (4,000 per 10 6 transcripts; 15% of satellite reads), a subtype whose expression is undetectable in normal pancreas ( FIG. 3B ).
  • mice sample reads were aligned to a custom made library for the mouse major satellite (sequence from UCSC genome browser). Human samples were aligned to a custom made reference library for all satellite repeats and LINE-1 variants generated from the Repbase library (Pushkarev et al., Nat Biotech 27, 847 (2009)). In addition, all samples were subjected to the DGE program for transcriptome analysis. Reads were normalized per 10 6 genomic aligned reads for all samples.
  • Line-1 the autonomous retrotransposon Line-1 had the highest expression level in mouse samples of diverse tissue types.
  • Mouse pancreatic tumors have a mean Line-1 expression 30,690 tpm (range 183-120,002), representing an average of 330-fold higher levels compared to Gapdh (Table 5).
  • SCGs satellite Correlated Genes
  • Histone modifications including H3K9 trimethylation (P. A. Cloos, J. Christensen, K. Agger, K. Helin, Genes Dev 22, 1115 (May 1, 2008)), combined with Dicer1 and Piwi-related protein-mediated ncRNA processing (A. A. Aravin, G. J. Hannon, J. Brennecke, Science 318, 761 (Nov. 2, 2007)) have been linked to maintenance of repression of satellite repeats.
  • To search for candidate regulators of satellite derepression in primary tumor specimens we first measured the quantitative DGE of known epigenetic regulators and RNA processing genes in mouse tumors, as a function of increasing major satellite expression.
  • a targeted gene expression analysis of demethylases and RNA processing enzymes was carried out in mouse and human PDAC samples.
  • a list of demethylases and RNA processing enzymes were generated from two recent publications (Cloos et al., Genes Dev 22, 1115 (2008); Aravin et al., Science 318, 761 (2007)).
  • Mouse PDACs with Kras G12D and Tp53 loss were used for this analysis.
  • Mouse tumors were separated into high vs low satellite levels using the median satellite expression (7%). A total of 37 genes were evaluated between high and low satellite tumors and fold change was calculated. Analysis of the population means was compared using the 2-tailed student t-test assuming equal variance.
  • mouse pancreatic tumors with satellite expression above the median had higher expression of the demethylases Hspbap1, Jmjd1B, Jmjd4, Jarid1d, Jmjd3, and Fbxl10 as well as the RNA processing enzyme Dicer1.
  • Hspbap1, Jmjd1B, Jmjd4, Jarid1d, Jmjd3, and Fbxl10 as well as the RNA processing enzyme Dicer1.
  • HSPBAP1, FBXL10 and DICER1 overexpression was also observed in human pancreatic adenocarcinomas (Table 7, p ⁇ 0.05, student t-test).
  • LINE-1 may drive expression of specific cellular mRNAs through its insertion upstream of their transcriptional start sites (T. Kuwabara et al., Nat Neurosci 12, 1097 (September, 2009)) or through alterations in flanking chromatin marks (J. A. Bailey, L. Carrel, A. Chakravarti, E. E. Eichler, Proceedings of the National Academy of Sciences of the United States of America 97, 6634 (Jun. 6, 2000, 2000); D. E.
  • the transcriptional start sites tissues and cell lines were determined (UCSC genome browser (D. Karolchik et al., Nucleic Acids Res 32, D493 (Jan. 1, 2004)) as well as the position of all Line-1 elements in the mouse genome with a threshold of 1 Kbp in length. Line-1 closest distance upstream of the transcriptional start sites of all annotated genes with a minimum expression level of 5 transcripts per million were calculated. Genes were then rank ordered according to the Pearson coefficient for linear regression. Genes were binned in 100 s and plotted by Excel. Randomization of all genes, followed by binning, and plotting was done as a control.
  • LINE-1 sequences within the proximity of cellular transcripts may contribute to their overexpression in primary tumors, in striking correlation with the expression of both LINE-1 and satellite repeats.
  • the consequence of increased expression of these cellular transcripts remains to be defined.
  • the high prevalence of genes linked to stem-like and neurogenic fates, along with the frequency of HOX and zinc finger transcriptional regulators raises the possibility that at least a subset of these may contribute to tumor-related phenotypes.
  • LINE-1 is a Specific and Sensitive Marker of CTCs
  • Satellite levels are most strongly linked with the expression of the autonomous retrotransposon Line-1, which has recently been shown to be a major cause for genomic variation in normal and tumor tissues (J. Berretta, A. Morillon, EMBO Rep 10, 973 (September, 2009); A. Jacquier, Nat Rev Genet 10, 833 (December, 2009); M. Guenatri, D. Bailly, C. Maison, G. Almouzni, J Cell Biol 166, 493 (Aug. 16, 2004)). Aberrant expression of cellular transcripts linked to stem cells and neural tissues is also highly correlated with satellite transcript levels, suggesting alteration of cell fate through derepression of a coordinated epigenetic program.
  • CTCs circulating tumor cells
  • HB herringbone chip
  • EpCAM epithelial cell adhesion molecule
  • RNA was extracted from the devices using the Qiagen RNeasy MinElute kit.
  • Krt Keratins
  • the HSATII satellite is overexpressed in pancreatic cancer and was confirmed to be overexpressed in human preneoplastic pancreatic lesions ( FIG. 6A ) using a branched DNA detection assay (QuantiGene® ViewRNA Assay, Affymetrix).
  • a branched DNA detection assay QuantantiGene® ViewRNA Assay, Affymetrix.
  • Breast cancer samples were also tested for HSATII using this method and were found to have significant expression compared to normal breast tissues.
  • Extension of this technique to potential circulating tumor cells captured on the HB-chip FIG. 6B ) has been accomplished indicating that HSATII may be used as a blood based diagnostic for epithelial cancers.
  • Serum was extracted from the blood of 8 metastatic pancreatic cancer patients by using Ficoll buffy coat method.
  • Serum RNA (cell free RNA), which includes exosomes, was purified using the Trizol method and then purified using Qiagen RNA MinElute columns kits. RNA was then subjected to Helicos DGE sequencing preparation and sequenced on a HeliScope next generation sequencer. Results of this data are summarized in Table 1.
  • HSATII was specific for cancer and GSATII was found to correlate with normal tissues. Therefore the ratio of HSATII to GSATII was evaluated as a marker for identifying cancer burden and potentially an early detection marker. In this case, one patient who had stable disease had the lowest HSATII/GSATII ratio as predicted (see Table 8).
  • Table 8 A total of 8 metastatic cancer patients with clinical status, total satellites, HSATII, and GSATII in transcripts per million aligned to genome (tpm) and the ratio of HSATII/GSATII in cell free RNA sequenced.
  • RNA cell free
  • HSATII and GSATII did not perform as well as expected, though the presence of.
  • other satellites like TAR1 seemed to be better predictors of “cancer” compared to “non-cancer” status as shown in Table 9.
  • Table 9 Average total satellites, HSATII, GSATII, HSATII/GSATII, and TAR1 (tpm) in a total of 8 metastatic PDAC patients and 4 healthy donors with cell free RNA sequenced. Student t-test was used to calculate significance.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Methods for diagnosing cancer based on detecting the presence of increased levels of expression of satellite repeats and/or Line-1.

Description

    CLAIM OF PRIORITY
  • This application claims the benefit of U.S. Provisional Patent Application Ser. No. 61/390,956, filed on Oct. 7, 2010, and 61/493,800, filed on Jun. 6, 2011. The entire contents of the foregoing are hereby incorporated by reference.
  • TECHNICAL FIELD
  • This invention relates to methods of diagnosing cancer, based on detecting the presence of increased levels of expression of satellite repeats and/or Line-1.
  • BACKGROUND
  • Genome-wide sequencing approaches have revealed an increasing set of transcribed non-coding sequences, including “pervasive transcription” by heterochromatic regions of the genome linked to transcriptional silencing and chromosomal integrity (J. Berretta, A. Morillon, EMBO Rep 10, 973 (September, 2009); A. Jacquier, Nat Rev Genet 10, 833 (December, 2009)). In the mouse, heterochromatin is comprised of centric (minor) and pericentric (major) satellite repeats that are required for formation of the mitotic spindle complex and faithful chromosome segregation (M. Guenatri, D. Bailly, C. Maison, G. Almouzni, J Cell Biol 166, 493 (Aug. 16, 2004)), whereas human satellite repeats have been divided into multiple classes with similar functions (J. Jurka et al., Cytogenet Genome Res 110, 462 (2005)). Bidirectional transcription of satellites in yeast maintains silencing of centromeric DNA through the Dicer mediated RNA-induced transcriptional silencing (RITS) and through a recently identified Dicer-independent pathway (M. Halic, D. Moazed, Cell 140, 504 (February 19)), although centromeric satellite silencing mechanisms in mammals are less well defined (A. A. Aravin, G. J. Hannon, J. Brennecke, Science 318, 761 (Nov. 2, 2007)). Accumulation of satellite transcripts in mouse and human cell lines results from defects in DICER1 (C. Kanellopoulou et al., Genes Dev 19, 489 (Feb. 15, 2005); T. Fukagawa et al., Nat Cell Biol 6, 784 (August, 2004)) and from DNA demethylation, heat shock, or the induction of apoptosis (H. Bouzinba-Segard, A. Guais, C. Francastel, Proc Natl Acad Sci USA 103, 8709 (Jun. 6, 2006); R. Valgardsdottir et al., Nucleic Acids Res 36, 423 (February, 2008)). Stress-induced transcription of satellites in cultured cells has also been linked to the activation of retroelements encoding RNA polymerase activity such as LINE-1 (L1TD1) (D. Ugarkovic, EMBO Rep 6, 1035 (November, 2005); D. M. Carone et al., Chromosoma 118, 113 (February, 2009)). Despite these in vitro models, the global expression of repetitive ncRNAs in primary tumors has not been analyzed, due to the bias of microarray platforms toward annotated coding sequences and the specific exclusion of repeat sequences from standard analytic programs.
  • SUMMARY
  • The present invention is based, at least in part, on the identification of massive expression of satellite repeats in tumor cells, and of increased levels of Line-1, e.g., in tumor cells including circulating tumor cells (CTCs). Described herein are methods for diagnosing cancer, e.g., solid malignancies of epithelial origin such as pancreatic, lung, breast, prostate, renal, ovarian or colon cancer, based on the presence of increased levels of expression of satellite repeats and/or Line-1.
  • In a first aspect, the invention provides methods, e.g., in vitro methods, for detecting the presence of cancer in a subject, including determining a level of LINE-1 in a sample from the subject to obtain a test value; and comparing the test value to a reference value, wherein a test value compared to the reference value indicates whether the subject has cancer.
  • In some embodiments, the reference value represents a threshold level of LINE-1, wherein the presence of a level of LINE-1 in the subject that is above the reference value indicates that the subject has cancer, and the presence of a level of LINE-1 in the subject that is below the reference value indicates that the subject is unlikely to have cancer.
  • In a first aspect, the invention provides methods, e.g., in vitro methods, for detecting the presence of cancer in a subject, including determining a level of satellite transcripts in a sample from the subject to obtain a test value; and comparing the test value to a reference value, wherein a test value compared to the reference value indicates whether the subject has cancer.
  • In some embodiments, the satellite transcripts comprise one or more of ALR, HSATII, GSATII, TAR1, and SST1. In some embodiments, the satellite transcript is ALR and/or HSATII, and the presence of a level of ALR and/or HSATII satellite transcripts above the reference level indicates that the subject has a tumor.
  • In some embodiments, the satellite transcript is GSATII, TAR1 and/or SST1, and the presence of a level of GSATII, TAR1 and/or SST1 satellite transcripts below the reference level indicates that subject has a tumor.
  • In another aspect, the invention provides methods, e.g., in vitro methods, for evaluating the efficacy of a treatment for cancer in a subject. The methods include determining a level of LINE-1 in a first sample from the subject to obtain a first value; administering a treatment for cancer to the subject; determining a level of LINE-1 in a subsequent sample obtained from the subject at a later time, to obtain a treatment value; and comparing the first value to the treatment value. A treatment value that is below the first value indicates that the treatment is effective.
  • In yet another aspect, the invention provides methods, e.g., in vitro methods, for evaluating the efficacy of a treatment for cancer in a subject. The methods include determining a level of satellite transcripts in a first sample from the subject to obtain a first value; administering a treatment for cancer to the subject; determining a level of satellite transcripts in a subsequent sample obtained from the subject at a later time, to obtain a treatment value; and comparing the first value to the treatment value, wherein a treatment value that is below the first value indicates that the treatment is effective.
  • In some embodiments, the satellite transcripts comprise one or more of ALR, HSATII, GSATII, TAR1, and SST1.
  • In some embodiments, the first and second samples are known or suspected to comprise tumor cells, e.g., blood samples known or suspected of comprising circulating tumor cells (CTCs), or biopsy samples known or suspected of comprising tumor cells. In some embodiments, the sample comprises free RNA in serum or RNA within exosomes in blood.
  • In some embodiments, the treatment includes administration of a surgical intervention, chemotherapy, radiation therapy, or a combination thereof.
  • In some embodiments of the methods described herein, the subject is a mammal, e.g., a human or veterinary subject, e.g., experimental animal.
  • In some embodiments of the methods described herein, the cancer is a solid tumor of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • In some embodiments, the methods described herein include measuring a level of LINE-1 transcript.
  • In some embodiments of the methods described herein, the level of a LINE-1 transcript or satellite is determined using a branched DNA assay.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
  • Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
  • DESCRIPTION OF DRAWINGS
  • FIG. 1A is a bar graph showing levels of major satellite in percent of all genomic aligned reads among different tumors, cell lines, and tissues. Genotype of primary tumors and cell lines indicated below each tumor type and cell line. (Kras=KrasG12D; Tp53, SMAD4, and APC represent genes deleted)
  • FIG. 1B is a graphical representation of sequence read contributions from major satellite among all primary tumors, cancer cell lines, and normal tissues.
  • FIG. 2A shows the results of Northern blot analysis of three KrasG12D, Tp53lox/+ pancreatic primary tumors (Tumors 1-3) and a stable cell line (CL3) derived from Tumor 3.
  • FIG. 2B shows the results of Northern blot analysis of CL3 before (0) and after (+) treatment with the DNA hypomethylating agent 5-azacitadine (AZA).
  • FIG. 2C shows the results of Northern blot analysis of total RNA from multiple adult and fetal mouse tissues. All Northern blots exposed for approximately 30 minutes.
  • FIG. 2D is a pair of photomicrographs showing the results of RNA in-situ hybridization (ISH) of normal pancreas (left) and primary pancreatic ductal adenocarcinoma (right), hybridized with a 1 kb major satellite repeat probe.
  • FIG. 2E is a set of three photomicrographs showing the results of ISH analysis of preneoplastic PanIN (P) lesion, adjacent to PDAC (T) and normal pancreas (N), showing positive staining in PanIN, with increased expression in full carcinoma. Higher magnification (40×) of PanIN (left) and PDAC (right) lesions.
  • FIG. 2F is a set of three photomicrographs showing marked expression of satellites in PDAC cells metastatic to liver, which itself does not express satellites (left). Large, glandular metastatic tumor deposits are readily identified by standard histological evaluation and stain for satellite (middle). Satellite ISH is sensitive enough to detect micrometastases in liver parenchyma not easily appreciated by standard histological analysis (right; arrowheads). All images at 20× magnification (scale bar=100 μm).
  • FIG. 3A is a bar graph showing the Total satellite expression in human pancreatic ductal adenocarcinoma (PDAC), normal pancreas, other cancers (L—lung, K—kidney, O—ovary, P—prostate), and other normal human tissues (1—fetal brain, 2—brain, 3—colon, 4—fetal liver, 5—liver, 6—lung, 7—kidney, 8—placenta, 9—prostate, and 10—uterus) quantitated by DGE. Satellite expression is shown as transcripts per million aligned to human genome.
  • FIG. 3B is a bar graph showing a breakdown of satellite repeat classes as percent of total satellites in human PDAC (Black, n=15) and normal human tissues (White, n=12) sequenced. Satellites are ordered from highest absolute difference in tumors to highest in normal tissue (left to right). Error bars represents standard error of the mean. Fold differential of top three cancer (left, black bars) and normal (right, white bars) tissue satellite classes shown (Bar graph, center).
  • FIG. 4A shows the results of multiple linear correlation analysis of major satellite to other cellular transcripts among all mouse tumors and normal tissues as depicted by a heat map. X-axis is samples ordered by expression of major satellite and y-axis is genes ordered by linear correlation to major satellite expression. Light grey (High) and dark grey (Low) color is log 2 (reads per million). Major satellite expression as percent genomic aligned reads (y-axis) rank ordered by satellite reads (x-axis) with expanded view of top genes with highest linearity (R≧0.85) with satellite levels.
  • FIG. 4B is a dot graph showing the Median distance of transcriptional start sites of all genes to Line-1 elements ordered by linearity to satellite expression (Dark gray; highest linearity to the left) or by random (Light gray). Plotted by genes binned in 100 s.
  • FIG. 4C is a dot graph showing Top genes with highest linearity (R>0.85) defining satellite correlated genes or SCGs plotted by frequency against distance of transcriptional start site to LINE-1 elements (Dark gray) compared to the expected frequency of these genes (Light gray).
  • FIG. 4D is a set of four photomicrographs showing the results of immunohistochemistry of mouse PDAC (KrasG12D, Tp53 lox/+) for the neuroendocrine marker chromogranin A. Tumors are depicted as a function of increasing chromogranin A staining (dark grey), with the relative level of major satellite expression noted for each tumor at the bottom of each image (percentage of all transcripts)
  • FIG. 5 is a bar graph indicating fold change expression of the indicated genes in CTC Device vs. control device. The subjects were newly diagnosed metastatic pancreatic adenocarcinoma patients. LINE-1 expression was seen in all patients at some point.
  • FIG. 6A is an image of RNA in situ hybridization (RNA-ISH) of human satellite HSATII in a human preneoplastic PanIN (P) lesion with adjacent non-cancerous stroma tissue (N) in formalin fixed paraffin embedded tissue (Top image) and fine needle aspirate biopsy of PDAC (T) and normal adjacent leukocytes (N). (Dark grey dots=HSATII). Scale bar=100 μm
  • FIG. 6B is an image of RNA in situ hybridization of HSATII using Affymetrix ViewRNA of a potential human pancreatic circulating tumor cell captured on the HB-chip. HSATII (lightest areas; yellow in original), DAPI nuclear stain (medium grey areas, blue in original). Scale bar=20 μm.
  • DETAILED DESCRIPTION
  • The present invention is based, at least in part, on the identification of a massive generation of LINE-1 protein and bidirectional ncRNAs from the major satellite repeat in mouse tumor models and from ALR and HSATII satellite repeats in human cancers. The exceptional magnitude of satellite levels in these cancers is unprecedented. This is likely to result from a general derepression of chromosomal marks affecting both satellites and LINE-1 retrotransposons, with proximity to LINE-1 activation potentially affecting the expression of selected cellular mRNAs. Together, the very high expression of satellites may affect chromosomal integrity and genetic stability, while the co-deregulated coding sequences may affect cell fates and biological behavior of cancer cells. In addition, the finding of massive expression of specific satellite subsets in human pancreatic cancer provides a novel biomarker for application in early detection of cancer. Finally, levels of LINE-1 are increased in circulating tumor cells from subjects with newly diagnosed metastatic pancreatic adenocarcinoma. Thus the present methods are useful in the early detection of cancer, and can be used to predict clinical outcomes.
  • Diagnosing Cancer using Transcript Biomarkers
  • The methods described herein can be used to diagnose the presence of, and monitor the efficacy of a treatment for, cancer, e.g., solid tumors of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer, in a subject.
  • As used herein, the term “hyperproliferative” refer to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth. Hyperproliferative disease states may be categorized as pathologic, i.e., characterizing or constituting a disease state, or may be categorized as non-pathologic, i.e., a deviation from normal but not associated with a disease state. The term is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. A “tumor” is an abnormal growth of hyperproliferative cells. “Cancer” refers to pathologic disease states, e.g., characterized by malignant tumor growth.
  • As demonstrated herein, the presence of cancer, e.g., solid tumors of epithelial origin, e.g., as defined by the ICD-O (International Classification of Diseases-Oncology) code (revision 3), section (8010-8790), e.g., early stage cancer, is associated with the presence of a massive levels of satellite due to increase in transcription and processing of satellite repeats in pancreatic cancer cells, and of increased levels of LINE-1 expression in circulating tumor cells. Thus the methods can include the detection of expression levels of satellite repeats in a sample comprising cells known or suspected of being tumor cells, e.g., cells from solid tumors of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer cells. Alternatively or in addition, the methods can include the detection of increased levels of LINE-1 in a sample, e.g., a sample known or suspected of including tumor cells, e.g., circulating tumor cells (CTCs), e.g., using a microfluidic device as described herein.
  • Cancers of epithelial origin can include pancreatic cancer (e.g., pancreatic adenocarcinoma or intraductal papillary mucinous carcinoma (IPMN, pancreatic mass)), lung cancer (e.g., non-small cell lung cancer), prostate cancer, breast cancer, renal cancer, ovarian cancer, or colon cancer. For example, the present methods can be used to distinguish between benign IPMN, for which surveillance is the standard treatment, and malignant IPMN, which require resection, a procedure associated with significant morbidity and a small but significant possibility of death. In some embodiments, in a subject diagnosed with IPMN, the methods described herein can be used for surveillance/monitoring of the subject, e.g., the methods can be repeated at selected intervals (e.g., every 3, 6, 12, or 24 months) to determine whether a benign IPMN has become a malignant IPMN warranting surgical intervention. In addition, in some embodiments the methods can be used to distinguish bronchioloalveolar carcinomas from reactive processes (e.g., postpneumonic reactive processes) in samples from subjects suspected of having non-small cell lung cancer. In some embodiments, in a sample from a subject who is suspected of having breast cancer, the methods can be used to distinguish ductal hyperplasia from atypical ductal hyperplasia and ductal carcinoma in situ (DCIS). The two latter categories receive resection/radiation; the former does not require intervention. In some embodiments, in subjects suspected of having prostate cancer, the methods can be used to distinguish between atypical small acinar proliferation and malignant cancer. In some embodiments, in subjects suspected of having bladder cancer, the methods can be used to detect, e.g., transitional cell carcinoma (TCC), e.g., in urine specimens. In some embodiments, in subjects diagnosed with Barrett's Esophagus (Sharma, N Engl J Med. 2009, 24; 361(26):2548-56. Erratum in: N Engl J Med. 2010 Apr. 15; 362(15):1450), the methods can be used for distinguishing dysplasia in Barrett's esophagus from a reactive process. The clinical implications are significant, as a diagnosis of dysplasia demands a therapeutic intervention. Other embodiments include, but are not limited to, diagnosis of well differentiated hepatocellular carcinoma, ampullary and bile duct carcinoma, glioma vs. reactive gliosis, melanoma vs. dermal nevus, low grade sarcoma, and pancreatic endocrine tumors, inter alia.
  • Therefore, included herein are methods for diagnosing cancer, e.g., tumors of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer, in a subject. In some embodiments, the methods include obtaining a sample from a subject, and evaluating the presence and/or level of LINE-1 or satellites in the sample, and comparing the presence and/or level with one or more references, e.g., a control reference that represents a normal level of LINE-1 or satellites, e.g., a level in an unaffected subject or a normal cell from the same subject, and/or a disease reference that represents a level of LINE-1 or satellites associated with cancer, e.g., a level in a subject having pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • The present methods can also be used to determine the stage of a cancer, e.g., whether a sample includes cells that are from a precancerous lesion, an early stage tumor, or an advanced tumor. For example, the present methods can be used to determine whether a subject has a precancerous pancreatic, breast, or prostate lesion. Where the markers used are LINE-1, or satellite transcript ALR and/or HSATII, increasing levels are correlated with advancing stage. For satellite transcripts GSATII, TAR1 and/or SST1, decreasing levels are correlated with increasing stage. Additionally, levels of LINE-1 and satellite ALR and/or HSATII may be prognostic and predictive to clinical outcomes.
  • Samples
  • In some embodiments of the present methods, the sample is or includes blood, serum, and/or plasma, or a portion or subfraction thereof, e.g., free RNA in serum or RNA within exosomes in blood. In some embodiments, the sample comprises (or is suspected of comprising) CTCs. In some embodiments, the sample is or includes urine or a portion or subfraction thereof. In some embodiments, the sample includes known or suspected tumor cells, e.g., is a biopsy sample, e.g., a fine needle aspirate (FNA), endoscopic biopsy, or core needle biopsy; in some embodiments the sample comprises cells from the pancreatic, lung, breast, prostate, renal, ovarian or colon of the subject. In some embodiments, the sample comprises lung cells obtained from a sputum sample or from the lung of the subject by brushing, washing, bronchoscopic biopsy, transbronchial biopsy, or FNA, e.g., bronchoscopic, fluoroscopic, or CT-guided FNA (such methods can also be used to obtain samples from other tissues as well). In some embodiments, the sample is frozen, fixed and/or permeabilized, e.g., is an formalin-fixed paraffin-embedded (FFPE) sample.
  • Satellite Expression Levels
  • In some embodiments, the level of satellite transcripts is detected, e.g., in a sample known or suspected to include tumor cells. In some embodiments, the level of satellite transcripts in a known or suspected tumor cell, e.g., a test cell, is compared to a reference level.
  • In some embodiments, the methods include detecting levels of alpha (ALR) satellite transcripts (D. Lipson et al., Nat Biotechnol 27, 652 (July, 2009)) or HSATII satellite transcripts (J. Jurka et al., Cytogenet Genome Res 110, 462 (2005)); in some embodiments, those levels are compared to a reference. In some embodiments, the reference level is a level of ALR and/or HSATII satellite transcripts in a normal (non-cancerous) cell, e.g., a normal cell from the same subject, or a reference level determined from a cohort of normal cells; the presence of levels of ALR and/or HSATII in the test cell above those in the normal cell indicate that the test cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer). In some embodiments, the reference level of ALR and/or HSATII transcripts is a threshold level, and the presence of a level of ALR and/or HSATII satellite transcripts above the threshold level indicates that the cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • In some embodiments, the methods include detecting levels of GSATII, TAR1 and/or SST1 transcripts; in some embodiments, those levels are compared to a reference. In some embodiments, the reference level is a level of GSATII, TAR1 and/or SST1 satellite transcripts in a normal (non-cancerous) cell, e.g., a normal cell from the same subject, or a reference level determined from a cohort of normal cells; the presence of levels of GSATII, TAR1 and/or SST1 in the test cell below those in the normal cell indicate that the test cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer). In some embodiments, the reference level of GSATII, TAR1 and/or SST1 transcripts is a threshold level, and the presence of a level of GSATII, TAR1 and/or SST1 satellite transcripts below the threshold level indicates that the cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • In some embodiments, the levels of the satellite transcripts are normalized to a relatively non-variant transcript such as GAPDH, actin, or tubulin, e.g., the level of expression of the satellite is compared to the non-variant transcript. For example, a ratio of expression levels can be calculated, and the ratio can be compared to the ratio in a normal (non-cancerous) cell. For example, in some embodiments the presence of a ratio of ALR:GAPDH of over 10:1, e.g., over 50:1, over 100:1, or over 150:1, indicates that the test cell is a cancer cell; in some embodiments the presence of a ratio of ALR:GAPDH of about 3:1 or 5:1 indicates that the test cell is a normal cell. In some embodiments, the presence of a ratio of HSATII satellites:GAPDH transcripts of over 10:1, e.g., 20:1, 30:1, 40:1, or 45:1, indicates that the test cell is a cancer cell. In some embodiments, the presence of significant (e.g., more than about 100 transcripts per million aligned) levels of HSATII indicates that the test cell is a cancer cell. In some embodiments, the absence or presence of very low levels (e.g., less than about 20 transcripts per million aligned) of HSATII indicates that the test cell is a normal cell.
  • Below are exemplary reference sequences that can be used for ALR (including its variants) and HSATII transcripts:
  • >ALR SAT Homo sapiens
    (SEQ ID NO: 1)
    aattctcagtaacttccttgtgttgtgtgtattcaactcacagagttgaacgatcctttacacagagcag
    acttgaaacactctttttgtggaatttgcaagtggagatttcagccgctttgaggtcaatggtagaatag
    gaaatatcttcctatagaaactagacagaat
    >ALR1 SAT Homo sapiens
    (SEQ ID NO: 2)
    tcattctcagaaactrctttgtgatgtgtgcrttcaactcacagagtttaacctttcttttgatagagca
    gtttggaaacactctgtttgtaaagtctgcaagtggatatttggacctctttgaggccttcgttggaaac
    gggatttcttcatataatgctagacagaaga
    >ALR2 SAT Homo sapiens
    (SEQ ID NO: 3)
    agctttctgagaaactgctttgtgatgtgtgcattcatctcacagagttaaacctttcttttgattcagc
    agtttggaaacactgtttttgtagaatctgtgaagggatatttgggagctcattgaggcctatggtgaaa
    aagaaaatatcttcagataaaaactagaaggaagctatc
    >ALRa SAT Primates
    (SEQ ID NO: 4)
    ctatctgagaaactgctttgtgatgtgtgcattcatctcacagagttaaacctttcttttgattcagcag
    tttggaaacactgtttttgtagaatctgcgaagggacatttgggagctcattgaggcctatggtgaaaaa
    gcgaatatccccagataaaaactagaaagaag
    >ALRa_ SAT Primates
    (SEQ ID NO: 5)
    ttgtagaatctgcgaagggacatttgggagctcattgaggcctatggtgaaaaagcgaatatccccagat
    aaaaactagaaagaagctatctgagaaactgctttgtgatgtgtgcattcatctcacagagttaaacctt
    tcttttgattcagcagtttggaaacactgttt
    >ALRb SAT Primates
    (SEQ ID NO: 6)
    ttgtggaatttgcaagtggagatttcaagcgctttgaggccaawnktagaaaaggaaatatcttcgtata
    aaaactagacagaataattctcagtaacttctttgtgttgtgtgtattcaactcacagagttgaaccttc
    ctttagacagagcagatttgaaacactcttt
    >ALR_ SAT Primates
    (SEQ ID NO: 7)
    ttgtagaatctgcaagtggatatttggasckctttgaggmcttcgktggaaacgggaatatcttcacata
    aaaactagacagaagcattctcagaaacttctttgtgatgtttgcattcaactcacagagttgaacmttc
    cttttgatagagcagttttgaaacactcttt
    >HSATII SAT Primates
    (SEQ ID NO: 8)
    ccattcgattccattcgatgattccattcgattccattcgatgatgattccattcgattccattcgatga
    ttccattcgattccattcgatgatgattccattcgattccattcgatgattccattcgattccattcgat
    gatgattccattcgattccattcgatgatt
  • Line-1 Levels
  • Long interspersed nucleotide element (LINE) non-LTR retrotransposons (Singer, Cell 28 (3): 433-4 (1982)) are a group of genetic elements that are found in large numbers in eukaryotic genomes, and generate insertion mutations, contribute to genomic instability and innovation, and can alter gene expression.
  • The canonical, full-length LINE-1 element is about 6 kilobases (kb) in length and includes a 5′ untranslated region (UTR) with an internal RNA polymerase II promoter (Swergold, Mol Cell Biol. 10(12):6718-29 (1990)), two open reading frames (designated ORF1 and ORF2) and a 3′ UTR containing a polyadenylation signal ending with an oligo dA-rich tail of variable length (Babushok and Kazazian, Hum Mutat. 28(6):527-39 (2007)). Although there are over 500,000 L1 elements inserted in the human genome, only about 80-100 copies are retrotransposition-competent (Brouha et al., Proc Natl Acad Sci USA. 100(9):5280-5. (2003)). For additional details, see Cordaux and Batzer, Nat Rev Genet. 10(10): 691-703 (2009)).
  • Exemplary LINE-1 sequences include GenBank Ref. No. NM001164835.1 (nucleic acid) and NP001158307.1 (protein) for variant (1); and GenBank Ref. No. NM019079.4 (nucleic acid) and NP061952.3 (protein) for variant 2, which is the shorter transcript. Variant 2 differs in the 5′ UTR compared to variant 1, but both variants 1 and 2 encode the same protein. See also Gene ID: 54596.
  • In some embodiments, the methods for diagnosing cancer described herein include determining a level of LINE-1 mRNA in a cell, e.g., in CTCs present in blood of a subject to obtain a LINE-1 value, and comparing the value to an appropriate reference value, e.g., a value that represents a threshold level, above which the subject can be diagnosed with cancer. The reference can also be a range of values, e.g., that indicate severity or stage of the cancer in the subject. A suitable reference value can be determined by methods known in the art.
  • In some embodiments, the reference level is a level of LINE-1 transcripts in a normal (non-cancerous) cell, e.g., a normal cell from the same subject, or a reference level determined from a cohort of normal cells; the presence of levels of LINE-1 in the test cell above those in the normal cell indicate that the test cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer). In some embodiments, the reference level of LINE-1 transcripts is a threshold level, and the presence of a level of LINE-1 transcripts above the threshold level indicates that the cell is a tumor cell (e.g., the subject from whom the test cell came has or can be diagnosed with cancer).
  • Methods of Detection
  • Any methods known in the art can be used to detect and/or quantify levels of a biomarker as described herein. For example, the level of a satellite transcript or LINE-1 mRNA (transcript) can be evaluated using methods known in the art, e.g., Northern blot, RNA in situ hybridization (RNA-ISH), RNA expression assays, e.g., microarray analysis, RT-PCR, deep sequencing, cloning, Northern blot, and quantitative real time polymerase chain reaction (qRT-PCR). Analytical techniques to determine RNA expression are known. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd Ed., Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (2001).
  • In some embodiments, the level of the LINE-1 protein is detected. The presence and/or level of a protein can be evaluated using methods known in the art, e.g., using quantitative immunoassay methods such as enzyme linked immunosorbent assays (ELISAs), immunoprecipitations, immunofluorescence, immunohistochemistry, enzyme immunoassay (EIA), radioimmunoassay (RIA), and Western blot analysis.
  • In some embodiments, the methods include contacting an agent that selectively binds to a biomarker, e.g., to a satellite transcript or LINE-1 mRNA or protein (such as an oligonucleotide probe, an antibody or antigen-binding portion thereof) with a sample, to evaluate the level of the biomarker in the sample. In some embodiments, the agent bears a detectable label. The term “labeled,” with regard to an agent encompasses direct labeling of the agent by coupling (i.e., physically linking) a detectable substance to the agent, as well as indirect labeling of the agent by reactivity with a detectable substance. Examples of detectable substances are known in the art and include chemiluminescent, fluorescent, radioactive, or colorimetric labels. For example, detectable substances can include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride, quantum dots, or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125I, 131I, 35S or 3H. In general, where a protein is to be detected, antibodies can be used. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or an antigen-binding fragment thereof (e.g., Fab or F(ab′)2) can be used.
  • In some embodiments, high throughput methods, e.g., protein or gene chips as are known in the art (see, e.g., Ch. 12, “Genomics,” in Griffiths et al., Eds. Modern genetic Analysis, 1999, W. H. Freeman and Company; Ekins and Chu, Trends in Biotechnology, 1999; 17:217-218; MacBeath and Schreiber, Science 2000, 289(5485):1760-1763; Simpson, Proteins and Proteomics: A Laboratory Manual, Cold Spring Harbor Laboratory Press; 2002; Hardiman, Microarrays Methods and Applications: Nuts & Bolts, DNA Press, 2003), can be used to detect the presence and/or level of satellites or LINE-1.
  • In some embodiments, the methods include using a modified RNA in situ hybridization technique using a branched-chain DNA assay to directly detect and evaluate the level of biomarker mRNA in the sample (see, e.g., Luo et al., U.S. Pat. No. 7,803,541B2, 2010; Canales et al., Nature Biotechnology 24(9):1115-1122 (2006); Nguyen et al., Single Molecule in situ Detection and Direct Quantification of miRNA in Cells and FFPE Tissues, poster available at panomics.com/index.php?id=product87). A kit for performing this assay is commercially-available from Affymetrix (ViewRNA).
  • Detection of LINE-1 and Satellite Transcripts in CTCs
  • In some embodiments, microfluidic (e.g., “lab-on-a-chip”) devices can be used in the present methods. Such devices have been successfully used for microfluidic flow cytometry, continuous size-based separation, and chromatographic separation. In general, methods in which expression of satellites or LINE-1 is detected in circulating tumor cells (CTCs) can be used for the early detection of cancer, e.g., early detection of tumors of epithelial origin, e.g., pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • The devices can be used for separating CTCs from a mixture of cells, or preparing an enriched population of CTCs. In particular, such devices can be used for the isolation of CTCs from complex mixtures such as whole blood.
  • A variety of approaches can be used to separate CTCs from a heterogeneous sample. For example, a device can include an array of multiple posts arranged in a hexagonal packing pattern in a microfluidic channel upstream of a block barrier. The posts and the block barrier can be functionalized with different binding moieties. For example, the posts can be functionalized with anti-EPCAM antibody to capture circulating tumor cells (CTCs); see, e.g., Nagrath et al., Nature 450:1235-1239 (2007), optionally with downstream block barriers functionalized with to capture LINE-1 nucleic acids or proteins, or satellites. See, e.g., (S. Maheswaran et al., N Engl J Med. 359, 366 (Jul. 24, 2008); S. Nagrath et al., Nature. 450, 1235 (Dec. 20, 2007); S. L. Stott et al., Sci Transl Med 2, 25ra23 (March 31)) and the applications and references listed herein.
  • Processes for enriching specific particles from a sample are generally based on sequential processing steps, each of which reduces the number of undesired cells/particles in the mixture, but one processing step may suffice in some embodiments. Devices for carrying out various processing steps can be separate or integrated into one microfluidic system. The devices include devices for cell/particle binding, devices for cell lysis, devices for arraying cells, and devices for particle separation, e.g., based on size, shape, and/or deformability or other criteria. In certain embodiments, processing steps are used to reduce the number of cells prior to introducing them into the device or system. In some embodiments, the devices retain at least 75%, e.g., 80%, 90%, 95%, 98%, or 99% of the desired cells compared to the initial sample mixture, while enriching the population of desired cells by a factor of at least 100, e.g., by 1000, 10,000, 100,000, or even 1,000,000 relative to one or more non-desired cell types.
  • Some devices for the separation of particles rely on size-based separation with or without simultaneous cell binding. Some size-based separation devices include one or more arrays of obstacles that cause lateral displacement of CTCs and other components of fluids, thereby offering mechanisms of enriching or otherwise processing such components. The array(s) of obstacles for separating particles according to size typically define a network of gaps, wherein a fluid passing through a gap is divided unequally into subsequent gaps. Both sieve and array sized-based separation devices can incorporate selectively permeable obstacles as described above with respect to cell-binding devices.
  • Devices including an array of obstacles that form a network of gaps can include, for example, a staggered two-dimensional array of obstacles, e.g., such that each successive row is offset by less than half of the period of the previous row. The obstacles can also be arranged in different patterns. Examples of possible obstacle shapes and patterns are discussed in more detail in WO 2004/029221.
  • In some embodiments, the device can provide separation and/or enrichment of CTCs using array-based size separation methods, e.g., as described in U.S. Pat. Pub. No. 2007/0026413. In general, the devices include one or more arrays of selectively permeable obstacles that cause lateral displacement of large particles such as CTCs and other components suspended in fluid samples, thereby offering mechanisms of enriching or otherwise processing such components, while also offering the possibility of selectively binding other, smaller particles that can penetrate into the voids in the dense matrices of nanotubes that make up the obstacles. Devices that employ such selectively permeable obstacles for size, shape, or deformability based enrichment of particles, including filters, sieves, and enrichment or separation devices, are described in International Publication Nos. 2004/029221 and 2004/113877, Huang et al. Science 304:987-990 (2004), U.S. Publication No. 2004/0144651, U.S. Pat. Nos. 5,837,115 and 6,692,952, and U.S. Application Nos. 60/703,833, 60/704,067, and Ser. No. 11/227,904; devices useful for affinity capture, e.g., those described in International Publication No. 2004/029221 and U.S. application Ser. No. 11/071,679; devices useful for preferential lysis of cells in a sample, e.g., those described in International Publication No. 2004/029221, U.S. Pat. No. 5,641,628, and U.S. Application No. 60/668,415; devices useful for arraying cells, e.g., those described in International Publication No. 2004/029221, U.S. Pat. No. 6,692,952, and U.S. application Ser. Nos. 10/778,831 and 11/146,581; and devices useful for fluid delivery, e.g., those described in U.S. application Ser. Nos. 11/071,270 and 11/227,469. Two or more devices can be combined in series, e.g., as described in International Publication No. WO 2004/029221. All of the foregoing are incorporated by reference herein.
  • In some embodiments, a device can contain obstacles that include binding moieties, e.g., monoclonal anti-EpCAM antibodies or fragments thereof, that selectively bind to particular cell types, e.g., cells of epithelial origin, e.g., tumor cells. All of the obstacles of the device can include these binding moieties; alternatively, only a subset of the obstacles include them. Devices can also include additional modules, e.g., a cell counting module or a detection module, which are in fluid communication with the microfluidic channel device. For example, the detection module can be configured to visualize an output sample of the device.
  • In one example, a detection module can be in fluid communication with a separation or enrichment device. The detection module can operate using any method of detection disclosed herein, or other methods known in the art. For example, the detection module includes a microscope, a cell counter, a magnet, a biocavity laser (see, e.g., Gourley et al., J. Phys. D: Appl. Phys., 36: R228-R239 (2003)), a mass spectrometer, a PCR device, an RT-PCR device, a microarray, RNA in situ hybridization system, or a hyperspectral imaging system (see, e.g., Vo-Dinh et al., IEEE Eng. Med. Biol. Mag., 23:40-49 (2004)). In some embodiments, a computer terminal can be connected to the detection module. For instance, the detection module can detect a label that selectively binds to cells, proteins, or nucleic acids of interest, e.g., LINE-1 DNA, mRNA, or proteins, or satellite DNA or mRNA.
  • In some embodiments, the microfluidic system includes (i) a device for separation or enrichment of CTCs; (ii) a device for lysis of the enriched CTCs; and (iii) a device for detection of LINE-1 DNA, mRNA, or proteins, or satellite DNA or mRNA.
  • In some embodiments, a population of CTCs prepared using a microfluidic device as described herein is used for analysis of expression of LINE-1 and/or satellites using known molecular biological techniques, e.g., as described above and in Sambrook, Molecular Cloning: A Laboratory Manual, Third Edition (Cold Spring Harbor Laboratory Press; 3rd edition (Jan. 15, 2001)); and Short Protocols in Molecular Biology, Ausubel et al., eds. (Current Protocols; 52 edition (Nov. 5, 2002)).
  • In general, devices for detection and/or quantification of expression of satellites or LINE-1 in an enriched population of CTCs are described herein and can be used for the early detection of cancer, e.g., tumors of epithelial origin, e.g., early detection of pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
  • Methods of Monitoring Disease Progress or Treatment Efficacy
  • In some embodiments, once it has been determined that a person has cancer, or has an increased risk of developing cancer, then a treatment, e.g., as known in the art, can be administered. The efficacy of the treatment can be monitored using the methods described herein; an additional sample can be evaluated after (or during) treatment, e.g., after one or more doses of the treatment are administered, and a decrease in the level of LINE-1, and/or ALR and/or HSATII satellites expression, or in the number of LINE-1-, and/or ALR and/or HSATII satellite-expressing cells in a sample, would indicate that the treatment was effective, while no change or an increase in the level of LINE-1, and/or ALR and/or HSATII satellite expression or LINE-1-, and/or ALR and/or HSATII satellite-expressing cells would indicate that the treatment was not effective (the converse would of course be true for levels of GSATII, TAR1 and/or SST1 satellites). The methods can be repeated multiple times during the course of treatment, and/or after the treatment has been concluded, e.g., to monitor potential recurrence of disease.
  • In some embodiments, e.g., for subjects who have been diagnosed with a benign condition that could lead to cancer, subjects who have been successfully treated for a cancer, or subjects who have an increased risk of cancer, e.g., due to a genetic predisposition or environmental exposure to cancer-causing agents, the methods can be repeated at selected intervals, e.g., at 3, 6, 12, or 24 month intervals, to monitor the disease in the subject for early detection of progression to malignancy or development of cancer in the subject.
  • EXAMPLES
  • The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.
  • Example 1 Major Satellite Levels are Massively Elevated in Tumor Tissues Compared to Cell Lines and Normal Tissue
  • The next generation digital gene expression (DGE) application from Helicos BioSciences (D. Lipson et al., Nat Biotechnol 27, 652 (July, 2009)) was utilized to compare expression of tumor markers in primary cancers and their derived metastatic precursors. We first determined DGE profiles of primary mouse pancreatic ductal adenocarcinoma (PDAC) generated through tissue-targeted expression of activated Kras and loss of Tp53 (KrasG12D, Tp53lox/+) (N. Bardeesy et al., Proc Natl Acad Sci USA 103, 5947 (Apr. 11, 2006)). These tumors are histopathological and genetic mimics of human PDAC, which exhibits virtually universal mutant KRAS (>90% of cases) and loss of TP53 (50-60%).
  • Mice with pancreatic cancer of different genotypes were bred as previously described in the Bardeesy laboratory (Bardeesy et al., Proc Natl Acad Sci USA 103, 5947 (2006)). Normal wild type mice were purchased from Jackson laboratories. Animals were euthanized as per animal protocol guidelines. Pancreatic tumors and normal tissue were extracted sterilely and then flash frozen with liquid nitrogen. Tissues were stored at −80° C. Cell lines were generated fresh for animals AH367 and AH368 as previously described (Aguirre et al., Genes Dev 17, 3112 (2003)) and established cell lines were cultured in RPMI-1640+10% FBS+1% Pen/Strep (Gibco/Invitrogen). Additional mouse tumors from colon and lung were generously provided by Kevin Haigis (Massachusetts General Hospital) and Kwok-Kin Wong (Dana Farber Cancer Institute).
  • Fresh frozen tissue was pulverized with a sterile pestle in a microfuge tube on dry ice. Cell lines were cultured and fresh frozen in liquid nitrogen prior to nucleic acid extraction. RNA and DNA from cell lines and fresh frozen tumor and normal tissues were all processed in the same manner. RNA was extracted using the TRIzol® Reagent (Invitrogen) per manufacturer's specifications. DNA from tissue and cell lines was extracted using the QIAamp Mini Kit (QIAGEN) per manufacturer's protocol.
  • Purified RNA was subjected to Digital Gene Expression (DGE) sample prepping and analysis on the HeliScope™ Single Molecule Sequencer from Helicos BioSciences. This method has been previously described (Lipson et al., Nat Biotechnol 27, 652 (2009)). Briefly, Single stranded cDNA was reverse transcribed from RNA with a dTU25V primer and the Superscript III cDNA synthesis kit (Invitrogen). RNA was digested and single stranded cDNA was purified using a solid phase reversible immobilization (SPRI) technique with Agencourt® AMPure® magnetic beads. Single stranded cDNA was denatured and then a poly-A tail was added to the 3′ end using terminal transferase (New England Biolabs).
  • Purified DNA was subjected to the DNA sequencing sample prepping protocol from Helicos that has been previously described (Pushkarev, N. F. Neff, S. R. Quake, Nat Biotech 27, 847 (2009)). Briefly, genomic DNA was sheared with a Covaris S2 acoustic sonicator producing fragments averaging 200 bps and ranging from 100-500 bps. Sheared DNA was then cleaned with SPRI. DNA was then denatured and a poly-A tail was added to the 3′ end using terminal transferase.
  • Tailed cDNA or DNA were then hybridized to the sequencing flow cell followed by “Fill and Lock” and single molecule sequencing. Gene expression sequence reads were then aligned to the known human or mouse transcriptome libraries using the DGE program. Genomic DNA sequence reads were aligned to the mouse genome and counted to determine copy number of the major mouse satellite (CNV).
  • The first mouse pancreatic tumor analyzed, AH284, was remarkable in that DGE sequences displayed a 48-52% discrepancy with the annotated mouse transcriptome, compared with a 3-4% difference for normal liver transcripts from the same mouse. Nearly all the discrepant sequences mapped to the pericentric (major) mouse satellite repeat. The satellite transcript accounts for ˜49% (495,421 tpm) of all cellular transcripts in the tumor, compared with 0.02-0.4% (196-4,115 tpm) in normal pancreas or liver (Table 1).
  • TABLE 1
    Total genomic aligned reads with breakdown of major
    satellite and transcriptome reads. Percentage of
    total genomic aligned reads in parentheses
    Major Satellite
    Total Reads Reads Transcriptome
    Pancreatic 18,063,363 8,460,135 (47%)   1,726,768 (10%)
    Tumor
    Normal Liver 2,270,669 8,973 (0.4%) 1,718,489 (75%)
    Normal 492,301 2,026 (0.4%)   63,160 (13%)
    Pancreas

    Satellite sequence reads were found in both sense and anti-sense directions and are absent from poly-A purified RNA. Tumor AH284 therefore contained massive amounts of a non-polyadenylated dsRNA element, quantitatively determined as >100-fold increased over that present in normal tissue from the same animal. By way of comparison, the levels of satellite transcripts in tumor tissues were about 8,000-fold higher than the abundant mRNA Gapdh. A second independent pancreatic tumor nodule from the same mouse showed a lower, albeit still greatly elevated, level of satellite transcript (4.5% of total cellular transcripts).
  • Analysis of 4 additional pancreatic tumors from (KrasG12D, Tp53lox/+) mice and 4 mice with an alternative pancreatic tumorigenic genotype (KrasG12D, SMAD4lox/lox) revealed increased satellite expression in 6/8 additional tumors (range 1-15% of all cellular transcripts). In 2/3 mouse colon cancer tumors (KrasG12D, APClox/−) and 2/2 lung cancers (KrasG12D, Tp53lox/lox), satellite expression level ranged from 2-16% of all cellular transcripts. In total, 12/15 (80%) independent mouse tumors had greatly increased levels of satellite expression, compared to normal mouse tissues (FIG. 1A, Table 2).
  • TABLE 2
    Total genomic reads and percentage of reads aligning to transcriptome and major
    satellite among multiple mouse tumors, cell lines, and normal tissues.
    %
    Total % Major
    Genomic Transcriptome Satellite
    Mouse ID Tissue Type Genotype Reads Reads Reads
    AH284 Rep 1 Pancreatic Cancer KrasG12D, Tp53lox/+ 18,063,363 9.56% 46.84%
    AH284 Rep 2 Pancreatic Cancer KrasG12D, Tp53lox/+ 16,948,693 10.15% 49.54%
    AH284-2* Pancreatic Cancer KrasG12D, Tp53lox/+ 1,613,592 48.67% 4.78%
    AH287 Pancreatic Cancer KrasG12D, Tp53lox/+ 2,227,850 54.70% 0.07%
    AH288 Pancreatic Cancer KrasG12D, Tp53lox/+ 6,780,821 26.57% 14.79%
    AH291 Pancreatic Cancer KrasG12D, Tp53lox/+ 1,388,906 43.12% 1.22%
    AH294 Pancreatic Cancer KrasG12D, Tp53lox/+ 969,896 37.20% 3.73%
    AH323 Pancreatic Cancer KrasG12D, SMAD4lox/lox 1,887,663 72.73% 0.29%
    AH346 Pancreatic Cancer KrasG12D, SMAD4lox/lox 1,291,648 32.92% 6.07%
    AH347 Pancreatic Cancer KrasG12D, SMAD4lox/lox 1,634,314 38.94% 8.59%
    AH348 Pancreatic Cancer KrasG12D, SMAD4lox/lox 2,030,197 45.84% 5.61%
    Colon 1 Colon Cancer - 1 KrasG12D, APClox/lox 2,954,930 77.49% 0.07%
    Colon 1 Colon Cancer - 2 KrasG12D, APClox/lox 985,510 53.13% 6.27%
    Colon 1 Colon Cancer - 3 KrasG12D, APClox/lox 1,017,319 30.71% 16.02%
    KN2128 Lung Cancer KrasG12D, Tp53lox/lox 2,233,183 60.78% 2.66%
    KN2199 Lung Cancer KrasG12D, Tp53lox/lox 1,653,948 43.21% 5.37%
    AH323 PDAC Cell Line KrasG12D, SMAD4lox/lox 1,958,108 83.13% 0.02%
    AH324 PDAC Cell Line KrasG12D, Tp53lox/+ 3,301,108 86.32% 0.04%
    NB490 PDAC Cell Line KrasG12D, Tp53lox/lox 15,378,802 76.85% 0.03%
    AH284 Rep 1 Matched Normal Liver KrasG12D, Tp53lox/+ 2,270,669 75.68% 0.40%
    AH284 Rep 2 Matched Normal Liver KrasG12D, Tp53lox/+ 1,627,749 56.59% 0.34%
    AH284-2* Matched Normal Liver KrasG12D, Tp53lox/+ 644,316 41.10% 0.31%
    Colon 1 Matched Normal Liver KrasG12D, APClox/lox 1,536,346 86.53% 0.02%
    Normal 1 Normal Pancreas WT 247,582 14.49% 0.41%
    Normal 2 Normal Pancreas WT 244,719 11.15% 0.41%
    *AH284-2 was RNA extraction from a different part of the pancreatic tumor and liver
  • Of note, the composite distribution of all RNA reads among coding, ribosomal and other non-coding transcripts showed significant variation between primary tumors and normal tissues (FIG. 1A), suggesting that the global cellular transcriptional machinery is affected by the massive expression of satellite transcripts in primary tumors. Immortalized cell lines established from 3 primary pancreatic tumors displayed minimal expression of satellite repeats, suggesting either negative selection pressure during in vitro proliferation or reestablishment of stable satellite silencing mechanisms under in vitro culture conditions (FIG. 1A). Of note in primary tumors overexpressing satellites, the composite distribution of all RNA reads among coding, ribosomal and other non-coding transcripts shows significant variation with that of normal tissues (FIG. 1B), suggesting that the cellular transcriptional machinery is affected by the massive expression of satellite transcripts in these tumors.
  • Example 2 Major Satellite Transcripts are of Various Sizes Depending on Tissue Type and Expression Levels are Linked to Genomic Methylation and Amplification
  • Northern blot analysis of mouse primary pancreatic tumors was carried out as follows. Northern Blot was performed using the NorthernMax-Gly Kit (Ambion). Total RNA (10 ug) was mixed with equal volume of Glyoxal Load Dye (Ambion) and incubated at 50° C. for 30 min. After electrophoresis in a 1% agarose gel, RNA was transferred onto BrightStar-Plus membranes (Ambion) and crosslinked with ultraviolet light. The membrane was prehybridized in ULTRAhyb buffer (Ambion) at 68° C. for 30 min. The mouse RNA probe (1100 bp) was prepared using the MAXIscript Kit (Ambion) and was nonisotopically labeled using the BrightStar Psoralen-Biotin Kit (Ambion) according to the manufacturer's instructions. Using 0.1 nM probe, the membrane was hybridized in ULTRAhyb buffer (Ambion) at 68° C. for 2 hours. The membrane was washed with a Low Stringency wash at room temperature for 10 min, followed by two High Stringency washes at 68° C. for 15 min. For nonisotopic chemiluminescent detection, the BrightStar BioDetect Kit was used according to the manufacturer's instructions.
  • The results demonstrated that the major satellite-derived transcripts range from 100 bp to 2.5 kb (FIG. 2A), consistent with the predicted cleavage of a large primary transcript comprised of multiple tandem repeats by Dicer1 (C. Kanellopoulou et al., Genes Dev 19, 489 (Feb. 15, 2005); T. Fukagawa et al., Nat Cell Biol 6, 784 (August, 2004); H. Bouzinba-Segard, A. Guais, C. Francastel, Proc Natl Acad Sci USA 103, 8709 (Jun. 6, 2006)), whose expression is 2.6-fold higher (p=0.0006, t-test) in mouse pancreatic tumors with satellite expression above the median. An established pancreatic cancer cell line derived from a primary tumor with high satellite expression has very little satellite expression confirming our sequencing results (T3 and CL3; FIG. 2A). Treatment of CL3 with 5-azacytidine leads to massive reexpression of satellite transcripts supporting DNA methylation as a mechanism for stable satellite silencing in vitro (FIG. 2B). Most normal adult mouse tissues, with the exception of lung, show minimal expression of satellite repeats (FIG. 2B). However, expression of the uncleaved 5 kb satellite transcript is evident in embryonic tissues (FIG. 2C). Thus, the aberrant expression of satellite repeats in primary pancreatic tumors does not simply recapitulate an embryonic cell fate, but also reflects altered processing of the primary 5 kb satellite transcript. The single molecule sequencing platform was exceptionally sensitive for quantitation of small repetitive ncRNA fragments, each of which is scored as a unique read. High level expression of the mouse major satellite was evident in all cells within the primary tumor (FIG. 2D), as shown by RNA in situ hybridization (ISH). Remarkably, expression was already elevated in early preneoplastic lesions, pancreatic intraepithelial neoplasia (PanIN), and it increased further upon transition to full pancreatic adenocarcinoma (FIG. 2E). Clearly defined metastatic lesions to the liver ware strongly positive by RNA ISH, as were individual PDAC cells within the liver parenchyma that otherwise would not have been detected by histopathological analysis (FIG. 2F). Low level diffuse expression was evident in liver and lung, as shown by whole mount embryo analysis, but no normal adult or embryonic tissues demonstrated satellite expression comparable to that evident in tumor cells.
  • To determine whether genomic amplification of satellite repeats also contributes toward the exceptional abundance of these transcripts in mouse pancreatic tumors, the index AH284 tumor was analyzed using next generation DNA digital copy number variation (CNV) analysis as described above for genomic DNA sequencing.
  • The results, shown in Table 3, indicated that satellite DNA comprised 18.8% of all genome-aligned reads in this tumor, compared with 2.3% of genomic sequences in matched normal liver. The major satellite repeat has previously been estimated at approximately 3% of the normal mouse genome (J. H. Martens et al., EMBO J 24, 800 (Feb. 23, 2005)). Thus, in this tumor with >100-fold increased expression of satellite repeats, approximately 8-fold gene amplification of the repeats may contribute to their abnormal expression.
  • TABLE 3
    CNV analysis of index pancreatic tumor and normal liver
    from mouse AH284. Major satellite reads as a percentage
    of all genomic aligned reads (last column)
    Major Satellite Reads Total Genomic Reads
    AH284 Liver  183,327 (2.3%) 7,995,538
    AH284 PDAC 2,283,436 (18.8%) 12,124,201
  • Example 3 Overexpression of Satellite Transcripts in Human Pancreatic Cancer and Other Epithelial Cancers
  • To test whether human tumors also overexpress satellite ncRNAs, we extended the DGE analysis to specimens of human pancreatic cancer. Human pancreatic tumor tissues were obtained as excess discarded human material per IRB protocol from the Massachusetts General Hospital. Gross tumor was excised and fresh frozen in liquid nitrogen prior to nucleic acid extraction. Normal pancreas RNA was obtained from two commercial vendors, Clontech and Ambion. The samples were prepared and analyzed as described above in Example 1.
  • Analysis of 15 PDACs showed a median 21-fold increased expression of total satellite transcripts compared with normal pancreas. A cohort of non-small cell lung cancer, renal cell carcinoma, ovarian cancer, and prostate cancer also had significant levels of satellites and the HSATII satellite. Other normal human tissues, including fetal brain, brain, colon, fetal liver, liver, lung, kidney, placenta, prostate, and uterus have somewhat higher levels of total satellite expression (Table 4, FIG. 3A).
  • TABLE 4
    Total
    Satellite ALR HSATII
    SAMPLE ID Genome (tpm) (tpm) (tpm)
    PDAC 1 4,472,810 25,209 14,688 3,589
    PDAC 2 1,668,281 22,001 12,653 3,295
    PDAC 3 5,211,399 27,366 15,921 5,057
    PDAC 4 1,649,041 23,556 13,428 3,167
    PDAC 5 239,483 15,095 8,259 509
    PDAC 6 1,520,470 374 195 14
    PDAC 7 1,449,321 7,738 4,400 750
    PDAC 8 1,950,197 574 316 9
    PDAC 9 3,853,773 19,572 12,563 1,731
    PDAC 10 2,748,850 28,225 18,767 2,489
    PDAC 11 2,848,599 23,163 14,634 2,589
    PDAC 12 3,723,326 21,243 12,940 2,122
    PDAC 13 1,834,743 24,549 15,342 3,150
    PDAC 14 2,481,332 25,650 18,016 2,564
    PDAC 15 1,752,081 38,514 25,899 5,210
    Normal Pancreas 1 1,196,372 908 284 0
    Normal Pancreas 2 975,676 1,043 303 0
    Lung Cancer 1 1,549,237 28,658 18,751 4,417
    Lung Cancer 2 13,829,845 33,030 26,143 2,555
    Kidney Cancer 1 2,104,859 10,814 6,505 1,501
    Kidney Cancer 2 4,753,409 5,025 2,739 625
    Ovarian Cancer 1 12,596,542 26,658 14,513 3,074
    Ovarian Cancer 2 7,290,000 4,089 2,058 403
    Prostate Cancer 1 3,376,849 43,730 22,244 9,793
    Prostate Cancer 2 12,052,244 23,947 14,201 3,209
    Prostate Cancer 3 3,631,148 21,411 12,390 2,804
    Normal Fetal Brain 384,453 2,843 1,516 3
    Normal Brain 371,161 5,184 2,573 3
    Normal Colon 183,855 13,059 7,229 5
    Normal Fetal Liver 147,977 11,218 5,879 7
    Normal Liver 117,976 7,968 3,730 25
    Normal Lung 208,089 15,027 7,857 5
    Normal Kidney 144,173 15,218 8,094 7
    Normal Placenta 207,929 13,990 7,815 0
    Normal Prostate 263,406 8,409 2,228 19
    Normal Uterus 477,480 2,702 1,395 2
  • Subdivision of human satellite among the multiple classes revealed major differences between tumors and all normal tissues. While mouse satellite repeats are broadly subdivided into major and minor satellites, human satellites have been classified more extensively. Of all human satellites, the greatest expression fold differential is evident for the pericentromeric satellite HSATII (mean 2,416 tpm; 10.3% of satellite reads), which is undetectable in normal human pancreas (FIG. 3B). In contrast, normal tissues have much higher representation of GSATII, Beta satellite (BSR), and TAR1 classes (21.1%, 17.3%, and 2.1% of all satellite reads respectively), while these constitute a small minority of satellite reads in pancreatic cancers.
  • The most abundant class of normally expressed human satellites, alpha (ALR) (Okada et al., Cell 131, 1287 (Dec. 28, 2007)) is expressed at 294 tpm in normal human pancreas, but comprises on average 12,535 tpm in human pancreatic adenocarcinomas (43-fold differential expression; 60.3% of satellite reads). Thus, while the overexpression of human ALR repeats is comparable to that of mouse major satellite repeats, it is the less abundant HSATII (49-fold above GAPDH), which shows exceptional specificity for human PDAC. The co-expression of LINE-1 with satellite transcripts in human pancreatic tumors is also striking, with a mean 16,089 tpm (range 358-38,419).
  • Beyond ALR repeats, the satellite expression profile of normal pancreas and PDAC are strikingly different; for instance normal pancreatic tissue has a much higher representation of GSATII, TAR1 and SST1 classes (26.4%, 10.6%, and 8.6% of all satellite reads), while these were a small minority of satellite reads in pancreatic cancers. In contrast, cancers express high levels of HSATII satellites (4,000 per 106 transcripts; 15% of satellite reads), a subtype whose expression is undetectable in normal pancreas (FIG. 3B). Quantitative comparison of satellite transcription in mouse versus human pancreatic cancers shows that mouse major satellites are expressed a median 466-fold above the abundant Gapdh mRNA, while the human ALR and HSATII satellites are respectively expressed 180-fold and 47-fold above GAPDH.
  • Example 4 Cellular Transcripts with Linear Correlation to Increasing Satellite Levels are Enriched for Stem Cell and Neural Elements that is Linked to Histone Demethylases and RNA Processing Enzymes
  • The generation of comprehensive DGE profiles for 25 different mouse tissues of different histologies and genetic backgrounds made it possible to correlate the expression of cellular transcripts with that of satellites across a broad quantitative range. To identify such co-regulated genes, all annotated transcripts quantified by DGE were subjected to linear regression analysis, and transcripts with the highest correlation coefficients to satellite expression were rank ordered.
  • All mouse sample reads were aligned to a custom made library for the mouse major satellite (sequence from UCSC genome browser). Human samples were aligned to a custom made reference library for all satellite repeats and LINE-1 variants generated from the Repbase library (Pushkarev et al., Nat Biotech 27, 847 (2009)). In addition, all samples were subjected to the DGE program for transcriptome analysis. Reads were normalized per 106 genomic aligned reads for all samples.
  • For linear correlation of mouse major satellite to transcriptome, all tissues and cell lines were rank ordered according to level of major satellite. All annotated genes were then subjected to linear regression analysis across all tissues. Genes were then ordered according to the Pearson coefficient for linear regression and plotted by Matlab.
  • Analysis of a set of 297 genes with highest linear correlation (R>0.85) revealed 190 annotated cellular mRNAs and a subset of transposable elements (FIG. 4A).
  • Of all transcripts analyzed with high linear correlation, the autonomous retrotransposon Line-1 had the highest expression level in mouse samples of diverse tissue types. Mouse pancreatic tumors have a mean Line-1 expression 30,690 tpm (range 183-120,002), representing an average of 330-fold higher levels compared to Gapdh (Table 5).
  • TABLE 5
    Normalized Reads per 106 transcripts
    LINE-1 GAPDH
    Mouse PDAC 30,690 (183-120,002) 171 (19-417)
    Human PDAC  6,091 (5,153-6,921) 48 (26-67)
  • The co-expression of LINE-1 with satellite transcripts in human pancreatic tumors was also striking, with an average of 6,091 per 106 transcripts (127-fold higher than GAPDH). Increased expression of the Tigger transposable elements 3 and 4 were also correlated with increasing satellite transcription in mouse tumors, but was not seen human tumors.
  • In addition to retroelements, a subset of cellular mRNAs showed a very high degree of correlation with the levels of satellite repeat expression across diverse mouse tumors (referred to herein as “Satellite Correlated Genes (SCGs)”). Linearly correlated genes with R>0.85 were mapped using the DAVID program (Dennis, Jr. et al., Genome Biol 4, P3 (2003); Huang et al., Nat Protoc 4, 44 (2009)). These genes were then analyzed with the Functional Annotation clustering program and the UP_TISSUE database to classify each of these mapped genes. Germ/Stem cell genes included genes expressed highly in testis, egg, trophoblast, and neural stem cells. Neural genes included genes expressed highly in brain, spinal cord, and specialized sensory neurons including olfactory, auditory, and visual perception. HOX and Zinc Finger proteins were classified using the INTERPRO database.
  • Analysis of 190 annotated transcripts using the DAVID gene ontology program identified 120 (63%) of these transcripts as being associated with neural cell fates and 50 (26%) linked with germ/stem cells pathways (Table 6).
  • TABLE 6
    Zinc Finger
    Germ/Stem Cell Neural HOX Region Domain
    TOTAL
    50 120 10 16
    COUNTS
    % Mapped 26% 63% 5% 8%
    (190)
  • In addition, significant enrichment was evident for transcriptional regulators, including HOX related (9, 5%) and zinc finger proteins (16, 8%). This gene set could not be matched to any known gene signature in the GSEA database (Subramanian et al., Proc Natl Acad Sci USA 102, 15545 (Oct. 25, 2005)), but the ontology analysis points towards a neuroendocrine phenotype. Neuroendocrine differentiation has been described in a variety of epithelial malignancies, including pancreatic cancer (Tezel et al., Cancer 89, 2230 (Dec. 1, 2000)), and is best characterized in prostate cancer where it is correlated with more aggressive disease (Cindolo et al., Urol Int 79, 287 (2007)). A striking increase in the number of carcinoma cells staining for the characteristic neuroendocrine marker chromogranin A, as a function of higher satellite expression in mouse PDACs (FIG. 4D), was observed, supporting the link between globally altered expression of ncRNAs and a specific cellular differentiation program.
  • SCGs were more readily identified in mouse tumors, since the large dynamic range in major satellite expression enabled linear correlation in expression level between satellites and protein-encoding genes. However, human orthologs were identified for 138 of the 190 annotated mouse SGCs, of which 54 (39%) showed >2-fold increased expression in human PDACs compared with normal pancreas (q-value<0.1). Together, these observations suggest that, as in the mouse genetic model, tumor-associated derepression of satellite-derived repeats is highly correlated with increased expression of Line-1 and a subset of cellular mRNAs.
  • Histone modifications, including H3K9 trimethylation (P. A. Cloos, J. Christensen, K. Agger, K. Helin, Genes Dev 22, 1115 (May 1, 2008)), combined with Dicer1 and Piwi-related protein-mediated ncRNA processing (A. A. Aravin, G. J. Hannon, J. Brennecke, Science 318, 761 (Nov. 2, 2007)) have been linked to maintenance of repression of satellite repeats. To search for candidate regulators of satellite derepression in primary tumor specimens, we first measured the quantitative DGE of known epigenetic regulators and RNA processing genes in mouse tumors, as a function of increasing major satellite expression.
  • A targeted gene expression analysis of demethylases and RNA processing enzymes was carried out in mouse and human PDAC samples. A list of demethylases and RNA processing enzymes were generated from two recent publications (Cloos et al., Genes Dev 22, 1115 (2008); Aravin et al., Science 318, 761 (2007)). Mouse PDACs with KrasG12D and Tp53 loss were used for this analysis. Mouse tumors were separated into high vs low satellite levels using the median satellite expression (7%). A total of 37 genes were evaluated between high and low satellite tumors and fold change was calculated. Analysis of the population means was compared using the 2-tailed student t-test assuming equal variance. Genes that had a significance value of p<0.05 (total of 7 gene) were then used to evaluate human PDAC versus human normal pancreatic tissue. Fold change and a 2-tailed student t-test with equal variance was then applied to human PDAC vs normal pancreatic tissue to find potential gene candidates involved with satellite expression.
  • Among 37 candidate genes tested, mouse pancreatic tumors with satellite expression above the median had higher expression of the demethylases Hspbap1, Jmjd1B, Jmjd4, Jarid1d, Jmjd3, and Fbxl10 as well as the RNA processing enzyme Dicer1. Among these, HSPBAP1, FBXL10 and DICER1 overexpression was also observed in human pancreatic adenocarcinomas (Table 7, p<0.05, student t-test).
  • TABLE 7
    List of candidate demethylases and RNA processing enzymes
    identified in mouse PDAC tumors compared to human PDAC.
    Mouse PDAC High vs Low Human PDAC vs
    Satellite Normal Panc
    FOLD FOLD
    Expres- T-test Expres- T-test
    GENE NAME sion p-value sion p-value
    HSPBAP1 5.11 0.0005 11.59 0.0069
    DICER1 2.56 0.0006 3.00 0.0023
    JMJD1B (KDM3B) −2.16 0.0010 1.16 0.8051
    JMJD4 3.10 0.0021 1.18 0.8080
    JARID1D (SMCY, 9.84 0.0031 1.62 0.7107
    KDM5D)
    JMJD3 (KDM6B) 1.62 0.0118 3.02 0.1109
    FBXL10 (KDM2B) 1.40 0.0412 7.96 0.0279
    Demethylases and RNA processing enzymes enriched in high vs low major satellite expressing mouse tumors (First two columns) ordered by highest significance (lowest p-value). Human PDAC vs normal tissue fold change and t-test p-value shown for each of these genes (Last two columns). Genes differentially expressed in both human and mouse tumors with a significance value of p < 0.05 are highlighted in bold.
  • The catalytic activity of HSPBAP1 and FBXL10 demethylases has not been extensively characterized, although the former is noteworthy for its contribution to the familial renal cancer DIRC3-HSPBAP1 fusion (D. Bodmer, M. Schepens, M. J. Eleveld, E. F. Schoenmakers, A. Geurts van Kessel, Genes Chromosomes Cancer 38, 107 (October, 2003)) and the latter appears to have some specificity for H3K36me2/me1 and H3K4me3, with an effect on ribosomal RNA expression and cellular proliferation (D. Frescas, D. Guardavaccaro, F. Bassermann, R. Koyama-Nasu, M. Pagano, Nature 450, 309 (Nov. 8, 2007)). While current understanding of multicomponent chromatin modifier complexes precludes linking satellite and Line-1 upregulation in primary tumors with aberrant expression of a single transcript, the relatively small number of genes with consistently altered expression may point to a key subset of epigenetic regulators that contribute to satellite and Line-1 derepression.
  • While failure of a common transcriptional silencing mechanism may contribute to repression of LINE-1 and satellite repeats, the diverse array of cellular transcripts whose overexpression is correlated with these repetitive sequences is less readily explained. Of note, recent findings have demonstrated that LINE-1 may drive expression of specific cellular mRNAs through its insertion upstream of their transcriptional start sites (T. Kuwabara et al., Nat Neurosci 12, 1097 (September, 2009)) or through alterations in flanking chromatin marks (J. A. Bailey, L. Carrel, A. Chakravarti, E. E. Eichler, Proceedings of the National Academy of Sciences of the United States of America 97, 6634 (Jun. 6, 2000, 2000); D. E. Montoya-Durango et al., Mutat Res 665, 20 (Jun. 1, 2009)). To test whether such a mechanism might also underlie the co-expression of the 297 cellular mRNAs identified here, we analyzed the genomic distance between their transcriptional start site and LINE-1 insertions in the genome.
  • The transcriptional start sites tissues and cell lines were determined (UCSC genome browser (D. Karolchik et al., Nucleic Acids Res 32, D493 (Jan. 1, 2004))) as well as the position of all Line-1 elements in the mouse genome with a threshold of 1 Kbp in length. Line-1 closest distance upstream of the transcriptional start sites of all annotated genes with a minimum expression level of 5 transcripts per million were calculated. Genes were then rank ordered according to the Pearson coefficient for linear regression. Genes were binned in 100 s and plotted by Excel. Randomization of all genes, followed by binning, and plotting was done as a control.
  • Focusing on the top linearly correlated genes (R>0.85), these genes were plotted as a frequency plot against distance of Line-1 elements upstream of the transcriptional start site. Enrichment was calculated at 5 Kbp and the Fisher Exact test was used to calculate the test statistic.
  • Remarkably, there was a striking correlation of LINE-1 genomic distance to expression of genes with the major satellite (FIG. 4B), and there was highly significant enrichment of our top 297 genes for presence of a LINE-1 element within 5 Kbp of the transcriptional start site (Enrichment 2.69, p=8.18×10−7, Fisher exact test, FIG. 4C).
  • Thus, activation of LINE-1 sequences within the proximity of cellular transcripts may contribute to their overexpression in primary tumors, in striking correlation with the expression of both LINE-1 and satellite repeats. The consequence of increased expression of these cellular transcripts remains to be defined. However, the high prevalence of genes linked to stem-like and neurogenic fates, along with the frequency of HOX and zinc finger transcriptional regulators raises the possibility that at least a subset of these may contribute to tumor-related phenotypes.
  • Example 5 LINE-1 is a Specific and Sensitive Marker of CTCs
  • Satellite levels are most strongly linked with the expression of the autonomous retrotransposon Line-1, which has recently been shown to be a major cause for genomic variation in normal and tumor tissues (J. Berretta, A. Morillon, EMBO Rep 10, 973 (September, 2009); A. Jacquier, Nat Rev Genet 10, 833 (December, 2009); M. Guenatri, D. Bailly, C. Maison, G. Almouzni, J Cell Biol 166, 493 (Aug. 16, 2004)). Aberrant expression of cellular transcripts linked to stem cells and neural tissues is also highly correlated with satellite transcript levels, suggesting alteration of cell fate through derepression of a coordinated epigenetic program.
  • Expression of LINE-1 in circulating tumor cells (CTCs) in newly diagnosed metastatic pancreatic adenocarcinoma patients was evaluated using a CTC device known as a herringbone chip (HB), which combines specific antibody mediated capture against the epithelial cell adhesion molecule (EpCAM) and the high-throughput advantages of microfluidics (Stott et al., Proc Natl Acad Sci USA, 107(43):18392-18397 2010). Blood was collected from cancer patients that had given consent. Approximately 3 mL of blood was processed on the CTC device and a control device over 2 hours. RNA was extracted from the devices using the Qiagen RNeasy MinElute kit. RNA was then subjected to cDNA synthesis with random primers using the Superscript III first strand synthesis kit (invitrogen). RNA was removed with RNase and cDNA was subject to qRT-PCR using Human LINE-1 Taqman assay (Applied Biosystems). LINE-1 expression was normalized to GAPDH in the CTC and control device. Fold difference between the CTC device and control device was then calculated by standard Ct calculation for qPCR. As shown in FIG. 5, LINE-1 expression was seen much more consistently and with higher frequency compared to keratins (Krt) which are typical CTC markers (S. Maheswaran et al., N Engl J Med. 359, 366 (Jul. 24, 2008); S. Nagrath et al., Nature. 450, 1235 (Dec. 20, 2007)). Preliminary data of HSATII positive cells suggests a 10 fold improvement in sensitivity of CTC detection. This demonstrates that LINE-1 is a specific and sensitive marker for detecting CTCs. The ability of satellites to increase the sensitivity of detecting pancreatic CTCs offers not only a more robust blood based diagnostic for cancer treatment monitoring, but a significant improvement in the ability to use CTCs as an early detection modality. This provides a screening tool for cancer, improving chances of early detection, which can improve the ability to provide curative therapy.
  • Example 6 HSATII RNA in Situ Technique using Branched DNA Detection Technology
  • As noted above, the HSATII satellite is overexpressed in pancreatic cancer and was confirmed to be overexpressed in human preneoplastic pancreatic lesions (FIG. 6A) using a branched DNA detection assay (QuantiGene® ViewRNA Assay, Affymetrix). Breast cancer samples were also tested for HSATII using this method and were found to have significant expression compared to normal breast tissues. Extension of this technique to potential circulating tumor cells captured on the HB-chip (FIG. 6B) has been accomplished indicating that HSATII may be used as a blood based diagnostic for epithelial cancers.
  • Example 7 Satellites Levels in Serum
  • Serum was extracted from the blood of 8 metastatic pancreatic cancer patients by using Ficoll buffy coat method. Serum RNA (cell free RNA), which includes exosomes, was purified using the Trizol method and then purified using Qiagen RNA MinElute columns kits. RNA was then subjected to Helicos DGE sequencing preparation and sequenced on a HeliScope next generation sequencer. Results of this data are summarized in Table 1. As described above, HSATII was specific for cancer and GSATII was found to correlate with normal tissues. Therefore the ratio of HSATII to GSATII was evaluated as a marker for identifying cancer burden and potentially an early detection marker. In this case, one patient who had stable disease had the lowest HSATII/GSATII ratio as predicted (see Table 8). These results suggest that detection of satellites in peripheral blood serum (cell free RNA) can be used as a predictive marker of disease response to therapy.
  • TABLE 8
    Total
    Patient Clinical Satellites HSATII GSATII HSATII/
    ID status (tpm) (tpm) (tpm) GSATII
    PDAC
    3 PROGRESSION 43,932 1,576 4,904 32%
    PDAC 6 PROGRESSION 21,845 735 3,151 23%
    PDAC 9 PROGRESSION 39,235 1,867 1,857 101% 
    PDAC 11 PROGRESSION 28,817 784 3,785 21%
    PDAC 12 PROGRESSION 2,472 59 83 71%
    PDAC 16 STABLE 43,629 162 6,437  3%
    PDAC 18 PROGRESSION 18,034 231 2,450  9%
    PDAC 19 PROGRESSION 38,425 399 5,287  8%
  • Table 8: A total of 8 metastatic cancer patients with clinical status, total satellites, HSATII, and GSATII in transcripts per million aligned to genome (tpm) and the ratio of HSATII/GSATII in cell free RNA sequenced.
  • However, in a preliminary evaluation of healthy donor serum (cell free) RNA (n=4) HSATII and GSATII did not perform as well as expected, though the presence of. However, other satellites like TAR1 seemed to be better predictors of “cancer” compared to “non-cancer” status as shown in Table 9. TAR1 was significantly different between the two populations with a p value=0.025.
  • TABLE 9
    Total
    Satellites HSATII GSATII HSATII/ TAR1
    (tpm) (tpm) (tpm) GSATII (tpm)
    AVG PDAC 29,549 727 3,494 0.33 100
    AVG HD 47,279 6,275 6,081 1.12 51
    TTEST 0.114 0.172 0.103 0.25 0.025
    FOLD 0.625 0.116 0.575 0.30 1.963
  • Table 9: Average total satellites, HSATII, GSATII, HSATII/GSATII, and TAR1 (tpm) in a total of 8 metastatic PDAC patients and 4 healthy donors with cell free RNA sequenced. Student t-test was used to calculate significance.
  • Other Embodiments
  • It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims (23)

1. An in vitro method of detecting the presence of cancer in a subject, the method comprising:
determining a level of Long Interspersed Nucleotide Element-1 (LINE-1) in a sample from the subject to obtain a test value;
comparing the test value to a reference value, wherein the reference value represents a threshold level of LINE-1, and
identifying the subject as having cancer when the level of LINE-1 in the subject is above the reference value, or identifying the subject as unlikely to have cancer when the level of LINE-1 in the subject is below the reference value.
2. (canceled)
3. An in vitro method of detecting the presence of cancer in a subject, the method comprising:
determining a level of satellite transcripts in a sample from the subject to obtain a test value; and
comparing the test value to a reference value,
wherein a test value compared to the reference value indicates whether the subject has cancer.
4. The method of claim 2, wherein the satellite transcripts comprise one or more of ALR, HSATII, GSATII, TAR1, and SST1.
5. The method of claim 4, wherein the satellite transcript is ALR and/or HSATII, and the presence of a level of ALR and/or HSATII satellite transcripts above the reference level indicates that the subject has a tumor.
6. The method of claim 4, wherein the satellite transcript is GSATII, TAR1 and/or SST1, and the presence of a level of GSATII, TAR1 and/or SST1 satellite transcripts below the reference level indicates that subject has a tumor.
7. The method of claim 1, wherein the sample is known or suspected to comprise tumor cells.
8. The method of claim 7, wherein the sample is a blood sample known or suspected of comprising circulating tumor cells (CTCs), or a biopsy sample known or suspected of comprising tumor cells.
9. The method of claim 3, wherein the sample comprises free RNA in serum or RNA within exosomes in blood.
10. The method of claim 1, wherein the subject is a human.
11. The method of claim 1, wherein the cancer is a solid tumor of epithelial origin.
12. An in vitro method of evaluating the efficacy of a treatment for cancer in a subject, the method comprising:
determining a level of LINE-1 in a first sample from the subject to obtain a first value;
administering a treatment for cancer to the subject;
determining a level of LINE-1 in a subsequent sample obtained from the subject at a later time, to obtain a treatment value; and
comparing the first value to the treatment value,
wherein a treatment value that is below the first value indicates that the treatment is effective.
13. An in vitro method of evaluating the efficacy of a treatment for cancer in a subject, the method comprising:
determining a level of satellite transcripts in a first sample from the subject to obtain a first value;
administering a treatment for cancer to the subject;
determining a level of satellite transcripts in a subsequent sample obtained from the subject at a later time, to obtain a treatment value; and
comparing the first value to the treatment value,
wherein a treatment value that is below the first value indicates that the treatment is effective.
14. The method of claim 13, wherein the satellite transcripts comprise one or more of ALR, HSATII, GSATII, TAR1, and SST1.
15. The method of claim 12, wherein the first and second samples are known or suspected to comprise tumor cells.
16. The method of claim 15, wherein the samples are blood samples known or suspected of comprising circulating tumor cells (CTCs), or biopsy samples known or suspected of comprising tumor cells.
17. The method of claim 13, wherein the sample comprises free RNA in serum or RNA within exosomes in blood.
18. The method of claim 12, wherein the treatment includes administration of a surgical intervention, chemotherapy, radiation therapy, or a combination thereof.
19. The method of claim 12, wherein the subject is a human.
20. The method of claim 12, wherein the cancer is a solid tumor of epithelial origin.
21. The method of claim 1, comprising measuring a level of LINE-1 transcript.
22. The method of claim 12, wherein the level is determined using a branched DNA assay.
23. The method of claim 11, wherein the solid tumor of epithelial origin is pancreatic, lung, breast, prostate, renal, ovarian or colon cancer.
US13/877,373 2010-10-07 2011-10-06 Biomarkers of Cancer Abandoned US20140031250A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/877,373 US20140031250A1 (en) 2010-10-07 2011-10-06 Biomarkers of Cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US39095610P 2010-10-07 2010-10-07
US201161493800P 2011-06-06 2011-06-06
US13/877,373 US20140031250A1 (en) 2010-10-07 2011-10-06 Biomarkers of Cancer
PCT/US2011/055108 WO2012048113A2 (en) 2010-10-07 2011-10-06 Biomarkers of cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/055108 A-371-Of-International WO2012048113A2 (en) 2010-10-07 2011-10-06 Biomarkers of cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/589,586 Continuation US11142800B2 (en) 2010-10-07 2017-05-08 Biomarkers of cancer

Publications (1)

Publication Number Publication Date
US20140031250A1 true US20140031250A1 (en) 2014-01-30

Family

ID=45928434

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/877,373 Abandoned US20140031250A1 (en) 2010-10-07 2011-10-06 Biomarkers of Cancer
US15/589,586 Active 2033-06-07 US11142800B2 (en) 2010-10-07 2017-05-08 Biomarkers of cancer
US17/497,270 Pending US20220098678A1 (en) 2010-10-07 2021-10-08 Biomarkers of Cancer

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/589,586 Active 2033-06-07 US11142800B2 (en) 2010-10-07 2017-05-08 Biomarkers of cancer
US17/497,270 Pending US20220098678A1 (en) 2010-10-07 2021-10-08 Biomarkers of Cancer

Country Status (4)

Country Link
US (3) US20140031250A1 (en)
EP (1) EP2625292B1 (en)
CN (1) CN103517990A (en)
WO (1) WO2012048113A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140213475A1 (en) * 2011-07-14 2014-07-31 University Of Massachusetts Methods of diagnosing cancer using epigenetic biomarkers
WO2016196284A1 (en) * 2015-05-29 2016-12-08 Epic Sciences, Inc. Intra-patient genomic heterogeneity of single circulating tumor cells (ctcs) associated to phenotypic ctc heterogeneity in metastatic castrate resistant prostate cancer (mcrpc)
EP3256608A4 (en) * 2015-02-13 2019-02-20 Icahn School of Medicine at Mount Sinai Rna containing compositions and methods of their use
US10527624B2 (en) 2014-01-27 2020-01-07 Epic Sciences, Inc. Circulating tumor cell diagnostics for prostate cancer biomarkers
US10545151B2 (en) 2014-02-21 2020-01-28 Epic Sciences, Inc. Methods for analyzing rare circulating cells
US10613089B2 (en) 2006-01-30 2020-04-07 The Scripps Research Institute Method of using non-rare cells to detect rare cells
US11142800B2 (en) 2010-10-07 2021-10-12 The General Hospital Corporation Biomarkers of cancer

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2510539B (en) * 2011-10-24 2016-04-20 Gen Hospital Corp Biomarkers of cancer
WO2014004945A1 (en) * 2012-06-28 2014-01-03 University Of Louisville Research Foundation, Inc. Use of protein line-1 orf-1 as a biomarker for cancer
US10993418B2 (en) 2012-08-13 2021-05-04 Life Genetics Lab, Llc Method for measuring tumor burden in patient derived xenograft (PDX) mice
DK3102722T3 (en) 2014-02-04 2020-11-16 Jumpcode Genomics Inc THROUGH FRACTIONING
US20160376663A1 (en) * 2014-02-27 2016-12-29 Igenomx International Genomics Corporation Methods for analysis of somatic mobile elements, and uses thereof
US10301624B2 (en) 2014-06-25 2019-05-28 The General Hospital Corporation Targeting human satellite II (HSATII)
WO2016109604A2 (en) 2014-12-29 2016-07-07 InnoGenomics Technologies, LLC Multiplexed assay for quantitating and assessing integrity of cell-free dna in biological fluids for cancer diagnosis, prognosis, and surveillance
US10968536B2 (en) 2015-02-25 2021-04-06 Jumpcode Genomics, Inc. Methods and compositions for sequencing
US11339427B2 (en) 2016-02-12 2022-05-24 Jumpcode Genomics, Inc. Method for target specific RNA transcription of DNA sequences
EP4317972A3 (en) 2018-02-06 2024-03-13 The General Hospital Corporation Repeat rna as biomarkers of tumor immune response
CN109709333B (en) * 2018-08-01 2021-09-24 东南大学 Application of detection reagent for trimethylation amounts of H4K20, H3K9 and H3K36 in esophageal cancer prognosis evaluation
WO2020142436A1 (en) * 2019-01-02 2020-07-09 Arizona Board Of Regents On Behalf Of The University Of Arizona Systems and methods for characterizing and treating disease
WO2022066880A1 (en) * 2020-09-23 2022-03-31 Transposon Therapeutics, Inc. Line-1 inhibitors to treat disease

Family Cites Families (243)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4476301A (en) 1982-04-29 1984-10-09 Centre National De La Recherche Scientifique Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
JPS5927900A (en) 1982-08-09 1984-02-14 Wakunaga Seiyaku Kk Oligonucleotide derivative and its preparation
FR2540122B1 (en) 1983-01-27 1985-11-29 Centre Nat Rech Scient NOVEL COMPOUNDS COMPRISING A SEQUENCE OF OLIGONUCLEOTIDE LINKED TO AN INTERCALATION AGENT, THEIR SYNTHESIS PROCESS AND THEIR APPLICATION
US4605735A (en) 1983-02-14 1986-08-12 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives
US4948882A (en) 1983-02-22 1990-08-14 Syngene, Inc. Single-stranded labelled oligonucleotides, reactive monomers and methods of synthesis
US4824941A (en) 1983-03-10 1989-04-25 Julian Gordon Specific antibody to the native form of 2'5'-oligonucleotides, the method of preparation and the use as reagents in immunoassays or for binding 2'5'-oligonucleotides in biological systems
US4587044A (en) 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
US5118802A (en) 1983-12-20 1992-06-02 California Institute Of Technology DNA-reporter conjugates linked via the 2' or 5'-primary amino group of the 5'-terminal nucleoside
US5550111A (en) 1984-07-11 1996-08-27 Temple University-Of The Commonwealth System Of Higher Education Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
US5258506A (en) 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
US4828979A (en) 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US4762779A (en) 1985-06-13 1988-08-09 Amgen Inc. Compositions and methods for functionalizing nucleic acids
US5317098A (en) 1986-03-17 1994-05-31 Hiroaki Shizuya Non-radioisotope tagging of fragments
JPS638396A (en) 1986-06-30 1988-01-14 Wakunaga Pharmaceut Co Ltd Poly-labeled oligonucleotide derivative
EP0260032B1 (en) 1986-09-08 1994-01-26 Ajinomoto Co., Inc. Compounds for the cleavage at a specific position of RNA, oligomers employed for the formation of said compounds, and starting materials for the synthesis of said oligomers
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
DE3851889T2 (en) 1987-06-24 1995-04-13 Florey Howard Inst NUCLEOSIDE DERIVATIVES.
US5585481A (en) 1987-09-21 1996-12-17 Gen-Probe Incorporated Linking reagents for nucleotide probes
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US5525465A (en) 1987-10-28 1996-06-11 Howard Florey Institute Of Experimental Physiology And Medicine Oligonucleotide-polyamide conjugates and methods of production and applications of the same
DE3738460A1 (en) 1987-11-12 1989-05-24 Max Planck Gesellschaft MODIFIED OLIGONUCLEOTIDS
JP3019994B2 (en) 1987-11-30 2000-03-15 ユニバーシティ オブ アイオワ リサーチ ファウンデーション Novel oligodeoxynucleotide, method of blocking expression of target gene using the same, and novel oligodeoxynucleotide and method of inhibiting expression of target gene using the same
US5403711A (en) 1987-11-30 1995-04-04 University Of Iowa Research Foundation Nucleic acid hybridization and amplification method for detection of specific sequences in which a complementary labeled nucleic acid probe is cleaved
US5082830A (en) 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
EP0406309A4 (en) 1988-03-25 1992-08-19 The University Of Virginia Alumni Patents Foundation Oligonucleotide n-alkylphosphoramidates
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5109124A (en) 1988-06-01 1992-04-28 Biogen, Inc. Nucleic acid probe linked to a label having a terminal cysteine
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5262536A (en) 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5599923A (en) 1989-03-06 1997-02-04 Board Of Regents, University Of Tx Texaphyrin metal complexes having improved functionalization
US5457183A (en) 1989-03-06 1995-10-10 Board Of Regents, The University Of Texas System Hydroxylated texaphyrins
US5391723A (en) 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
GB2236186B (en) 1989-08-22 1994-01-05 Finnigan Mat Gmbh Process and device for laser desorption of analyte molecular ions, especially of biomolecules
US5451463A (en) 1989-08-28 1995-09-19 Clontech Laboratories, Inc. Non-nucleoside 1,3-diol reagents for labeling synthetic oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5254469A (en) 1989-09-12 1993-10-19 Eastman Kodak Company Oligonucleotide-enzyme conjugate that can be used as a probe in hybridization assays and polymerase chain reaction procedures
US5045694A (en) 1989-09-27 1991-09-03 The Rockefeller University Instrument and method for the laser desorption of ions in mass spectrometry
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5641628A (en) 1989-11-13 1997-06-24 Children's Medical Center Corporation Non-invasive method for isolation and detection of fetal DNA
US5292873A (en) 1989-11-29 1994-03-08 The Research Foundation Of State University Of New York Nucleic acids labeled with naphthoquinone probe
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5623065A (en) 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5220007A (en) 1990-02-15 1993-06-15 The Worcester Foundation For Experimental Biology Method of site-specific alteration of RNA and production of encoded polypeptides
US5149797A (en) 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
WO1991013080A1 (en) 1990-02-20 1991-09-05 Gilead Sciences, Inc. Pseudonucleosides and pseudonucleotides and their polymers
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
EP0455905B1 (en) 1990-05-11 1998-06-17 Microprobe Corporation Dipsticks for nucleic acid hybridization assays and methods for covalently immobilizing oligonucleotides
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
JPH0874B2 (en) 1990-07-27 1996-01-10 アイシス・ファーマシューティカルス・インコーポレーテッド Nuclease-resistant, pyrimidine-modified oligonucleotides that detect and modulate gene expression
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5688941A (en) 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5245022A (en) 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
KR100211552B1 (en) 1990-08-03 1999-08-02 디. 꼬쉬 Compounds and methods for inhibiting gene expression
US5177196A (en) 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
US5512667A (en) 1990-08-28 1996-04-30 Reed; Michael W. Trifunctional intermediates for preparing 3'-tailed oligonucleotides
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
CA2092002A1 (en) 1990-09-20 1992-03-21 Mark Matteucci Modified internucleoside linkages
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
DE69132510T2 (en) 1990-11-08 2001-05-03 Hybridon Inc CONNECTION OF MULTIPLE REPORTING GROUPS ON SYNTHETIC OLIGONUCLEOTIDS
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5719262A (en) 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US5714331A (en) 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5371241A (en) 1991-07-19 1994-12-06 Pharmacia P-L Biochemicals Inc. Fluorescein labelled phosphoramidites
US5571799A (en) 1991-08-12 1996-11-05 Basco, Ltd. (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
JP4111463B2 (en) 1991-08-23 2008-07-02 ボード・オブ・リージェンツ・オブ・ジ・ユニバーシティ・オブ・ネブラスカ Cell reprogramming method and composition
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
US5565552A (en) 1992-01-21 1996-10-15 Pharmacyclics, Inc. Method of expanded porphyrin-oligonucleotide conjugate synthesis
US5595726A (en) 1992-01-21 1997-01-21 Pharmacyclics, Inc. Chromophore probe for detection of nucleic acid
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
US5272250A (en) 1992-07-10 1993-12-21 Spielvogel Bernard F Boronated phosphoramidate compounds
US5652355A (en) 1992-07-23 1997-07-29 Worcester Foundation For Experimental Biology Hybrid oligonucleotide phosphorothioates
US5574142A (en) 1992-12-15 1996-11-12 Microprobe Corporation Peptide linkers for improved oligonucleotide delivery
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
JPH08508491A (en) 1993-03-31 1996-09-10 スターリング ウインスロップ インコーポレイティド Oligonucleotides with phosphodiester bonds replaced by amide bonds
ES2201077T3 (en) 1993-05-28 2004-03-16 Baylor College Of Medicine METHOD AND SPECTROMETER OF MASSES FOR DESORTION AND IONIZATION OF ANALYTS.
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
EP0733059B1 (en) 1993-12-09 2000-09-13 Thomas Jefferson University Compounds and methods for site-directed mutations in eukaryotic cells
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US5597696A (en) 1994-07-18 1997-01-28 Becton Dickinson And Company Covalent cyanine dye oligonucleotide conjugates
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5716928A (en) 1995-06-07 1998-02-10 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
US5652356A (en) 1995-08-17 1997-07-29 Hybridon, Inc. Inverted chimeric and hybrid oligonucleotides
US5858401A (en) 1996-01-22 1999-01-12 Sidmak Laboratories, Inc. Pharmaceutical composition for cyclosporines
US6007839A (en) 1996-02-16 1999-12-28 The Liposome Company, Inc. Preparation of pharmaceutical compositions containing etherlipid-containing multiple lipid liposomes
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides and oligonucleotide analogues
US6630137B1 (en) 1997-04-28 2003-10-07 Eli Lilly And Company Activated protein C formulations
NZ516848A (en) 1997-06-20 2004-03-26 Ciphergen Biosystems Inc Retentate chromatography apparatus with applications in biology and medicine
AU8285998A (en) 1997-07-02 1999-01-25 Sdg, Inc. Targeted liposomal constructs for diagnostic and therapeutic uses
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US7572582B2 (en) 1997-09-12 2009-08-11 Exiqon A/S Oligonucleotide analogues
US20030166132A1 (en) 1998-08-26 2003-09-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP1140137A2 (en) 1998-12-18 2001-10-10 Scios Inc. Method for detection and use of differentially expressed genes in disease states
US7084125B2 (en) 1999-03-18 2006-08-01 Exiqon A/S Xylo-LNA analogues
US6734291B2 (en) 1999-03-24 2004-05-11 Exiqon A/S Synthesis of [2.2.1]bicyclo nucleosides
US7053207B2 (en) 1999-05-04 2006-05-30 Exiqon A/S L-ribo-LNA analogues
DE60035057T2 (en) 1999-10-04 2008-01-31 Novartis Vaccines and Diagnostics, Inc., Emeryville CD40 antagonist for the treatment of psoriasis
US20060128006A1 (en) 1999-11-10 2006-06-15 Gerhardt Antimony L Hydrodynamic capture and release mechanisms for particle manipulation
US6692952B1 (en) 1999-11-10 2004-02-17 Massachusetts Institute Of Technology Cell analysis and sorting apparatus for manipulation of cells
US6287860B1 (en) 2000-01-20 2001-09-11 Isis Pharmaceuticals, Inc. Antisense inhibition of MEKK2 expression
US20030059427A1 (en) 2000-04-28 2003-03-27 Force Walker R. Isolation and characterization of highly active anti-CD40 antibody
US7033758B2 (en) 2000-06-02 2006-04-25 Bayer Corporation Highly sensitive gene detection and localization using in situ branched-DNA hybridization
US20020042072A1 (en) 2000-06-26 2002-04-11 Jacobus Van Meel Method for monitoring the effect of cancer therapies
DE60219292T2 (en) 2001-04-10 2008-04-10 Pfizer Inc. PYRAZONE DERIVATIVES FOR THE TREATMENT OF HIV
CN101508734A (en) 2001-04-27 2009-08-19 协和发酵麒麟株式会社 Anti-CD40 monoclonal antibody
US8207142B2 (en) 2001-07-31 2012-06-26 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Inhibitor of DNA methylation
JP2005514005A (en) 2001-09-04 2005-05-19 エクシコン エ/エス Novel LNA compositions and uses thereof
ATE481985T1 (en) 2002-07-03 2010-10-15 Ono Pharmaceutical Co IMMUNOPOTENTATING COMPOSITIONS
EP2359689B1 (en) 2002-09-27 2015-08-26 The General Hospital Corporation Microfluidic device for cell separation and use thereof
CN1703524B (en) 2002-09-30 2010-04-07 肿瘤疗法科学股份有限公司 Genes and polypeptides relating to human pancreatic cancer
AU2003273778A1 (en) 2002-10-21 2004-05-04 Exiqon A/S Oligonucleotide analogues for detecting and analyzing nucleic acids
US20050113324A1 (en) 2003-01-15 2005-05-26 Bondarev Igor E. Modulation of line-1 reverse transcriptase
US20060160087A1 (en) 2003-01-31 2006-07-20 Mcgrath Michael Monitoring and treatment of amyotrophic lateral sclerosis
AU2004221760B2 (en) 2003-03-21 2010-03-18 Roche Innovation Center Copenhagen A/S Short interfering RNA (siRNA) analogues
US20060115806A1 (en) * 2003-04-29 2006-06-01 University Of Georgia Research Foundation, Inc. Global analysis of transposable elements as molecular markers of cancer
US8277810B2 (en) 2003-11-04 2012-10-02 Novartis Vaccines & Diagnostics, Inc. Antagonist anti-CD40 antibodies
PT1707627E (en) 2003-12-25 2013-01-24 Kyowa Hakko Kirin Co Ltd Antagonistic anti-cd40 antibody mutant
DE602005017131D1 (en) 2004-01-21 2009-11-26 Gilead Sciences Inc USE OF ADEFOVIR OR TENOFOVIR FOR INHIBITING MMTV-LIKE VIRUSES IN CONNECTION WITH BREAST CANCER AND PRIMARY BILIARY CIRRHOSIS
WO2005073378A1 (en) 2004-01-30 2005-08-11 Santaris Pharma A/S MODIFIED SHORT INTERFERING RNA (MODIFIED siRNA)
WO2005084374A2 (en) 2004-03-03 2005-09-15 The General Hospital Corporation Magnetic device for isolation of cells and biomolecules in a microfluidic environment
US20060121624A1 (en) 2004-03-03 2006-06-08 Huang Lotien R Methods and systems for fluid delivery
EP1765503A2 (en) 2004-03-03 2007-03-28 The General Hospital Corporation System for delivering a diluted solution
US7618947B2 (en) 2004-08-25 2009-11-17 Isis Pharmaceuticals, Inc. Modulation of HIF-1 beta expression
NZ555645A (en) 2004-11-09 2009-08-28 Santaris Pharma As LNA oligonucleotides and the treatment of cancer
EP1871391B1 (en) 2005-03-30 2011-12-28 Viroxis Endogenous retrovirus and proteins encoded by env as a target for cancer treatment
US20070196820A1 (en) 2005-04-05 2007-08-23 Ravi Kapur Devices and methods for enrichment and alteration of cells and other particles
US8652786B2 (en) 2005-04-05 2014-02-18 The General Hospital Corporation Method for predicting responsiveness to drugs
US8628918B2 (en) 2005-05-09 2014-01-14 Affymetrix, Inc. Multiplex capture of nucleic acids
SI2439273T1 (en) 2005-05-09 2019-05-31 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US7803541B2 (en) 2005-05-12 2010-09-28 Panomics, Inc. Multiplex branched-chain DNA assays
CA2609303A1 (en) * 2005-05-18 2006-11-23 Alt Solutions, Inc. Pharmacological modulation of telomere length in cancer cells for prevention and treatment of cancer
EP1888786A4 (en) * 2005-05-27 2009-12-30 Wayne John Cancer Inst Use of free circulating dna for diagnosis, prognosis, and treatment of cancer
CN101495650B (en) 2005-06-20 2015-02-04 领先细胞医疗诊断有限公司 Methods of detecting nucleic acids in individual cells and of identifying rare cells from large heterogeneous cell populations
AR057455A1 (en) 2005-07-22 2007-12-05 Merck & Co Inc INHIBITORS OF HIV REVERSE TRANSCRIPTASE AND PHARMACEUTICAL COMPOSITION
ES2324128A1 (en) 2005-09-29 2009-07-30 Proyecto De Biomedicina Cima, S.L. Molecular markers of hepatocellular carcinoma and their applications
WO2007045572A1 (en) 2005-10-19 2007-04-26 F. Hoffmann-La Roche Ag N-phenyl phenylacetamide non-nucleoside reverse transcriptase inihibitors
AU2007234191B2 (en) 2006-04-03 2012-07-12 Roche Innovation Center Copenhagen A/S Pharmaceutical composition comprising anti-miRNA antisense oligonucleotides
AU2007240507B2 (en) 2006-04-21 2013-07-18 Novartis Ag Antagonist anti-CD40 antibody pharmaceutical compositions
NZ573888A (en) 2006-07-21 2012-02-24 Gilead Sciences Inc Aza-peptide protease inhibitors
WO2008030605A2 (en) 2006-09-08 2008-03-13 The Regents Of The University Of Michigan Herv group ii viruses in lymphoma and cancer
WO2008058285A2 (en) 2006-11-09 2008-05-15 Ardea Biosciences, Inc. 4-cyanophenylamino-substituted bicyclic heterocyclic compounds as hiv inhibitors
CZ200718A3 (en) 2007-01-09 2008-07-16 Ústav organické chemie a biochemie AV CR, v.v.i. 1-(2-Deoxy-alpha-D-erythro-pentofuranosyl)-5-azacytosine for use as medicament
EP2118118B1 (en) 2007-01-19 2017-09-27 Exiqon A/S Mediated cellular delivery of lna oligonucleotides
PT2510946E (en) 2007-02-07 2015-11-23 Univ California Conjugates of synthetic tlr agonists and uses therefor
EP1961825A1 (en) 2007-02-26 2008-08-27 INSERM (Institut National de la Santé et de la Recherche Medicale) Method for predicting the occurrence of metastasis in breast cancer patients
PE20090064A1 (en) 2007-03-26 2009-03-02 Novartis Ag DOUBLE-CHAIN RIBONUCLEIC ACID TO INHIBIT THE EXPRESSION OF THE HUMAN E6AP GENE AND THE PHARMACEUTICAL COMPOSITION THAT INCLUDES IT
US20090068660A1 (en) * 2007-04-25 2009-03-12 John Wayne Cancer Institute Use of methylated or unmethylated line-1 dna as a cancer marker
US20100261175A1 (en) 2007-06-15 2010-10-14 Exiqon A/S Use of short oligonucleotides for reagent redundancy experiments in rna functional analysis
CN101969987A (en) 2007-09-20 2011-02-09 J·大卫格莱斯顿学会 Long interspersed nuclear element polypeptide compositions and methods of use thereof
EP2623598B1 (en) 2007-10-04 2018-08-01 Roche Innovation Center Copenhagen A/S Micromirs
WO2009048530A2 (en) 2007-10-05 2009-04-16 Panomics, Inc. Highly multiplexed particle-based assays
UA100253C2 (en) 2007-11-26 2012-12-10 Сантаріс Фарма А/С Androgenic receptor lna-antagonists
EP2220091B1 (en) 2007-12-14 2012-07-25 Ardea Biosciences, Inc. Reverse transcriptase inhibitors
WO2009090639A2 (en) 2008-01-15 2009-07-23 Quark Pharmaceuticals, Inc. Sirna compounds and methods of use thereof
US8361980B2 (en) 2008-03-07 2013-01-29 Santaris Pharma A/S Pharmaceutical compositions for treatment of microRNA related diseases
US8597946B2 (en) 2008-03-10 2013-12-03 H. Lee Moffitt Caner Center and Research Institute, Inc. Enhanced dendritic cells for cancer immunotherapy
SI2342226T1 (en) 2008-09-26 2016-11-30 Dana-Farber Cancer Institute Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses thereof
EP2351739B1 (en) 2008-10-09 2013-07-31 Institute Of Pharmacology And Toxicology Academy Of Military Medical Sciences P.L.A. China 2-(4-substituted phenylamino) polysubstituted pyridine compounds as the inhibitors of non-nucleoside hiv reverse transcriptase, praparation methods and uses thereof
WO2010135690A1 (en) 2009-05-21 2010-11-25 The Regents Of The University Of Michigan Antiviral treatment of lymphoma and cancer
KR20120107456A (en) 2009-07-30 2012-10-02 안티센스 파마 게엠베하 Combination of a chemotherapeutic agent and an inhibitor of the tgf-beta system
WO2011038403A1 (en) 2009-09-28 2011-03-31 Yuling Luo Methods of detecting nucleic acid sequences with high specificity
US20130171621A1 (en) 2010-01-29 2013-07-04 Advanced Cell Diagnostics Inc. Methods of in situ detection of nucleic acids
US20110218170A1 (en) 2010-03-02 2011-09-08 Southern Research Institute Use of 2'-deoxy-4'-thiocytidine and its analogues as dna hypomethylating anticancer agents
MX2012011379A (en) 2010-03-30 2012-11-30 Merck Canada Inc Non-nucleoside reverse transcriptase inhibitors.
SI3178851T1 (en) 2010-03-31 2020-09-30 Boehringer Inhelheim International Gmbh Anti-cd40 antibodies
KR20130043104A (en) 2010-04-06 2013-04-29 카리스 라이프 사이언스 룩셈부르크 홀딩스 Circulating biomarkers for disease
US20120052498A1 (en) 2010-07-01 2012-03-01 Affymetrix, Inc. Detection of Nucleic Acids
US20120003648A1 (en) 2010-07-01 2012-01-05 Affymetrix, Inc. Signal Multiplexing and Signal Amplification
US20120004132A1 (en) 2010-07-02 2012-01-05 Affymetrix, Inc. Detection of Nucleic Acids and Proteins
EP2619321B1 (en) 2010-09-20 2018-08-01 Advanced Cell Diagnostics, Inc. Biomarkers for differentiating melanoma from benign nevus in the skin
CN103517990A (en) 2010-10-07 2014-01-15 通用医疗公司 Biomarkers of cancer
ES2648564T3 (en) 2010-10-21 2018-01-04 Advanced Cell Diagnostics, Inc. Ultrasensitive method for in situ detection of nucleic acids
US20120172246A1 (en) 2010-12-31 2012-07-05 Affymetrix, Inc. Detection of Nucleic Acids
EP2668290B1 (en) 2011-01-28 2017-05-31 Advanced Cell Diagnostics, Inc. Rnascope® hpv assay for determining hpv status in head and neck cancers and cervical lesions
JP5458188B2 (en) 2011-02-17 2014-04-02 協和発酵キリン株式会社 High concentration formulation of anti-CD40 antibody
CN103635488B (en) 2011-04-29 2016-12-14 埃派斯进有限公司 Anti-CD 40 antibodies and using method thereof
TWI454478B (en) 2011-05-13 2014-10-01 Academia Sinica Tlr-2 agonists and use thereof for stimulating immune responses
US20120321637A1 (en) 2011-06-20 2012-12-20 The Board Of Regents Of The University Of Texas System Combination cancer therapy with herv inhibition
WO2013035114A1 (en) 2011-09-08 2013-03-14 Decode Genetics Ehf Tp53 genetic variants predictive of cancer
GB2510539B (en) 2011-10-24 2016-04-20 Gen Hospital Corp Biomarkers of cancer
DK2785375T3 (en) 2011-11-28 2020-10-12 Merck Patent Gmbh ANTI-PD-L1 ANTIBODIES AND USES THEREOF
JP6448533B2 (en) 2012-05-15 2019-01-09 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Cancer immunotherapy by disrupting PD-1 / PD-L1 signaling
JO3470B1 (en) 2012-10-08 2020-07-05 Merck Sharp & Dohme 5-phenoxy-3h-pyrimidin-4-one derivatives and their use as hiv reverse transcriptase inhibitors
SI2914254T1 (en) 2012-10-30 2020-07-31 Mei Pharma, Inc. Combination therapies to treat chemoresistant cancers
CA2888763A1 (en) 2012-10-30 2014-05-08 Apexigen, Inc. Anti-cd40 antibodies and methods of use
US20150290235A1 (en) 2012-11-23 2015-10-15 Ab Science Use of small molecule inhibitors/activators in combination with (deoxy)nucleoside or (deoxy)nucleotide analogs for treatment of cancer and hematological malignancies or viral infections
WO2014133085A1 (en) 2013-02-27 2014-09-04 国立大学法人京都大学 Pharmaceutical composition for prevention and/or treatment of cancer
US10119133B2 (en) 2013-03-15 2018-11-06 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
JP2016520643A (en) 2013-06-03 2016-07-14 ノバルティス アーゲー Combination of anti-PD-L1 antibody and MEK inhibitor and / or BRAF inhibitor
US9382245B2 (en) 2013-10-11 2016-07-05 Yale University Compounds and methods for treating HIV infections
SG11201606819QA (en) 2014-02-18 2016-09-29 Univ Duke Compositions for the inactivation of virus replication and methods of making and using the same
US10301624B2 (en) 2014-06-25 2019-05-28 The General Hospital Corporation Targeting human satellite II (HSATII)
RU2715038C2 (en) 2014-07-11 2020-02-21 Дженентек, Инк. Anti-pd-l1 antibodies and methods for their diagnostic use
TWI716362B (en) 2014-10-14 2021-01-21 瑞士商諾華公司 Antibody molecules to pd-l1 and uses thereof
AR105654A1 (en) 2015-08-24 2017-10-25 Lilly Co Eli ANTIBODIES PD-L1 (LINKING 1 OF PROGRAMMED CELL DEATH)
US9512446B1 (en) 2015-08-28 2016-12-06 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US20170267667A1 (en) 2016-03-15 2017-09-21 Purdue Research Foundation Alkenyldiarylmethanes as non-nucleoside reverse transcriptase inhibitors for anti-hiv-1 chemotherapy
EP3488001A1 (en) 2016-07-25 2019-05-29 Mayo Foundation for Medical Education and Research Treating cancer

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Choi I-S. et al. Modern Pathology (2007) 20, 802–810 *
Florl A.R. et al. British Journal of Cancer (1999) 80(9), 1312–1321. *
Horad B. et al Epigenetics 4:5, 339-350; 1 July 2009 *
Player A. et al. The Journal of Histochemistry & Cytochemistry, Volume 49(5): 603–611, 2001 *
Yonezawa S. et al.Human Pathology, Volume 28, Issue 3, March 1997, Pages 344–352 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10613089B2 (en) 2006-01-30 2020-04-07 The Scripps Research Institute Method of using non-rare cells to detect rare cells
US11142800B2 (en) 2010-10-07 2021-10-12 The General Hospital Corporation Biomarkers of cancer
US20140213475A1 (en) * 2011-07-14 2014-07-31 University Of Massachusetts Methods of diagnosing cancer using epigenetic biomarkers
US10527624B2 (en) 2014-01-27 2020-01-07 Epic Sciences, Inc. Circulating tumor cell diagnostics for prostate cancer biomarkers
US10545151B2 (en) 2014-02-21 2020-01-28 Epic Sciences, Inc. Methods for analyzing rare circulating cells
US11340228B2 (en) 2014-02-21 2022-05-24 Epic Sciences, Inc. Methods for analyzing rare circulating cells
EP3256608A4 (en) * 2015-02-13 2019-02-20 Icahn School of Medicine at Mount Sinai Rna containing compositions and methods of their use
WO2016196284A1 (en) * 2015-05-29 2016-12-08 Epic Sciences, Inc. Intra-patient genomic heterogeneity of single circulating tumor cells (ctcs) associated to phenotypic ctc heterogeneity in metastatic castrate resistant prostate cancer (mcrpc)

Also Published As

Publication number Publication date
EP2625292A4 (en) 2014-03-19
EP2625292A2 (en) 2013-08-14
US20170356054A1 (en) 2017-12-14
EP2625292B1 (en) 2018-12-05
CN103517990A (en) 2014-01-15
US20220098678A1 (en) 2022-03-31
WO2012048113A3 (en) 2012-07-26
US11142800B2 (en) 2021-10-12
WO2012048113A2 (en) 2012-04-12

Similar Documents

Publication Publication Date Title
US20220098678A1 (en) Biomarkers of Cancer
Hunter et al. Divergent routes to oral cancer
Ginestier et al. Prognosis and gene expression profiling of 20q13-amplified breast cancers
Wang et al. Wdr66 is a novel marker for risk stratification and involved in epithelial-mesenchymal transition of esophageal squamous cell carcinoma
Li et al. MicroRNAs involved in neoplastic transformation of liver cancer stem cells
US20170292942A1 (en) Biomarkers of Cancer
Bender et al. Sleeping beauty–mediated somatic mutagenesis implicates csf1 in the formation of high-grade astrocytomas
Casanova-Salas et al. MiR-187 targets the androgen-regulated gene ALDH1A3 in prostate cancer
Pongor et al. Extrachromosomal DNA amplification contributes to small cell lung cancer heterogeneity and is associated with worse outcomes
WO2017136508A1 (en) Dissociation of human tumor to single cell suspension followed by biological analysis
White-Al Habeeb et al. Alpha-enolase is a potential prognostic marker in clear cell renal cell carcinoma
JP2011517937A (en) In vitro diagnostic method for diagnosis of somatic and ovarian cancer
De Palma et al. The abundance of the long intergenic non-coding RNA 01087 differentiates between luminal and triple-negative breast cancers and predicts patient outcome
Pavlič et al. Tumour budding and poorly differentiated clusters in colon cancer–different manifestations of partial epithelial–mesenchymal transition
Bosotti et al. Establishment and genomic characterization of the new chordoma cell line Chor-IN-1
CA2989497A1 (en) Methods for diagnosis of bladder cancer
Morotti et al. The oncosuppressors MEN1 and CDC73 are involved in lncRNA deregulation in human parathyroid tumors
WO2019169336A1 (en) Methods for prostate cancer detection
Carofino et al. Head and neck squamous cancer progression is marked by CLIC4 attenuation in tumor epithelium and reciprocal stromal upregulation of miR-142-3p, a novel post-transcriptional regulator of CLIC4
Dai et al. Genome-wide analysis of barrett's adenocarcinoma. a first step towards identifying patients at risk and developing therapeutic paths
Bojovic et al. Dysfunctional telomeres promote genomic instability and metastasis in the absence of telomerase activity in oncogene induced mammary cancer
US20210285053A1 (en) L1td1 as predictive biomarker of colon cancer
TWI651414B (en) Biomarkers, probes, kits for detecting biological samples and methods for non-invasive qualitative determination of the degree of deterioration of endometriosis
Kowalewska et al. Estimation of groin recurrence risk in patients with squamous cell vulvar carcinoma by the assessment of marker gene expression in the lymph nodes
KR102384992B1 (en) Age-specific biomarker of a patient with colorectal cancer and use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HABER, DANIEL A.;REEL/FRAME:037563/0109

Effective date: 20130430

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TING, DAVID TSAI;MAHESWARAN, SHYAMALA;REEL/FRAME:037562/0979

Effective date: 20130430

STCB Information on status: application discontinuation

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