US10385406B2 - Detection of lung neoplasia by analysis of methylated DNA - Google Patents

Detection of lung neoplasia by analysis of methylated DNA Download PDF

Info

Publication number
US10385406B2
US10385406B2 US15/471,337 US201715471337A US10385406B2 US 10385406 B2 US10385406 B2 US 10385406B2 US 201715471337 A US201715471337 A US 201715471337A US 10385406 B2 US10385406 B2 US 10385406B2
Authority
US
United States
Prior art keywords
methylation
dna
max
sample
marker
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.)
Active, expires
Application number
US15/471,337
Other languages
English (en)
Other versions
US20170335401A1 (en
Inventor
Hatim Allawi
Graham P. Lidgard
Maria Giakoumopoulos
David A. Ahlquist
William R. Taylor
Douglas Mahoney
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Mayo Foundation for Medical Education and Research
Exact Sciences Corp
Original Assignee
Mayo Foundation for Medical Education and Research
Exact Sciences Development Co LLC
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
Priority to US15/471,337 priority Critical patent/US10385406B2/en
Application filed by Mayo Foundation for Medical Education and Research, Exact Sciences Development Co LLC filed Critical Mayo Foundation for Medical Education and Research
Assigned to EXACT SCIENCES CORPORATION reassignment EXACT SCIENCES CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ALLAWI, HATIM, GIAKOUMOPOULOS, Maria, LIDGARD, GRAHAM P.
Assigned to MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH reassignment MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TAYLOR, WILLIAM R., AHLQUIST, DAVID A., MAHONEY, DOUGLAS
Assigned to EXACT SCIENCES DEVELOPMENT COMPANY, LLC reassignment EXACT SCIENCES DEVELOPMENT COMPANY, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EXACT SCIENCES CORPORATION
Publication of US20170335401A1 publication Critical patent/US20170335401A1/en
Priority to US16/460,647 priority patent/US11028447B2/en
Publication of US10385406B2 publication Critical patent/US10385406B2/en
Application granted granted Critical
Priority to US17/314,099 priority patent/US20210262042A1/en
Assigned to EXACT SCIENCES CORPORATION reassignment EXACT SCIENCES CORPORATION MERGER (SEE DOCUMENT FOR DETAILS). Assignors: EXACT SCIENCES DEVELOPMENT COMPANY, LLC
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • 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
    • C12Q2600/154Methylation markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/16Primer sets for multiplex assays

Definitions

  • neoplasia Provided herein is technology relating to detecting neoplasia and particularly, but not exclusively, to methods, compositions, and related uses for detecting neoplasms such as lung cancer.
  • Lung cancer remains the number one cancer killer in the US, and effective screening approaches are urgent needed. Lung cancer alone accounts for 221,000 deaths annually. DNA methylation profiling has shown unique patterns in DNA promoter regions with cancer and has potential application for detection of lung malignancies. However, optimally discriminant markers and marker panels are needed.
  • markers selected from the collection can be used alone or in a panel, for example, to characterize blood or bodily fluid, with applications in lung cancer screening and discrimination of malignant from benign nodules.
  • markers from the panel are used to distinguish one form of lung cancer from another, e.g., for distinguishing the presence of a lung adenocarcinoma or large cell carcinoma from the presence of a lung small cell carcinoma, or for detecting mixed pathology carcinomas.
  • technology for screening markers that provide a high signal-to-noise ratio and a low background level when detected from samples taken from a subject.
  • Methylation markers and/or panels of markers having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST8SIA_22, FLJ45983, DLX4, SHOX2,
  • the technology provides a number of methylation markers and subsets thereof (e.g., sets of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more markers) with high discrimination for lung cancer and, in some embodiments, with discrimination between lung cancer types.
  • a selection filter applied to candidate markers to identify markers that provide a high signal to noise ratio and a low background level to provide high specificity and selectivity for purposes of characterizing biological samples, e.g., for cancer screening or diagnosis.
  • a panel of 6 markers (SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI) resulted in a sensitivity of 92.2% at 93% specificity
  • a panel of 4 markers (ZNF781, BARX1, EMX1, and HOXA9) resulted in an overall sensitivity of 96% and specificity of 94%.
  • the methylation state of the methylation marker is determined by measuring the amounts of a methylated marker and of a reference marker in the sample, and comparing the amount of the methylated marker to the amount of reference marker in the sample to determine a methylation state for the methylation marker in the sample.
  • the method finds use, e.g., in characterizing samples from a subject having or suspected of having lung cancer, when the methylation state of the methylation marker is different than a methylation state of that marker assayed in a subject that does not have a neoplasm.
  • the methylation marker comprises a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST8SIA_22, FLJ45983, DLX4, SHOX2, EMX1, HOXB2,
  • the technology comprises assaying a plurality of markers, e.g., comprising assaying the methylation states of 2 to 21 markers, preferably 2 to 8 markers, preferably 4 to 6 markers.
  • the method comprises analysis of the methylation status of two or more markers selected from SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, EMX1, CYP26C1, SOBP, SUCLG2, SHOX2, ZDHHC1, NFIX, FLJ45983, HOXA9, B3GALT6, ZNF781, SP9, BARX1, and SKI.
  • the method comprises analysis of the methylation status of a set of markers comprising SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, and EMX1.
  • the method comprises analysis of the methylation status of a set of markers selected from: the group consisting of ZNF781, BARX1, and EMX1; the group consisting of SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI; the group consisting of SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, and EMX1; the group consisting of SHOX2, SOBP, ZNF781, BTACT, CYP26C1, and DLX4; and the group consisting of SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI.
  • the at least one methylation marker comprises the group selected from ZNF781, BARX1, and EMX1, and further comprises SOBP and/or HOXA9.
  • assessing the methylation state of the methylation marker in the sample comprises determining the methylation state of one base.
  • assaying the methylation state of the marker in the sample comprises determining the extent of methylation at a plurality of bases.
  • the methylation state of the marker comprises an increased methylation of the marker relative to a normal methylation state of the marker.
  • the methylation state of the marker comprises a decreased methylation of the marker relative to a normal methylation state of the marker.
  • the methylation state of the marker comprises a different pattern of methylation of the marker relative to a normal methylation state of the marker.
  • the technology provides a method of generating a record reporting a lung neoplasm in a subject, the method comprising the steps of:
  • the sample is assayed for at least two of the markers, and preferably the at least two methylated marker genes are selected from the group consisting of SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, EMX1 CYP26C1, SOBP, SUCLG2, SHOX2, ZDHHC1, NFIX, FLJ45983, HOXA9, B3GALT6, ZNF781, SP9, BARX1, and SKI.
  • the at least two methylated marker genes are selected from the group consisting of SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, EMX1 CYP26C1, SOBP, SUCLG2, SHOX2, ZDHHC1, NFIX, FLJ45983, HOXA9, B3GALT6, ZNF781, SP9, BARX1, and SKI.
  • the method comprises analysis of the methylation status of a set of markers selected from: the group consisting of ZNF781, BARX1, and EMX1; the group consisting of SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI; the group consisting of SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, and EMX1; the group consisting of SHOX2, SOBP, ZNF781, BTACT, CYP26C1, and DLX4; and the group consisting of SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI.
  • a set of markers selected from: the group consisting of ZNF781, BARX1, and EMX1; the group consisting of SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI; the group consisting of SLC12A8, KLHDC
  • the at least one methylation marker comprises the group selected from ZNF781, BARX1, and EMX1, and further comprises SOBP and/or HOXA9.
  • methylation markers are selected such that the methylation status of said one or more markers is indicative of only one of lung adenocarcinoma, large cell carcinoma, squamous cell carcinoma, or small cell carcinoma.
  • methylation markers are selected such that the methylation status of said one or more markers is indicative of more than one of lung adenocarcinoma, large cell carcinoma, squamous cell carcinoma, and small cell carcinoma.
  • methylation markers are selected such that the methylation status of said one or more markers is indicative of any one of or combination of lung adenocarcinoma, large cell carcinoma, squamous cell carcinoma, small cell carcinoma, generic non-small cell lung cancer, and/or undefined lung carcinoma.
  • the method used for assaying comprises obtaining a sample comprising DNA from a subject, and treating DNA obtained from the sample with a reagent that selectively modifies unmethylated cytosine residues in the obtained DNA to produce modified residues.
  • the reagent comprises a bisulfate reagent.
  • assaying the methylation state of the methylation marker in the sample comprises determining the methylation state of one base, while in other embodiments the assay comprises determining the extent of methylation at a plurality of bases.
  • the methylation state of the marker comprises an increased or decreased methylation of the marker relative to a normal methylation state of the marker, e.g., as the marker would appear in a non-cancerous sample, while in some embodiments the methylation state of the marker comprises a different pattern of methylation of the marker relative to a normal methylation state of the marker.
  • the reference marker is a methylated reference marker.
  • the sample is a tissue sample, a blood sample, a plasma sample, a serum sample, or a sputum sample.
  • a tissue sample comprises lung tissue.
  • the sample comprises DNA isolated from plasma.
  • the technology is not limited to any particular method of assaying DNA from samples.
  • the assaying comprises using polymerase chain reaction, nucleic acid sequencing, mass spectrometry, methylation specific nuclease, mass-based separation, and/or target capture.
  • the assaying comprises using a flap endonuclease assay.
  • sample DNA and/or reference marker DNA are bisulfite-converted and the assay for determining the methylation level of the DNA is achieved by a technique comprising the use of methylation-specific PCR, quantitative methylation-specific PCR, methylation-sensitive DNA restriction enzyme analysis, quantitative bisulfite pyrosequencing, flap endonuclease assay (e.g., a QUARTS flap endonuclease assay), and/or bisulfite genomic sequencing PCR.
  • a technique comprising the use of methylation-specific PCR, quantitative methylation-specific PCR, methylation-sensitive DNA restriction enzyme analysis, quantitative bisulfite pyrosequencing, flap endonuclease assay (e.g., a QUARTS flap endonuclease assay), and/or bisulfite genomic sequencing PCR.
  • kits comprising a) at least one oligonucleotide, wherein at least a portion of the oligonucleotide specifically hybridizes to a marker selected from the group consisting of BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GR
  • the portion of the oligonucleotide that hybridizes to the marker specifically hybridizes to bisulfite-treated DNA comprising the methylation marker.
  • the kit comprises at least one additional oligonucleotide, wherein at least a portion of the additional oligonucleotide specifically hybridizes to a reference nucleic acid.
  • the kit comprises at least two additional oligonucleotides and, in some embodiments, the kit further comprises a bisulfite reagent.
  • At least a portion of the oligonucleotide specifically hybridizes to a least one the marker selected from the group consisting of SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, EMX1, CYP26C1, SOBP, SUCLG2, SHOX2, ZDHHC1, NFIX, FLJ45983, HOXA9, B3GALT6, ZNF781, SP9, BARX1, and SKI.
  • a marker selected from the group consisting of SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, EMX1, CYP26C1, SOBP, SUCLG2, SHOX2, ZDHHC1, NFIX, FLJ45983, HOXA9, B3GALT6, ZNF781, SP9, BARX1, and SKI.
  • the kit comprises a set of oligonucleotides, each of which hybridizes to one marker in a set of markers, the set of markers selected from: the group consisting of ZNF781, BARX1, and EMX1; the group consisting of SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI the group consisting of SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, and EMX1; the group consisting of SHOX2, SOBP, ZNF781, BTACT, CYP26C1, and DLX4; and the group consisting of SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI.
  • the set of methylation markers comprises the group selected from ZNF781, BARX1, and EMX1, and further comprises SOBP and/or HOXA9.
  • the at least one oligonucleotide in the kit is selected to hybridize to methylation marker(s) that are indicative of only one of type of lung carcinoma, e.g., lung adenocarcinoma, large cell carcinoma, squamous cell carcinoma, or small cell carcinoma. In other embodiments, the at least one oligonucleotide is selected to hybridize to methylation marker(s) that are indicative of more than one of lung adenocarcinoma, large cell carcinoma, squamous cell carcinoma, and small cell carcinoma.
  • the at least one oligonucleotide is selected to hybridize to methylation marker(s) that are indicative of any one of, or any combination of lung adenocarcinoma, large cell carcinoma, squamous cell carcinoma, small cell carcinoma, and/or undefined lung carcinoma.
  • oligonucleotide(s) provided in the kit are selected from one or more of a capture oligonucleotide, a pair of nucleic acid primers, a nucleic acid probe, and an invasive oligonucleotide. In preferred embodiments, oligonucleotide(s) specifically hybridize to bisulfite-treated DNA comprising said methylation marker(s).
  • the kit further comprises a solid support, such a magnetic bead or particle.
  • a solid support comprises one or more capture reagents, e.g., oligonucleotides complementary said one or more markers genes.
  • compositions comprising a mixture, e.g., a reaction mixture, that comprises a complex of a target nucleic acid selected from the group consisting of BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PR
  • the target nucleic acid is bisulfite-converted target nucleic acid.
  • the mixture comprises a complex of a target nucleic acid selected from the group consisting of SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, EMX1, CYP26C1, SOBP, SUCLG2, SHOX2, ZDHHC1, NFIX, FLJ45983, HOXA9, B3GALT6, ZNF781, SP9, BARX1, and SKI, and an oligonucleotide that specifically hybridizes to the target nucleic acid (whether unconverted or bisulfite-converted).
  • Oligonucleotides in the mixture include but are not limited to one or more of a capture oligonucleotide, a pair of nucleic acid primers, a hybridization probe, a hydrolysis probe, a flap assay probe, and an invasive oligonucleotide.
  • the target nucleic acid in the mixture comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 1, 6, 11, 16, 21, 28, 33, 38, 43, 48, 53, 58, 63, 68, 73, 78, 86, 91, 96, 101, 106, 111, 116, 121, 126, 131, 136, 141, 146, 151, 156, 161, 166, 171, 176, 181, 186, 191, 196, 201, 214, 219, 224, 229, 234, 239, 247, 252, 257, 262, 267, 272, 277, 282, 287, 292, 298, 303, 308, 313, 319, 327, 336, 341, 346, 351, 356, 361, 366, 371, 384, and 403.
  • the mixture comprises bisulfate-converted target nucleic acid that comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 2, 7, 12, 17, 22, 29, 34, 39, 44, 49, 54, 59, 64, 69, 74, 79, 87, 92, 97, 102, 107, 112, 117, 122, 127, 132, 137, 142, 147, 152, 157, 162, 167, 172, 177, 182, 187, 192, 197, 202, 210, 215, 220, 225, 230, 235, 240, 248, 253, 258, 263, 268, 273, 278, 283, 288, 293, 299, 304, 309, 314, 320, 328, 337, 342, 347, 352, 357, 362, 367, 372, 385, and 404.
  • an oligonucleotide in said mixture comprises a reporter molecule, and in preferred embodiments, the reporter molecule comprises a fluorophore. In some embodiments the oligonucleotide comprises a flap sequence. In some embodiments the mixture further comprises one or more of a FRET cassette; a FEN-1 endonuclease and/or a thermostable DNA polymerase, preferably a bacterial DNA polymerase.
  • the term “or” is an inclusive “or” operator and is equivalent to the term “and/or” unless the context clearly dictates otherwise.
  • the term “based on” is not exclusive and allows for being based on additional factors not described, unless the context clearly dictates otherwise.
  • the meaning of “a”, “an”, and “the” include plural references.
  • the meaning of “in” includes “in” and “on.”
  • composition “consisting essentially of” as used in claims in the present application limits the scope of a claim to the specified materials or steps “and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention, as discussed in In re Herz, 537 E2d 549, 551-52, 190 USPQ 461, 463 (CCPR 1976).
  • a composition “consisting essentially of” recited elements may contain an unrecited contaminant at a level such that, though present, the contaminant does not alter the function of the recited composition as compared to a pure composition, i.e., a composition “consisting of” the recited components.
  • methylation refers to cytosine methylation at positions C5 or N4 of cytosine, the N6 position of adenine, or other types of nucleic acid methylation.
  • In vitro amplified DNA is usually unmethylated because typical in vitro DNA amplification methods do not retain the methylation pattern of the amplification template.
  • unmethylated DNA or “methylated DNA” can also refer to amplified DNA whose original template was unmethylated or methylated, respectively.
  • a “methylated nucleotide” or a “methylated nucleotide base” refers to the presence of a methyl moiety on a nucleotide base, where the methyl moiety is not present in a recognized typical nucleotide base.
  • cytosine does not contain a methyl moiety on its pyrimidine ring, but 5-methylcytosine contains a methyl moiety at position 5 of its pyrimidine ring. Therefore, cytosine is not a methylated nucleotide and 5-methylcytosine is a methylated nucleotide.
  • thymine contains a methyl moiety at position 5 of its pyrimidine ring; however, for purposes herein, thymine is not considered a methylated nucleotide when present in DNA since thymine is a typical nucleotide base of DNA.
  • a “methylated nucleic acid molecule” refers to a nucleic acid molecule that contains one or more methylated nucleotides.
  • a “methylation state”, “methylation profile”, and “methylation status” of a nucleic acid molecule refers to the presence of absence of one or more methylated nucleotide bases in the nucleic acid molecule.
  • a nucleic acid molecule containing a methylated cytosine is considered methylated (e.g., the methylation state of the nucleic acid molecule is methylated).
  • a nucleic acid molecule that does not contain any methylated nucleotides is considered unmethylated.
  • a nucleic acid may be characterized as “unmethylated” if it is not methylated at a specific locus (e.g., the locus of a specific single CpG dinucleotide) or specific combination of loci, even if it is methylated at other loci in the same gene or molecule.
  • a specific locus e.g., the locus of a specific single CpG dinucleotide
  • specific combination of loci even if it is methylated at other loci in the same gene or molecule.
  • the methylation state of a particular nucleic acid sequence can indicate the methylation state of every base in the sequence or can indicate the methylation state of a subset of the bases (e.g., of one or more cytosines) within the sequence, or can indicate information regarding regional methylation density within the sequence with or without providing precise information of the locations within the sequence the methylation occurs.
  • the terms “marker gene” and “marker” are used interchangeably to refer to DNA that is associated with a condition, e.g., cancer, regardless of whether the marker region is in a coding region of DNA. Markers may include, e.g., regulatory regions, flanking regions, intergenic regions, etc.
  • the methylation state of a nucleotide locus in a nucleic acid molecule refers to the presence or absence of a methylated nucleotide at a particular locus in the nucleic acid molecule.
  • the methylation state of a cytosine at the 7th nucleotide in a nucleic acid molecule is methylated when the nucleotide present at the 7th nucleotide in the nucleic acid molecule is 5-methylcytosine.
  • the methylation state of a cytosine at the 7th nucleotide in a nucleic acid molecule is unmethylated when the nucleotide present at the 7th nucleotide in the nucleic acid molecule is cytosine (and not 5-methylcytosine).
  • the methylation status can optionally be represented or indicated by a “methylation value” (e.g., representing a methylation frequency, fraction, ratio, percent, etc.)
  • a methylation value can be generated, for example, by quantifying the amount of intact nucleic acid present following restriction digestion with a methylation dependent restriction enzyme or by comparing amplification profiles after bisulfite reaction or by comparing sequences of bisulfite-treated and untreated nucleic acids. Accordingly, a value, e.g., a methylation value, represents the methylation status and can thus be used as a quantitative indicator of methylation status across multiple copies of a locus. This is of particular use when it is desirable to compare the methylation status of a sequence in a sample to a threshold or reference value.
  • methylation frequency or “methylation percent (%)” refer to the number of instances in which a molecule or locus is methylated relative to the number of instances the molecule or locus is unmethylated.
  • the methylation state describes the state of methylation of a nucleic acid (e.g., a genomic sequence).
  • the methylation state refers to the characteristics of a nucleic acid segment at a particular genomic locus relevant to methylation. Such characteristics include, but are not limited to, whether any of the cytosine (C) residues within this DNA sequence are methylated, the location of methylated C residue(s), the frequency or percentage of methylated C throughout any particular region of a nucleic acid, and allelic differences in methylation due to, e.g., difference in the origin of the alleles.
  • C cytosine
  • methylation state also refer to the relative concentration, absolute concentration, or pattern of methylated C or unmethylated C throughout any particular region of a nucleic acid in a biological sample.
  • cytosine (C) residue(s) within a nucleic acid sequence are methylated it may be referred to as “hypermethylated” or having “increased methylation”
  • cytosine (C) residue(s) within a DNA sequence are not methylated it may be referred to as “hypomethylated” or having “decreased methylation”.
  • cytosine (C) residue(s) within a nucleic acid sequence are methylated as compared to another nucleic acid sequence (e.g., from a different region or from a different individual, etc.) that sequence is considered hypermethylated or having increased methylation compared to the other nucleic acid sequence.
  • the cytosine (C) residue(s) within a DNA sequence are not methylated as compared to another nucleic acid sequence (e.g., from a different region or from a different individual, etc.) that sequence is considered hypomethylated or having decreased methylation compared to the other nucleic acid sequence.
  • methylation pattern refers to the collective sites of methylated and unmethylated nucleotides over a region of a nucleic acid.
  • Two nucleic acids may have the same or similar methylation frequency or methylation percent but have different methylation patterns when the number of methylated and unmethylated nucleotides is the same or similar throughout the region but the locations of methylated and unmethylated nucleotides are different.
  • Sequences are said to be “differentially methylated” or as having a “difference in methylation” or having a “different methylation state” when they differ in the extent (e.g., one has increased or decreased methylation relative to the other), frequency, or pattern of methylation.
  • the term “differential methylation” refers to a difference in the level or pattern of nucleic acid methylation in a cancer positive sample as compared with the level or pattern of nucleic acid methylation in a cancer negative sample. It may also refer to the difference in levels or patterns between patients that have recurrence of cancer after surgery versus patients who not have recurrence. Differential methylation and specific levels or patterns of DNA methylation are prognostic and predictive biomarkers, e.g., once the correct cut-off or predictive characteristics have been defined.
  • Methylation state frequency can be used to describe a population of individuals or a sample from a single individual.
  • a nucleotide locus having a methylation state frequency of 50% is methylated in 50% of instances and unmethylated in 50% of instances.
  • Such a frequency can be used, for example, to describe the degree to which a nucleotide locus or nucleic acid region is methylated in a population of individuals or a collection of nucleic acids.
  • the methylation state frequency of the first population or pool will be different from the methylation state frequency of the second population or pool.
  • Such a frequency also can be used, for example, to describe the degree to which a nucleotide locus or nucleic acid region is methylated in a single individual.
  • a frequency can be used to describe the degree to which a group of cells from a tissue sample are methylated or unmethylated at a nucleotide locus or nucleic acid region.
  • nucleotide locus refers to the location of a nucleotide in a nucleic acid molecule.
  • a nucleotide locus of a methylated nucleotide refers to the location of a methylated nucleotide in a nucleic acid molecule.
  • methylation of human DNA occurs on a dinucleotide sequence including an adjacent guanine and cytosine where the cytosine is located 5′ of the guanine (also termed CpG dinucleotide sequences).
  • CpG dinucleotide sequences also termed CpG dinucleotide sequences.
  • Most cytosines within the CpG dinucleotides are methylated in the human genome, however some remain unmethylated in specific CpG dinucleotide rich genomic regions, known as CpG islands (see, e.g., Antequera, et al. (1990) Cell 62: 503-514).
  • a “CpG island” refers to a G:C-rich region of genomic DNA containing an increased number of CpG dinucleotides relative to total genomic DNA.
  • a CpG island can be at least 100, 200, or more base pairs in length, where the G:C content of the region is at least 50% and the ratio of observed CpG frequency over expected frequency is 0.6; in some instances, a CpG island can be at least 500 base pairs in length, where the G:C content of the region is at least 55%) and the ratio of observed CpG frequency over expected frequency is 0.65.
  • the observed CpG frequency over expected frequency can be calculated according to the method provided in Gardiner-Garden et al (1987) J. Mol. Biol. 196: 261-281.
  • Methylation state is typically determined in CpG islands, e.g., at promoter regions. It will be appreciated though that other sequences in the human genome are prone to DNA methylation such as CpA and CpT (see Ramsahoye (2000) Proc.
  • a “methylation-specific reagent” refers to a reagent that modifies a nucleotide of the nucleic acid molecule as a function of the methylation state of the nucleic acid molecule, or a methylation-specific reagent, refers to a compound or composition or other agent that can change the nucleotide sequence of a nucleic acid molecule in a manner that reflects the methylation state of the nucleic acid molecule.
  • Methods of treating a nucleic acid molecule with such a reagent can include contacting the nucleic acid molecule with the reagent, coupled with additional steps, if desired, to accomplish the desired change of nucleotide sequence.
  • Such methods can be applied in a manner in which unmethylated nucleotides (e.g., each unmethylated cytosine) is modified to a different nucleotide.
  • a reagent can deaminate unmethylated cytosine nucleotides to produce deoxy uracil residues.
  • An exemplary reagent is a bisulfite reagent.
  • bisulfite reagent refers to a reagent comprising bisulfite, disulfite, hydrogen sulfite, or combinations thereof, useful as disclosed herein to distinguish between methylated and unmethylated CpG dinucleotide sequences.
  • Methods of said treatment are known in the art (e.g., PCT/EP2004/011715 and WO 2013/116375, each of which is incorporated by reference in its entirety).
  • bisulfite treatment is conducted in the presence of denaturing solvents such as but not limited to n-alkyleneglycol or diethylene glycol dimethyl ether (DME), or in the presence of dioxane or dioxane derivatives.
  • the denaturing solvents are used in concentrations between 1% and 35% (v/v).
  • the bisulfite reaction is carried out in the presence of scavengers such as but not limited to chromane derivatives, e.g., 6-hydroxy-2,5,7,8,-tetramethylchromane 2-carboxylic acid or trihydroxybenzone acid and derivates thereof, e.g., Gallic acid (see: PCT/EP2004/011715, which is incorporated by reference in its entirety).
  • the bisulfite reaction comprises treatment with ammonium hydrogen sulfite, e.g., as described in WO 2013/116375.
  • a change in the nucleic acid nucleotide sequence by a methylation—specific reagent can also result in a nucleic acid molecule in which each methylated nucleotide is modified to a different nucleotide.
  • methylation assay refers to any assay for determining the methylation state of one or more CpG dinucleotide sequences within a sequence of a nucleic acid.
  • the “sensitivity” of a given marker refers to the percentage of samples that report a DNA methylation value above a threshold value that distinguishes between neoplastic and non-neoplastic samples.
  • a positive is defined as a histology-confirmed neoplasia that reports a DNA methylation value above a threshold value (e.g., the range associated with disease)
  • a false negative is defined as a histology-confirmed neoplasia that reports a DNA methylation value below the threshold value (e.g., the range associated with no disease).
  • the value of sensitivity therefore, reflects the probability that a DNA methylation measurement for a given marker obtained from a known diseased sample will be in the range of disease-associated measurements.
  • the clinical relevance of the calculated sensitivity value represents an estimation of the probability that a given marker would detect the presence of a clinical condition when applied to a subject with that condition.
  • the “specificity” of a given marker refers to the percentage of non-neoplastic samples that report a DNA methylation value below a threshold value that distinguishes between neoplastic and non-neoplastic samples.
  • a negative is defined as a histology-confirmed non-neoplastic sample that reports a DNA methylation value below the threshold value (e.g., the range associated with no disease) and a false positive is defined as a histology-confirmed non-neoplastic sample that reports a DNA methylation value above the threshold value (e.g., the range associated with disease).
  • the value of specificity therefore, reflects the probability that a DNA methylation measurement for a given marker obtained from a known non-neoplastic sample will be in the range of non-disease associated measurements.
  • the clinical relevance of the calculated specificity value represents an estimation of the probability that a given marker would detect the absence of a clinical condition when applied to a patient without that condition.
  • a “selected nucleotide” refers to one nucleotide of the four typically occurring nucleotides in a nucleic acid molecule (C, G, T, and A for DNA and C, G, U, and A for RNA), and can include methylated derivatives of the typically occurring nucleotides (e.g., when C is the selected nucleotide, both methylated and unmethylated C are included within the meaning of a selected nucleotide), whereas a methylated selected nucleotide refers specifically to a nucleotide that is typically methylated and an unmethylated selected nucleotides refers specifically to a nucleotide that typically occurs in unmethylated form.
  • methylation-specific restriction enzyme or “methylation-sensitive restriction enzyme” refers to an enzyme that selectively digests a nucleic acid dependent on the methylation state of its recognition site.
  • a restriction enzyme that specifically cuts if the recognition site is not methylated or is hemi-methylated, the cut will not take place or will take place with a significantly reduced efficiency if the recognition site is methylated.
  • a restriction enzyme that specifically cuts if the recognition site is methylated, the cut will not take place or will take place with a significantly reduced efficiency if the recognition site is not methylated.
  • methylation-specific restriction enzymes the recognition sequence of which contains a CG dinucleotide (for instance a recognition sequence such as CGCG or CCCGGG). Further preferred for some embodiments are restriction enzymes that do not cut if the cytosine in this dinucleotide is methylated at the carbon atom C5.
  • primer refers to an oligonucleotide, whether occurring naturally as, e.g., a nucleic acid fragment from a restriction digest, or produced synthetically, that is capable of acting as a point of initiation of synthesis when placed under conditions in which synthesis of a primer extension product that is complementary to a nucleic acid template strand is induced, (e.g., in the presence of nucleotides and an inducing agent such as a DNA polymerase, and at a suitable temperature and pH).
  • the primer is preferably single stranded for maximum efficiency in amplification, but may alternatively be double stranded. If double stranded, the primer is first treated to separate its strands before being used to prepare extension products.
  • the primer is an oligodeoxyribonucleotide.
  • the primer must be sufficiently long to prime the synthesis of extension products in the presence of the inducing agent. The exact lengths of the primers will depend on many factors, including temperature, source of primer, and the use of the method.
  • probe refers to an oligonucleotide (e.g., a sequence of nucleotides), whether occurring naturally as in a purified restriction digest or produced synthetically, recombinantly, or by PCR amplification, that is capable of hybridizing to another oligonucleotide of interest.
  • a probe may be single-stranded or double-stranded. Probes are useful in the detection, identification, and isolation of particular gene sequences (e.g., a “capture probe”).
  • any probe used in the present invention may, in some embodiments, be labeled with any “reporter molecule,” so that is detectable in any detection system, including, but not limited to enzyme (e.g., ELISA, as well as enzyme-based histochemical assays), fluorescent, radioactive, and luminescent systems. It is not intended that the present invention be limited to any particular detection system or label.
  • target refers to a nucleic acid sought to be sorted out from other nucleic acids, e.g., by probe binding, amplification, isolation, capture, etc.
  • target refers to the region of nucleic acid bounded by the primers used for polymerase chain reaction
  • a target comprises the site at which a probe and invasive oligonucleotides (e.g., INVADER oligonucleotide) bind to form an invasive cleavage structure, such that the presence of the target nucleic acid can be detected.
  • a “segment” is defined as a region of nucleic acid within the target sequence.
  • marker refers to a substance (e.g., a nucleic acid, or a region of a nucleic acid, or a protein) that may be used to distinguish non-normal cells (e.g., cancer cells) from normal cells (non-cancerous cells), e.g., based on presence, absence, or status (e.g., methylation state) of the marker substance.
  • non-normal cells e.g., cancer cells
  • normal cells e.g., based on presence, absence, or status (e.g., methylation state) of the marker substance.
  • normal methylation of a marker refers to a degree of methylation typically found in normal cells, e.g., in non-cancerous cells.
  • neoplasm refers to any new and abnormal growth of tissue.
  • a neoplasm can be a premalignant neoplasm or a malignant neoplasm.
  • nucleic acid-specific marker refers to any biological material or element that can be used to indicate the presence of a neoplasm.
  • biological materials include, without limitation, nucleic acids, polypeptides, carbohydrates, fatty acids, cellular components (e.g., cell membranes and mitochondria), and whole cells.
  • markers are particular nucleic acid regions (e.g., genes, intragenic regions, specific loci, etc.). Regions of nucleic acid that are markers may be referred to, e.g., as “marker genes,” “marker regions,” “marker sequences,” “marker loci,” etc.
  • sample is used in its broadest sense. In one sense it can refer to an animal cell or tissue. In another sense, it refers to a specimen or culture obtained from any source, as well as biological and environmental samples. Biological samples may be obtained from plants or animals (including humans) and encompass fluids, solids, tissues, and gases. Environmental samples include environmental material such as surface matter, soil, water, and industrial samples. These examples are not to be construed as limiting the sample types applicable to the present invention.
  • the terms “patient” or “subject” refer to organisms to be subject to various tests provided by the technology.
  • the term “subject” includes animals, preferably mammals, including humans.
  • the subject is a primate.
  • the subject is a human.
  • a preferred subject is a vertebrate subject.
  • a preferred vertebrate is warm-blooded; a preferred warm-blooded vertebrate is a mammal.
  • a preferred mammal is most preferably a human.
  • the term “subject’ includes both human and animal subjects. Thus, veterinary therapeutic uses are provided herein.
  • the present technology provides for the diagnosis of mammals such as humans, as well as those mammals of importance due to being endangered, such as Siberian tigers; of economic importance, such as animals raised on farms for consumption by humans; and/or animals of social importance to humans, such as animals kept as pets or in zoos.
  • animals include but are not limited to: carnivores such as cats and dogs; swine, including pigs, hogs, and wild boars; ruminants and/or ungulates such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels; pinnipeds; and horses.
  • the presently-disclosed subject matter further includes a system for diagnosing a lung cancer in a subject.
  • the system can be provided, for example, as a commercial kit that can be used to screen for a risk of lung cancer or diagnose a lung cancer in a subject from whom a biological sample has been collected.
  • An exemplary system provided in accordance with the present technology includes assessing the methylation state of a marker described herein.
  • amplifying or “amplification” in the context of nucleic acids refers to the production of multiple copies of a polynucleotide, or a portion of the polynucleotide, typically starting from a small amount of the polynucleotide (e.g., a single polynucleotide molecule), where the amplification products or amplicons are generally detectable.
  • Amplification of polynucleotides encompasses a variety of chemical and enzymatic processes. The generation of multiple DNA copies from one or a few copies of a target or template DNA molecule during a polymerase chain reaction (PCR) or a ligase chain reaction (LCR; see, e.g., U.S. Pat. No.
  • PCR polymerase chain reaction
  • the mixture is denatured and the primers then annealed to their complementary sequences within the target molecule.
  • the primers are extended with a polymerase so as to form a new pair of complementary strands.
  • the steps of denaturation, primer annealing, and polymerase extension can be repeated many times (i.e., denaturation, annealing and extension constitute one “cycle”; there can be numerous “cycles”) to obtain a high concentration of an amplified segment of the desired target sequence.
  • the length of the amplified segment of the desired target sequence is determined by the relative positions of the primers with respect to each other, and therefore, this length is a controllable parameter.
  • PCR polymerase chain reaction
  • nucleic acid detection assay refers to any method of determining the nucleotide composition of a nucleic acid of interest.
  • Nucleic acid detection assay include but are not limited to, DNA sequencing methods, probe hybridization methods, structure specific cleavage assays (e.g., the INVADER assay, (Hologic, Inc.) and are described, e.g., in U.S. Pat. Nos. 5,846,717, 5,985,557, 5,994,069, 6,001,567, 6,090,543, and 6,872,816; Lyamichev et al., Nat.
  • target nucleic acid is amplified (e.g., by PCR) and amplified nucleic acid is detected simultaneously using an invasive cleavage assay.
  • Assays configured for performing a detection assay e.g., invasive cleavage assay
  • an amplification assay are described in U.S. Pat. No. 9,096,893, incorporated herein by reference in its entirety for all purposes.
  • Additional amplification plus invasive cleavage detection configurations, termed the QUARTS method are described in, e.g., in U.S. Pat. Nos. 8,361,720; 8,715,937; 8,916,344; and 9,212,392, each of which is incorporated herein by reference for all purposes.
  • invasive cleavage structure refers to a cleavage structure comprising i) a target nucleic acid, ii) an upstream nucleic acid (e.g., an invasive or “INVADER” oligonucleotide), and iii) a downstream nucleic acid (e.g., a probe), where the upstream and downstream nucleic acids anneal to contiguous regions of the target nucleic acid, and where an overlap forms between the a 3′ portion of the upstream nucleic acid and duplex formed between the downstream nucleic acid and the target nucleic acid.
  • an upstream nucleic acid e.g., an invasive or “INVADER” oligonucleotide
  • a downstream nucleic acid e.g., a probe
  • an overlap occurs where one or more bases from the upstream and downstream nucleic acids occupy the same position with respect to a target nucleic acid base, whether or not the overlapping base(s) of the upstream nucleic acid are complementary with the target nucleic acid, and whether or not those bases are natural bases or non-natural bases.
  • the 3′ portion of the upstream nucleic acid that overlaps with the downstream duplex is a non-base chemical moiety such as an aromatic ring structure, e.g., as disclosed, for example, in U.S. Pat. No. 6,090,543, incorporated herein by reference in its entirety.
  • one or more of the nucleic acids may be attached to each other, e.g., through a covalent linkage such as nucleic acid stem-loop, or through a non-nucleic acid chemical linkage (e.g., a multi-carbon chain).
  • a covalent linkage such as nucleic acid stem-loop
  • a non-nucleic acid chemical linkage e.g., a multi-carbon chain.
  • the term “flap endonuclease assay” includes “INVADER” invasive cleavage assays and QuARTS assays, as described above.
  • probe oligonucleotide or “flap oligonucleotide” when used in reference to flap assay, refers to an oligonucleotide that interacts with a target nucleic acid to form a cleavage structure in the presence of an invasive oligonucleotide.
  • invasive oligonucleotide refers to an oligonucleotide that hybridizes to a target nucleic acid at a location adjacent to the region of hybridization between a probe and the target nucleic acid, wherein the 3′ end of the invasive oligonucleotide comprises a portion (e.g., a chemical moiety, or one or more nucleotides) that overlaps with the region of hybridization between the probe and target.
  • the 3′ terminal nucleotide of the invasive oligonucleotide may or may not base pair a nucleotide in the target.
  • the invasive oligonucleotide contains sequences at its 3′ end that are substantially the same as sequences located at the 5′ end of a portion of the probe oligonucleotide that anneals to the target strand.
  • overlap endonuclease refers to a class of nucleolytic enzymes, typically 5′ nucleases, that act as structure-specific endonucleases on DNA structures with a duplex containing a single stranded 5′ overhang, or flap, on one of the strands that is displaced by another strand of nucleic acid (e.g., such that there are overlapping nucleotides at the junction between the single and double-stranded DNA). FENs catalyze hydrolytic cleavage of the phosphodiester bond at the junction of single and double stranded DNA, releasing the overhang, or the flap.
  • FENs may be individual enzymes, multi-subunit enzymes, or may exist as an activity of another enzyme or protein complex (e.g., a DNA polymerase).
  • a flap endonuclease may be thermostable.
  • FEN-1 flap endonuclease from archival thermophiles organisms are typical thermostable.
  • the term “FEN-1” refers to a non-polymerase flap endonuclease from a eukaryote or archaeal organism. See, e.g., WO 02/070755, and Kaiser M. W., et al. (1999) J. Biol. Chem., 274:21387, which are incorporated by reference herein in their entireties for all purposes.
  • cleaved flap refers to a single-stranded oligonucleotide that is a cleavage product of a flap assay.
  • cassette when used in reference to a flap cleavage reaction, refers to an oligonucleotide or combination of oligonucleotides configured to generate a detectable signal in response to cleavage of a flap or probe oligonucleotide, e.g., in a primary or first cleavage structure formed in a flap cleavage assay.
  • the cassette hybridizes to a non-target cleavage product produced by cleavage of a flap oligonucleotide to form a second overlapping cleavage structure, such that the cassette can then be cleaved by the same enzyme, e.g., a FEN-1 endonuclease.
  • the cassette is a single oligonucleotide comprising a hairpin portion (i.e., a region wherein one portion of the cassette oligonucleotide hybridizes to a second portion of the same oligonucleotide under reaction conditions, to form a duplex).
  • a cassette comprises at least two oligonucleotides comprising complementary portions that can form a duplex under reaction conditions.
  • the cassette comprises a label, e.g., a fluorophore.
  • a cassette comprises labeled moieties that produce a FRET effect.
  • FRET refers to fluorescence resonance energy transfer, a process in which moieties (e.g., fluorophores) transfer energy e.g., among themselves, or, from a fluorophore to a non-fluorophore (e.g., a quencher molecule).
  • FRET involves an excited donor fluorophore transferring energy to a lower-energy acceptor fluorophore via a short-range (e.g., about 10 nm or less) dipole-dipole interaction.
  • FRET involves a loss of fluorescence energy from a donor and an increase in fluorescence in an acceptor fluorophore.
  • FRET energy can be exchanged from an excited donor fluorophore to a non-fluorescing molecule (e.g., a “dark” quenching molecule).
  • FRET is known to those of skill in the art and has been described (See, e.g., Stryer et al., 1978, Ann. Rev. Biochem., 47:819; Selvin, 1995, Methods Enzymol., 246:300; Orpana, 2004 Biomol Eng 21, 45-50; Olivier, 2005 Mutant Res 573, 103-110, each of which is incorporated herein by reference in its entirety).
  • an invasive oligonucleotide and flap oligonucleotide are hybridized to a target nucleic acid to produce a first complex having an overlap as described above.
  • An unpaired “flap” is included on the 5′ end of the flap oligonucleotide.
  • the first complex is a substrate for a flap endonuclease, e.g., a FEN-1 endonuclease, which cleaves the flap oligonucleotide to release the 5′ flap portion.
  • the released 5′ flap product serves as an invasive oligonucleotide on a FRET cassette to again create the structure recognized by the flap endonuclease, such that the FRET cassette is cleaved.
  • a detectable fluorescent signal above background fluorescence is produced.
  • real time refers to the detection or measurement of the accumulation of products or signal in the reaction while the reaction is in progress, e.g., during incubation or thermal cycling. Such detection or measurement may occur continuously, or it may occur at a plurality of discrete points during the progress of the amplification reaction, or it may be a combination. For example, in a polymerase chain reaction, detection (e.g., of fluorescence) may occur continuously during all or part of thermal cycling, or it may occur transiently, at one or more points during one or more cycles.
  • detection e.g., of fluorescence
  • real time detection of PCR or QUARTS reactions is accomplished by determining a level of fluorescence at the same point (e.g., a time point in the cycle, or temperature step in the cycle) in each of a plurality of cycles, or in every cycle.
  • Real time detection of amplification may also be referred to as detection “during” the amplification reaction.
  • the term “quantitative amplification data set” refers to the data obtained during quantitative amplification of the target sample, e.g., target DNA.
  • the quantitative amplification data set is a collection of fluorescence values obtained at during amplification, e.g., during a plurality of, or all of the thermal cycles.
  • Data for quantitative amplification is not limited to data collected at any particular point in a reaction, and fluorescence may be measured at a discrete point in each cycle or continuously throughout each cycle.
  • Ct and Cp as used herein in reference to data collected during real time PCR and PCR+INVADER assays refer to the cycle at which signal (e.g., fluorescent signal) crosses a predetermined threshold value indicative of positive signal.
  • signal e.g., fluorescent signal
  • Various methods have been used to calculate the threshold that is used as a determinant of signal verses concentration, and the value is generally expressed as either the “crossing threshold” (Ct) or the “crossing point” (Cp).
  • Ct crossing threshold
  • Cp crossing point
  • Either Cp values or Ct values may be used in embodiments of the methods presented herein for analysis of real-time signal for the determination of the percentage of variant and/or non-variant constituents in an assay or sample.
  • kits refers to any delivery system for delivering materials.
  • reaction assays such delivery systems include systems that allow for the storage, transport, or delivery of reaction reagents (e.g., oligonucleotides, enzymes, etc. in the appropriate containers) and/or supporting materials (e.g., buffers, written instructions for performing the assay etc.) from one location to another.
  • kits include one or more enclosures (e.g., boxes) containing the relevant reaction reagents and/or supporting materials.
  • fragmentous kit refers to delivery systems comprising two or more separate containers that each contains a subportion of the total kit components. The containers may be delivered to the intended recipient together or separately.
  • a first container may contain an enzyme for use in an assay, while a second container contains oligonucleotides.
  • system refers to a collection of articles for use for a particular purpose.
  • the articles comprise instructions for use, as information supplied on e.g., an article, on paper, or on recordable media (e.g., DVD, CD, flash drive, etc.).
  • instructions direct a user to an online location, e.g., a website.
  • the term “information” refers to any collection of facts or data. In reference to information stored or processed using a computer system(s), including but not limited to internets, the term refers to any data stored in any format (e.g., analog, digital, optical, etc.).
  • the term “information related to a subject” refers to facts or data pertaining to a subject (e.g., a human, plant, or animal).
  • the term “genomic information” refers to information pertaining to a genome including, but not limited to, nucleic acid sequences, genes, percentage methylation, allele frequencies, RNA expression levels, protein expression, phenotypes correlating to genotypes, etc.
  • Allele frequency information refers to facts or data pertaining to allele frequencies, including, but not limited to, allele identities, statistical correlations between the presence of an allele and a characteristic of a subject (e.g., a human subject), the presence or absence of an allele in an individual or population, the percentage likelihood of an allele being present in an individual having one or more particular characteristics, etc.
  • FIG. 1 shows schematic diagrams of marker target regions in unconverted form and bisulfite-converted form. Flap assay primers and probes for detection of bisulfate-converted target DNA are shown.
  • FIGS. 2-5 provide tables comparing Reduced Representation Bisulfite Sequencing (RRBS) results for selecting markers associated with lung carcinomas as described in Example 2, with each row showing the mean values for the indicated marker region (identified by chromosome and start and stop positions).
  • the ratio of mean methylation for each tissue type normal (Norm), adenocarcinoma (Ad), large cell carcinoma (LC), small cell carcinoma(SC), squamous cell carcinoma (SQ) and undefined cancer (UND) is compared to the mean methylation of buffy coat samples from normal subjects (WBC or BC)) is shown for each region, and genes and transcripts identified with each region are indicated.
  • RRBS Reduced Representation Bisulfite Sequencing
  • FIG. 2 provides a table comparing RRBS results for selecting markers associated with lung adenocarcinoma.
  • FIG. 3 provides a table comparing RRBS results for selecting markers associated with lung large cell carcinoma.
  • FIG. 4 provides a table comparing RRBS results for selecting markers associated with lung small cell carcinoma.
  • FIG. 5 provides a table comparing RRBS results for selecting markers associated with lung squamous cell carcinoma.
  • FIG. 6 provides a table of nucleic acid sequences of assay targets and detection oligonucleotides, with corresponding SEQ ID NOS.
  • FIG. 7 provides a graph showing a 6-marker logistic fit of data from Example 3, using markers SHOX2, SOBP, ZNF781, BTACT, CYP26C1, and DLX4.
  • the ROC curve analysis shows an area under the curve (AUC) of 0.973.
  • FIG. 8 provides a graph showing a 6-marker logistic fit of data from Example 3, using markers SHOX2, SOBP, ZNF781, CYP26C1, SUCLG2, and SKI.
  • the ROC curve analysis shows an area under the curve (AUC) of 0.97982.
  • the technology relates to selection of nucleic acid markers for use in assays for detection and quantification of DNA, e.g., methylated DNA, and use of the markers in nucleic acid detection assays.
  • the technology relates to use of methylation assays to detect lung cancer.
  • a marker is a region of 100 or fewer bases, the marker is a region of 500 or fewer bases, the marker is a region of 1000 or fewer bases, the marker is a region of 5000 or fewer bases, or, in some embodiments, the marker is one base. In some embodiments the marker is in a high CpG density promoter.
  • the technology is not limited by sample type.
  • the sample is a stool sample, a tissue sample, sputum, a blood sample (e.g., plasma, serum, whole blood), an excretion, or a urine sample.
  • the technology is not limited in the method used to determine methylation state.
  • the assaying comprises using methylation specific polymerase chain reaction, nucleic acid sequencing, mass spectrometry, methylation specific nuclease, mass-based separation, or target capture.
  • the assaying comprises use of a methylation specific oligonucleotide.
  • the technology uses massively parallel sequencing (e.g., next-generation sequencing) to determine methylation state, e.g., sequencing-by-synthesis, real-time (e.g., single-molecule) sequencing, bead emulsion sequencing, nanopore sequencing, etc.
  • an oligonucleotide comprising a sequence complementary to a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSFJ, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GR
  • Kit embodiments are provided, e.g., a kit comprising a bisulfate reagent; and a control nucleic acid comprising a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST
  • kits comprise a bisulfite reagent and an oligonucleotide as described herein. In some embodiments, kits comprise a bisulfite reagent; and a control nucleic acid comprising a sequence from such a chromosomal region and having a methylation state associated with a subject who has lung cancer.
  • compositions e.g., reaction mixtures.
  • a composition comprising a nucleic acid comprising a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28,
  • compositions comprising a nucleic acid comprising a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST8SIA_22, FLJ45983, DLX4, SHOX2, EMX
  • compositions comprising a nucleic acid comprising a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST8SIA_22, FLJ45983, DLX4, SHOX2, EMX
  • compositions comprising a nucleic acid comprising a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST8SIA_22, FLJ45983, DLX4, SHOX2, EMX
  • Additional related method embodiments are provided for screening for a neoplasm (e.g., lung carcinoma) in a sample obtained from a subject, e.g., a method comprising determining a methylation state of a marker in the sample comprising a base in a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP
  • the confidence interval is 90%, 95%, 97.5%, 98%, 99%, 99.5%, 99.9% or 99.99% and the p value is 0.1, 0.05, 0.025, 0.02, 0.01, 0.005, 0.001, or 0.0001.
  • Some embodiments of methods provide steps of reacting a nucleic acid comprising a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST8SIA_22, FLJ45983, DLX4, SHOX2,
  • Systems for screening for lung cancer in a sample obtained from a subject are provided by the technology.
  • Exemplary embodiments of systems include, e.g., a system for screening for lung cancer in a sample obtained from a subject, the system comprising an analysis component configured to determine the methylation state of a sample, a software component configured to compare the methylation state of the sample with a control sample or a reference sample methylation state recorded in a database, and an alert component configured to alert a user of a cancer-associated methylation state.
  • An alert is determined in some embodiments by a software component that receives the results from multiple assays (e.g., determining the methylation states of multiple markers, e.g., a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, B
  • Some embodiments provide a database of weighted parameters associated with each a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST8SIA_22, FLJ45983, DLX4, SHOX2, EMX1,
  • results from multiple assays are reported and in some embodiments one or more results are used to provide a score, value, or result based on a composite of one or more results from multiple assays that is indicative of a lung cancer risk in a subject.
  • a sample comprises a nucleic acid comprising a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GRIN2D, MATK, BCAT1, PRKCB_28, ST8SIA_22, FLJ45983, DLX4, SHOX2,
  • the system further comprises a component for isolating a nucleic acid, a component for collecting a sample such as a component for collecting a stool sample.
  • the system comprises nucleic acid sequences comprising a chromosomal region having an annotation selected from BARU, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR,
  • the database comprises nucleic acid sequences from subjects who do not have lung cancer.
  • nucleic acids e.g., a set of nucleic acids, each nucleic acid having a sequence comprising a chromosomal region having an annotation selected from BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4, CYP26C1, ZNF781, PTGDR, GR
  • Related system embodiments comprise a set of nucleic acids as described and a database of nucleic acid sequences associated with the set of nucleic acids. Some embodiments further comprise a bisulfate reagent. And, some embodiments further comprise a nucleic acid sequencer.
  • methods for characterizing a sample obtained from a human subject comprising a) obtaining a sample from a human subject; b) assaying a methylation state of one or more markers in the sample, wherein the marker comprises a base in a chromosomal region having an annotation selected from the following groups of markers: BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF
  • the technology is related to assessing the presence of and methylation state of one or more of the markers identified herein in a biological sample. These markers comprise one or more differentially methylated regions (DMR) as discussed herein. Methylation state is assessed in embodiments of the technology.
  • DMR differentially methylated regions
  • Methylation state is assessed in embodiments of the technology.
  • the technology provided herein is not restricted in the method by which a gene's methylation state is measured.
  • the methylation state is measured by a genome scanning method.
  • one method involves restriction landmark genomic scanning (Kawai et al. (1994) Mol. Cell. Biol. 14: 7421-7427) and another example involves methylation-sensitive arbitrarily primed PCR (Gonzalgo et al. (1997) Cancer Res.
  • changes in methylation patterns at specific CpG sites are monitored by digestion of genomic DNA with methylation-sensitive restriction enzymes followed by Southern analysis of the regions of interest (digestion-Southern method).
  • analyzing changes in methylation patterns involves a PCR-based process that involves digestion of genomic DNA with methylation-sensitive restriction enzymes prior to PCR amplification (Singer-Sam et al. (1990) Nucl. Acids Res. 18: 687).
  • MSP methylation-specific PCR
  • Such techniques use internal primers, which anneal to a PCR-generated template and terminate immediately 5′ of the single nucleotide to be assayed.
  • Methods using a “quantitative Ms-SNuPE assay” as described in U.S. Pat. No. 7,037,650 are used in some embodiments.
  • the methylation state is often expressed as the fraction or percentage of individual strands of DNA that is methylated at a particular site (e.g., at a single nucleotide, at a particular region or locus, at a longer sequence of interest, e.g., up to a ⁇ 100-bp, 200-bp, 500-bp, 1000-bp subsequence of a DNA or longer) relative to the total population of DNA in the sample comprising that particular site.
  • the amount of the unmethylated nucleic acid is determined by PCR using calibrators.
  • a known amount of DNA is bisulfite treated and the resulting methylation-specific sequence is determined using either a real-time PCR or other exponential amplification, e.g., a QuARTS assay (e.g., as provided by U.S. Pat. Nos. 8,361,720; 8,715,937; 8,916,344; and 9,212,392).
  • a QuARTS assay e.g., as provided by U.S. Pat. Nos. 8,361,720; 8,715,937; 8,916,344; and 9,212,392.
  • methods comprise generating a standard curve for the unmethylated target by using external standards.
  • the standard curve is constructed from at least two points and relates the real-time Ct value for unmethylated DNA to known quantitative standards.
  • a second standard curve for the methylated target is constructed from at least two points and external standards. This second standard curve relates the Ct for methylated DNA to known quantitative standards.
  • the test sample Ct values are determined for the methylated and unmethylated populations and the genomic equivalents of DNA are calculated from the standard curves produced by the first two steps.
  • the percentage of methylation at the site of interest is calculated from the amount of methylated DNAs relative to the total amount of DNAs in the population, e.g., (number of methylated DNAs)/(the number of methylated DNAs+number of unmethylated DNAs) ⁇ 100.
  • compositions and kits for practicing the methods.
  • reagents e.g., primers, probes
  • sets e.g., sets of primers pairs for amplifying a plurality of markers.
  • Additional reagents for conducting a detection assay may also be provided (e.g., enzymes, buffers, positive and negative controls for conducting QuARTS, PCR, sequencing, bisulfite, or other assays).
  • the kits containing one or more reagent necessary, sufficient, or useful for conducting a method are provided.
  • reactions mixtures containing the reagents.
  • master mix reagent sets containing a plurality of reagents that may be added to each other and/or to a test sample to complete a reaction mixture.
  • Some embodiments comprise isolation of nucleic acids as described in U.S. patent application Ser. No. 13/470,251 (“Isolation of Nucleic Acids”), incorporated herein by reference in its entirety.
  • Genomic DNA may be isolated by any means, including the use of commercially available kits. Briefly, wherein the DNA of interest is encapsulated by a cellular membrane the biological sample must be disrupted and lysed by enzymatic, chemical or mechanical means. The DNA solution may then be cleared of proteins and other contaminants, e.g., by digestion with proteinase K. The genomic DNA is then recovered from the solution. This may be carried out by means of a variety of methods including salting out, organic extraction, or binding of the DNA to a solid phase support. The choice of method will be affected by several factors including time, expense, and required quantity of DNA.
  • neoplastic matter or pre-neoplastic matter are suitable for use in the present method, e.g., cell lines, histological slides, biopsies, paraffin-embedded tissue, body fluids, stool, colonic effluent, urine, blood plasma, blood serum, whole blood, isolated blood cells, cells isolated from the blood, and combinations thereof.
  • a DNA is isolated from a stool sample or from blood or from a plasma sample using direct gene capture, e.g., as detailed in U.S. Pat. Appl. Ser. No. 61/485,386 or by a related method.
  • the technology relates to the analysis of any sample that may be associated with lung cancer, or that may be examined to establish the absence of lung cancer.
  • the sample comprises a tissue and/or biological fluid obtained from a patient.
  • the sample comprises a secretion.
  • the sample comprises sputum, blood, serum, plasma, gastric secretions, lung tissue samples, lung cells or lung DNA recovered from stool.
  • the subject is human.
  • Such samples can be obtained by any number of means known in the art, such as will be apparent to the skilled person.
  • Candidate methylated DNA markers were identified by unbiased whole methylome sequencing of selected lung cancer case and lung control tissues. The top marker candidates were further evaluated in 255 independent patients with 119 controls, of which 37 were from benign nodules, and 136 cases inclusive of all lung cancer subtypes. DNA extracted from patient tissue samples was bisulfite treated and then candidate markers and ⁇ -actin (ACTB) as a normalizing gene were assayed by Quantitative Allele-Specific Real-time Target and Signal amplification (QUARTS amplification). QuARTS assay chemistry yields high discrimination for methylated marker selection and screening.
  • ACTB ⁇ -actin
  • AUCs areas under the curve
  • a combined panel of 8 methylation markers SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX 12.526, HOXB2, and EMX1 yielded a sensitivity of 98.5% across all subtypes of lung cancer.
  • 8 markers panel benign lung nodules yielded no false positives.
  • the markers described herein find use in a variety of methylation detection assays.
  • the most frequently used method for analyzing a nucleic acid for the presence of 5-methylcytosine is based upon the bisulfite method described by Frommer, et al. for the detection of 5-methylcytosines in DNA (Frommer et al. (1992) Proc. Natl. Acad. Sci. USA 89: 1827-31 explicitly incorporated herein by reference in its entirety for all purposes) or variations thereof.
  • the bisulfite method of mapping 5-methylcytosines is based on the observation that cytosine, but not 5-methylcytosine, reacts with hydrogen sulfite ion (also known as bisulfite).
  • the reaction is usually performed according to the following steps: first, cytosine reacts with hydrogen sulfite to form a sulfonated cytosine. Next, spontaneous deamination of the sulfonated reaction intermediate results in a sulfonated uracil. Finally, the sulfonated uracil is desulfonated under alkaline conditions to form uracil. Detection is possible because uracil base pairs with adenine (thus behaving like thymine), whereas 5-methylcytosine base pairs with guanine (thus behaving like cytosine).
  • methylated cytosines from non-methylated cytosines possible by, e.g., bisulfite genomic sequencing (Grigg G, & Clark S, Bioessays (1994) 16: 431-36; Grigg G, DNA Seq. (1996) 6: 189-98),methylation-specific PCR (MSP) as is disclosed, e.g., in U.S. Pat. No. 5,786,146, or using an assay comprising sequence-specific probe cleavage, e.g., a QuARTS flap endonuclease assay (see, e.g., Zou et al.
  • MSP methylation-specific PCR
  • Some conventional technologies are related to methods comprising enclosing the DNA to be analyzed in an agarose matrix, thereby preventing the diffusion and renaturation of the DNA (bisulfite only reacts with single-stranded DNA), and replacing precipitation and purification steps with a fast dialysis (Olek A, et al. (1996) “A modified and improved method for bisulfite based cytosine methylation analysis” Nucleic Acids Res. 24: 5064-6). It is thus possible to analyze individual cells for methylation status, illustrating the utility and sensitivity of the method.
  • An overview of conventional methods for detecting 5-methylcytosine is provided by Rein, T., et al. (1998) Nucleic Acids Res. 26: 2255.
  • the bisulfite technique typically involves amplifying short, specific fragments of a known nucleic acid subsequent to a bisulfite treatment, then either assaying the product by sequencing (Olek & Walter (1997) Nat. Genet. 17: 275-6) or a primer extension reaction (Gonzalgo & Jones (1997) Nucleic Acids Res. 25: 2529-31; WO 95/00669; U.S. Pat. No. 6,251,594) to analyze individual cytosine positions. Some methods use enzymatic digestion (Xiong & Laird (1997) Nucleic Acids Res. 25: 2532-4). Detection by hybridization has also been described in the art (Olek et al., WO 99/28498).
  • methylation assay procedures can be used in conjunction with bisulfite treatment according to the present technology. These assays allow for determination of the methylation state of one or a plurality of CpG dinucleotides (e.g., CpG islands) within a nucleic acid sequence. Such assays involve, among other techniques, sequencing of bisulfite-treated nucleic acid, PCR (for sequence-specific amplification), Southern blot analysis, and use of methylation-sensitive restriction enzymes.
  • genomic sequencing has been simplified for analysis of methylation patterns and 5-methylcytosine distributions by using bisulfite treatment (Frommer et al. (1992) Proc. Natl. Acad. Sci. USA 89: 1827-1831).
  • restriction enzyme digestion of PCR products amplified from bisulfite-converted DNA finds use in assessing methylation state, e.g., as described by Sadri & Hornsby (1997) Nucl. Acids Res. 24: 5058-5059 or as embodied in the method known as COBRA (Combined Bisulfite Restriction Analysis) (Xiong & Laird (1997) Nucleic Acids Res. 25: 2532-2534).
  • COBRATM analysis is a quantitative methylation assay useful for determining DNA methylation levels at specific loci in small amounts of genomic DNA (Xiong & Laird, Nucleic Acids Res. 25:2532-2534, 1997). Briefly, restriction enzyme digestion is used to reveal methylation-dependent sequence differences in PCR products of sodium bisulfite-treated DNA. Methylation-dependent sequence differences are first introduced into the genomic DNA by standard bisulfite treatment according to the procedure described by Frommer et al. (Proc. Natl. Acad. Sci. USA 89:1827-1831, 1992).
  • PCR amplification of the bisulfite converted DNA is then performed using primers specific for the CpG islands of interest, followed by restriction endonuclease digestion, gel electrophoresis, and detection using specific, labeled hybridization probes.
  • Methylation levels in the original DNA sample are represented by the relative amounts of digested and undigested PCR product in a linearly quantitative fashion across a wide spectrum of DNA methylation levels.
  • this technique can be reliably applied to DNA obtained from microdissected paraffin-embedded tissue samples.
  • Typical reagents for COBRATM analysis may include, but are not limited to: PCR primers for specific loci (e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, etc.); restriction enzyme and appropriate buffer; gene-hybridization oligonucleotide; control hybridization oligonucleotide; kinase labeling kit for oligonucleotide probe; and labeled nucleotides.
  • bisulfite conversion reagents may include: DNA denaturation buffer; sulfonation buffer; DNA recovery reagents or kits (e.g., precipitation, ultrafiltration, affinity column); desulfonation buffer; and DNA recovery components.
  • Assays such as “MethyLightTM” (a fluorescence-based real-time PCR technique) (Eads et al., Cancer Res. 59:2302-2306, 1999), Ms-SNuPETM (Methylation-sensitive Single Nucleotide Primer Extension) reactions (Gonzalgo & Jones, Nucleic Acids Res. 25:2529-2531, 1997), methylation-specific PCR (“MSP”; Herman et al., Proc. Natl. Acad. Sci. USA 93:9821-9826, 1996; U.S. Pat. No. 5,786,146), and methylated CpG island amplification (“MCA”; Toyota et al., Cancer Res. 59:2307-12, 1999) are used alone or in combination with one or more of these methods.
  • MSP methylation-specific PCR
  • MCA methylated CpG island amplification
  • the “HeavyMethylTM” assay, technique is a quantitative method for assessing methylation differences based on methylation-specific amplification of bisulfite-treated DNA.
  • Methylation-specific blocking probes (“blockers”) covering CpG positions between, or covered by, the amplification primers enable methylation-specific selective amplification of a nucleic acid sample.
  • HeavyMethylTM MethyLightTM assay refers to a HeavyMethylTM MethyLightTM assay, which is a variation of the MethyLightTM assay, wherein the MethyLightTM assay is combined with methylation specific blocking probes covering CpG positions between the amplification primers.
  • the HeavyMethylTM assay may also be used in combination with methylation specific amplification primers.
  • Typical reagents for HeavyMethylTM analysis may include, but are not limited to: PCR primers for specific loci (e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, or bisulfite treated DNA sequence or CpG island, etc.); blocking oligonucleotides; optimized PCR buffers and deoxynucleotides; and Taq polymerase.
  • specific loci e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, or bisulfite treated DNA sequence or CpG island, etc.
  • blocking oligonucleotides e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, or bisulfite treated DNA sequence or CpG island, etc.
  • blocking oligonucleotides e.g., specific genes, markers, regions of genes, regions of markers,
  • MSP methylation-specific PCR
  • DNA is modified by sodium bisulfite, which converts unmethylated, but not methylated cytosines, to uracil, and the products are subsequently amplified with primers specific for methylated versus unmethylated DNA.
  • MSP requires only small quantities of DNA, is sensitive to 0.1% methylated alleles of a given CpG island locus, and can be performed on DNA extracted from paraffin-embedded samples.
  • Typical reagents e.g., as might be found in a typical MSP-based kit
  • MSP analysis may include, but are not limited to: methylated and unmethylated PCR primers for specific loci (e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, etc.); optimized PCR buffers and deoxynucleotides, and specific probes.
  • the MethyLightTM assay is a high-throughput quantitative methylation assay that utilizes fluorescence-based real-time PCR (e.g., TaqMan0) that requires no further manipulations after the PCR step (Eads et al., Cancer Res. 59:2302-2306, 1999). Briefly, the MethyLightTM process begins with a mixed sample of genomic DNA that is converted, in a sodium bisulfite reaction, to a mixed pool of methylation-dependent sequence differences according to standard procedures (the bisulfite process converts unmethylated cytosine residues to uracil).
  • fluorescence-based real-time PCR e.g., TaqMan0
  • the MethyLightTM process begins with a mixed sample of genomic DNA that is converted, in a sodium bisulfite reaction, to a mixed pool of methylation-dependent sequence differences according to standard procedures (the bisulfite process converts unmethylated cytosine residues to uracil).
  • Fluorescence-based PCR is then performed in a “biased” reaction, e.g., with PCR primers that overlap known CpG dinucleotides. Sequence discrimination occurs both at the level of the amplification process and at the level of the fluorescence detection process.
  • the MethyLightTM assay is used as a quantitative test for methylation patterns in a nucleic acid, e.g., a genomic DNA sample, wherein sequence discrimination occurs at the level of probe hybridization.
  • a quantitative version the PCR reaction provides for a methylation specific amplification in the presence of a fluorescent probe that overlaps a particular putative methylation site.
  • An unbiased control for the amount of input DNA is provided by a reaction in which neither the primers, nor the probe, overlie any CpG dinucleotides.
  • a qualitative test for genomic methylation is achieved by probing the biased PCR pool with either control oligonucleotides that do not cover known methylation sites (e.g., a fluorescence-based version of the HeavyMethylTM and MSP techniques) or with oligonucleotides covering potential methylation sites.
  • the MethyLightTM process is used with any suitable probe (e.g. a “TaqMan®” probe, a Lightcycler® probe, etc.)
  • a “TaqMan®” probe e.g. a “TaqMan®” probe, a Lightcycler® probe, etc.
  • double-stranded genomic DNA is treated with sodium bisulfite and subjected to one of two sets of PCR reactions using TaqMan® probes, e.g., with MSP primers and/or HeavyMethyl blocker oligonucleotides and a TaqMan® probe.
  • the TaqMan® probe is dual-labeled with fluorescent “reporter” and “quencher” molecules and is designed to be specific for a relatively high GC content region so that it melts at about a 10° C. higher temperature in the PCR cycle than the forward or reverse primers.
  • TaqMan® probe This allows the TaqMan® probe to remain fully hybridized during the PCR annealing/extension step. As the Taq polymerase enzymatically synthesizes a new strand during PCR, it will eventually reach the annealed TaqMan® probe. The Taq polymerase 5′ to 3′ endonuclease activity will then displace the TaqMan® probe by digesting it to release the fluorescent reporter molecule for quantitative detection of its now unquenched signal using a real-time fluorescent detection system.
  • Typical reagents for MethyLightTM analysis may include, but are not limited to: PCR primers for specific loci (e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, etc.); TaqMan® or Lightcycler® probes; optimized PCR buffers and deoxynucleotides; and Taq polymerase.
  • specific loci e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, etc.
  • TaqMan® or Lightcycler® probes e.g., optimized PCR buffers and deoxynucleotides
  • Taq polymerase e.g., as might be found in a typical MethyLightTM-based kit
  • the QMTM (quantitative methylation) assay is an alternative quantitative test for methylation patterns in genomic DNA samples, wherein sequence discrimination occurs at the level of probe hybridization.
  • the PCR reaction provides for unbiased amplification in the presence of a fluorescent probe that overlaps a particular putative methylation site.
  • An unbiased control for the amount of input DNA is provided by a reaction in which neither the primers, nor the probe, overlie any CpG dinucleotides.
  • a qualitative test for genomic methylation is achieved by probing the biased PCR pool with either control oligonucleotides that do not cover known methylation sites (a fluorescence-based version of the HeavyMethylTM and MSP techniques) or with oligonucleotides covering potential methylation sites.
  • the QMTM process can be used with any suitable probe, e.g., “TaqMan®” probes, Lightcycler® probes, in the amplification process.
  • any suitable probe e.g., “TaqMan®” probes, Lightcycler® probes
  • double-stranded genomic DNA is treated with sodium bisulfite and subjected to unbiased primers and the TaqMan® probe.
  • the TaqMan® probe is dual-labeled with fluorescent “reporter” and “quencher” molecules, and is designed to be specific for a relatively high GC content region so that it melts out at about a 10° C. higher temperature in the PCR cycle than the forward or reverse primers. This allows the TaqMan® probe to remain fully hybridized during the PCR annealing/extension step.
  • Taq polymerase As the Taq polymerase enzymatically synthesizes a new strand during PCR, it will eventually reach the annealed TaqMan® probe. The Taq polymerase 5′ to 3′ endonuclease activity will then displace the TaqMan® probe by digesting it to release the fluorescent reporter molecule for quantitative detection of its now unquenched signal using a real-time fluorescent detection system.
  • Typical reagents for QMTM analysis may include, but are not limited to: PCR primers for specific loci (e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, etc.); TaqMan® or Lightcycler® probes; optimized PCR buffers and deoxynucleotides; and Taq polymerase.
  • specific loci e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, etc.
  • TaqMan® or Lightcycler® probes e.g., optimized PCR buffers and deoxynucleotides
  • Taq polymerase e.g., as might be found in a typical QMTM-based kit
  • the Ms-SNuPETM technique is a quantitative method for assessing methylation differences at specific CpG sites based on bisulfite treatment of DNA, followed by single-nucleotide primer extension (Gonzalgo & Jones, Nucleic Acids Res. 25:2529-2531, 1997). Briefly, genomic DNA is reacted with sodium bisulfite to convert unmethylated cytosine to uracil while leaving 5-methylcytosine unchanged. Amplification of the desired target sequence is then performed using PCR primers specific for bisulfite-converted DNA, and the resulting product is isolated and used as a template for methylation analysis at the CpG site of interest. Small amounts of DNA can be analyzed (e.g., microdissected pathology sections) and it avoids utilization of restriction enzymes for determining the methylation status at CpG sites.
  • Typical reagents for Ms-SNuPETM analysis may include, but are not limited to: PCR primers for specific loci (e.g., specific genes, markers, regions of genes, regions of markers, bisulfite treated DNA sequence, CpG island, etc.); optimized PCR buffers and deoxynucleotides; gel extraction kit; positive control primers; Ms-SNuPETM primers for specific loci; reaction buffer (for the Ms-SNuPE reaction); and labeled nucleotides.
  • bisulfite conversion reagents may include: DNA denaturation buffer; sulfonation buffer; DNA recovery reagents or kit (e.g., precipitation, ultrafiltration, affinity column); desulfonation buffer; and DNA recovery components.
  • RRBS Reduced Representation Bisulfite Sequencing
  • every fragment produced by the restriction enzyme digestion contains DNA methylation information for at least one CpG dinucleotide.
  • RRBS enriches the sample for promoters, CpG islands, and other genomic features with a high frequency of restriction enzyme cut sites in these regions and thus provides an assay to assess the methylation state of one or more genomic loci.
  • a typical protocol for RRBS comprises the steps of digesting a nucleic acid sample with a restriction enzyme such as Mspl, filling in overhangs and A-tailing, ligating adaptors, bisulfite conversion, and PCR.
  • a restriction enzyme such as Mspl
  • a quantitative allele-specific real-time target and signal amplification (QUARTS) assay is used to evaluate methylation state.
  • Three reactions sequentially occur in each QuARTS assay, including amplification (reaction 1) and target probe cleavage (reaction 2) in the primary reaction; and FRET cleavage and fluorescent signal generation (reaction 3) in the secondary reaction.
  • amplification reaction 1
  • target probe cleavage reaction 2
  • FRET cleavage and fluorescent signal generation reaction 3
  • target nucleic acid is amplified with specific primers
  • a specific detection probe with a flap sequence loosely binds to the amplicon.
  • the presence of the specific invasive oligonucleotide at the target binding site causes a 5′ nuclease, e.g., a FEN-1 endonuclease, to release the flap sequence by cutting between the detection probe and the flap sequence.
  • the flap sequence is complementary to a non-hairpin portion of a corresponding FRET cassette. Accordingly, the flap sequence functions as an invasive oligonucleotide on the FRET cassette and effects a cleavage between the FRET cassette fluorophore and a quencher, which produces a fluorescent signal.
  • the cleavage reaction can cut multiple probes per target and thus release multiple fluorophore per flap, providing exponential signal amplification.
  • QuARTS can detect multiple targets in a single reaction well by using FRET cassettes with different dyes. See, e.g., in Zou et al. (2010) “Sensitive quantification of methylated markers with a novel methylation specific technology” Clin Chem 56: A199), and U.S. Pat. Nos. 8,361,720; 8,715,937; 8,916,344; and 9,212,392, each of which is incorporated herein by reference for all purposes.
  • the bisulfite-treated DNA is purified prior to the quantification. This may be conducted by any means known in the art, such as but not limited to ultrafiltration, e.g., by means of MicroconTM columns (manufactured by MilliporeTM). The purification is carried out according to a modified manufacturer's protocol (see, e.g., PCT/EP2004/011715, which is incorporated by reference in its entirety).
  • the bisulfate treated DNA is bound to a solid support, e.g., a magnetic bead, and desulfonation and washing occurs while the DNA is bound to the support. Examples of such embodiments are provided, e.g., in WO 2013/116375 and U.S. Pat.
  • support-bound DNA is ready for a methylation assay immediately after desulfonation and washing on the support.
  • the desulfonated DNA is eluted from the support prior to assay.
  • fragments of the treated DNA are amplified using sets of primer oligonucleotides according to the present invention (e.g., see FIG. 1 ) and an amplification enzyme.
  • the amplification of several DNA segments can be carried out simultaneously in one and the same reaction vessel.
  • the amplification is carried out using a polymerase chain reaction (PCR).
  • the markers described herein find use in QUARTS assays performed on stool samples.
  • methods for producing DNA samples and, in particular, to methods for producing DNA samples that comprise highly purified, low-abundance nucleic acids in a small volume (e.g., less than 100, less than 60 microliters) and that are substantially and/or effectively free of substances that inhibit assays used to test the DNA samples e.g., PCR, INVADER, QuARTS assays, etc.
  • Such DNA samples find use in diagnostic assays that qualitatively detect the presence of, or quantitatively measure the activity, expression, or amount of, a gene, a gene variant (e.g., an allele), or a gene modification (e.g., methylation) present in a sample taken from a patient.
  • a gene e.g., an allele
  • a gene modification e.g., methylation
  • some cancers are correlated with the presence of particular mutant alleles or particular methylation states, and thus detecting and/or quantifying such mutant alleles or methylation states has predictive value in the diagnosis and treatment of cancer.
  • the sample comprises blood, serum, plasma, or saliva.
  • the subject is human.
  • Such samples can be obtained by any number of means known in the art, such as will be apparent to the skilled person.
  • Cell free or substantially cell free samples can be obtained by subjecting the sample to various techniques known to those of skill in the art which include, but are not limited to, centrifugation and filtration. Although it is generally preferred that no invasive techniques are used to obtain the sample, it still may be preferable to obtain samples such as tissue homogenates, tissue sections, and biopsy specimens. The technology is not limited in the methods used to prepare the samples and provide a nucleic acid for testing.
  • a DNA is isolated from a stool sample or from blood or from a plasma sample using direct gene capture, e.g., as detailed in U.S. Pat. Nos. 8,808,990 and 9,169,511, and in WO 2012/155072, or by a related method.
  • markers can be carried out separately or simultaneously with additional markers within one test sample. For example, several markers can be combined into one test for efficient processing of multiple samples and for potentially providing greater diagnostic and/or prognostic accuracy.
  • one skilled in the art would recognize the value of testing multiple samples (for example, at successive time points) from the same subject.
  • Such testing of serial samples can allow the identification of changes in marker methylation states over time. Changes in methylation state, as well as the absence of change in methylation state, can provide useful information about the disease status that includes, but is not limited to, identifying the approximate time from onset of the event, the presence and amount of salvageable tissue, the appropriateness of drug therapies, the effectiveness of various therapies, and identification of the subject's outcome, including risk of future events.
  • biomarkers can be carried out in a variety of physical formats.
  • the use of microtiter plates or automation can be used to facilitate the processing of large numbers of test samples.
  • single sample formats could be developed to facilitate immediate treatment and diagnosis in a timely fashion, for example, in ambulatory transport or emergency room settings.
  • kits comprise embodiments of the compositions, devices, apparatuses, etc. described herein, and instructions for use of the kit.
  • Such instructions describe appropriate methods for preparing an analyte from a sample, e.g., for collecting a sample and preparing a nucleic acid from the sample.
  • Individual components of the kit are packaged in appropriate containers and packaging (e.g., vials, boxes, blister packs, ampules, jars, bottles, tubes, and the like) and the components are packaged together in an appropriate container (e.g., a box or boxes) for convenient storage, shipping, and/or use by the user of the kit.
  • liquid components may be provided in a lyophilized form to be reconstituted by the user.
  • Kits may include a control or reference for assessing, validating, and/or assuring the performance of the kit.
  • a kit for assaying the amount of a nucleic acid present in a sample may include a control comprising a known concentration of the same or another nucleic acid for comparison and, in some embodiments, a detection reagent (e.g., a primer) specific for the control nucleic acid.
  • the kits are appropriate for use in a clinical setting and, in some embodiments, for use in a user's home.
  • the components of a kit in some embodiments, provide the functionalities of a system for preparing a nucleic acid solution from a sample. In some embodiments, certain components of the system are provided by the user.
  • diagnostic assays identify the presence of a disease or condition in an individual.
  • the disease is cancer (e.g., lung cancer).
  • markers whose aberrant methylation is associated with a lung cancer e.g., one or more markers selected from the markers listed in Table 1, or preferably one or more of BARX1, LOC100129726, SPOCK2, TSC22D4, MAX.chr8.124, RASSF1, ZNF671, ST8SIA1, NKX6_2, FAM59B, DIDO1, MAX_Chr1.110, AGRN, SOBP, MAX_chr10.226, ZMIZ1, MAX_chr8.145, MAX_chr10.225, PRDM14, ANGPT1, MAX.chr16.50, PTGDR_9, ANKRD13B, DOCK2, MAX_chr19.163, ZNF132, MAX chr19.372, HOXA9, TRH, SP9, DMRTA2, ARHGEF4,
  • an assay further comprises detection of a reference gene (e.g., ⁇ -actin, ZDHHC1, B3GALT6.
  • a reference gene e.g., ⁇ -actin, ZDHHC1, B3GALT6. See, e.g., U.S. patent application Ser. No. 14/966,617, filed Dec. 11, 2015, and U.S. Pat. Appl. No. 62/364,082, filed Jul. 19, 2016, each of which is incorporated herein by reference for all purposes).
  • the technology finds application in treating a patient (e.g., a patient with lung cancer, with early stage lung cancer, or who may develop lung cancer), the method comprising determining the methylation state of one or more markers as provided herein and administering a treatment to the patient based on the results of determining the methylation state.
  • the treatment may be administration of a pharmaceutical compound, a vaccine, performing a surgery, imaging the patient, performing another test.
  • said use is in a method of clinical screening, a method of prognosis assessment, a method of monitoring the results of therapy, a method to identify patients most likely to respond to a particular therapeutic treatment, a method of imaging a patient or subject, and a method for drug screening and development.
  • the technology finds application in methods for diagnosing lung cancer in a subject.
  • diagnosis and “diagnosis” as used herein refer to methods by which the skilled artisan can estimate and even determine whether or not a subject is suffering from a given disease or condition or may develop a given disease or condition in the future.
  • the skilled artisan often makes a diagnosis on the basis of one or more diagnostic indicators, such as for example a biomarker, the methylation state of which is indicative of the presence, severity, or absence of the condition.
  • clinical cancer prognosis relates to determining the aggressiveness of the cancer and the likelihood of tumor recurrence to plan the most effective therapy. If a more accurate prognosis can be made or even a potential risk for developing the cancer can be assessed, appropriate therapy, and in some instances less severe therapy for the patient can be chosen. Assessment (e.g., determining methylation state) of cancer biomarkers is useful to separate subjects with good prognosis and/or low risk of developing cancer who will need no therapy or limited therapy from those more likely to develop cancer or suffer a recurrence of cancer who might benefit from more intensive treatments.
  • “making a diagnosis” or “diagnosing”, as used herein, is further inclusive of making determining a risk of developing cancer or determining a prognosis, which can provide for predicting a clinical outcome (with or without medical treatment), selecting an appropriate treatment (or whether treatment would be effective), or monitoring a current treatment and potentially changing the treatment, based on the measure of the diagnostic biomarkers disclosed herein.
  • multiple determinations of the biomarkers over time can be made to facilitate diagnosis and/or prognosis.
  • a temporal change in the biomarker can be used to predict a clinical outcome, monitor the progression of lung cancer, and/or monitor the efficacy of appropriate therapies directed against the cancer.
  • the technology further finds application in methods for determining whether to initiate or continue prophylaxis or treatment of a cancer in a subject.
  • the method comprises providing a series of biological samples over a time period from the subject; analyzing the series of biological samples to determine a methylation state of at least one biomarker disclosed herein in each of the biological samples; and comparing any measurable change in the methylation states of one or more of the biomarkers in each of the biological samples. Any changes in the methylation states of biomarkers over the time period can be used to predict risk of developing cancer, predict clinical outcome, determine whether to initiate or continue the prophylaxis or therapy of the cancer, and whether a current therapy is effectively treating the cancer.
  • a first time point can be selected prior to initiation of a treatment and a second time point can be selected at some time after initiation of the treatment.
  • Methylation states can be measured in each of the samples taken from different time points and qualitative and/or quantitative differences noted.
  • a change in the methylation states of the biomarker levels from the different samples can be correlated with risk for developing lung, prognosis, determining treatment efficacy, and/or progression of the cancer in the subject.
  • the methods and compositions of the invention are for treatment or diagnosis of disease at an early stage, for example, before symptoms of the disease appear. In some embodiments, the methods and compositions of the invention are for treatment or diagnosis of disease at a clinical stage.
  • a diagnostic marker can be determined at an initial time, and again at a second time.
  • an increase in the marker from the initial time to the second time can be diagnostic of a particular type or severity of cancer, or a given prognosis.
  • a decrease in the marker from the initial time to the second time can be indicative of a particular type or severity of cancer, or a given prognosis.
  • the degree of change of one or more markers can be related to the severity of the cancer and future adverse events.
  • comparative measurements can be made of the same biomarker at multiple time points, one can also measure a given biomarker at one time point, and a second biomarker at a second time point, and a comparison of these markers can provide diagnostic information.
  • the phrase “determining the prognosis” refers to methods by which the skilled artisan can predict the course or outcome of a condition in a subject.
  • the term “prognosis” does not refer to the ability to predict the course or outcome of a condition with 100% accuracy, or even that a given course or outcome is predictably more or less likely to occur based on the methylation state of a biomarker.
  • the term “prognosis” refers to an increased probability that a certain course or outcome will occur; that is, that a course or outcome is more likely to occur in a subject exhibiting a given condition, when compared to those individuals not exhibiting the condition. For example, in individuals not exhibiting the condition, the chance of a given outcome (e.g., suffering from lung cancer) may be very low.
  • a statistical analysis associates a prognostic indicator with a predisposition to an adverse outcome. For example, in some embodiments, a methylation state different from that in a normal control sample obtained from a patient who does not have a cancer can signal that a subject is more likely to suffer from a cancer than subjects with a level that is more similar to the methylation state in the control sample, as determined by a level of statistical significance. Additionally, a change in methylation state from a baseline (e.g., “normal”) level can be reflective of subject prognosis, and the degree of change in methylation state can be related to the severity of adverse events.
  • a baseline e.g., “normal”
  • Statistical significance is often determined by comparing two or more populations and determining a confidence interval and/or ap value. See, e.g., Dowdy and Wearden, Statistics for Research, John Wiley & Sons, New York, 1983, incorporated herein by reference in its entirety.
  • Exemplary confidence intervals of the present subject matter are 90%, 95%, 97.5%, 98%, 99%, 99.5%, 99.9% and 99.99%, while exemplary p values are 0.1, 0.05, 0.025, 0.02, 0.01, 0.005, 0.001, and 0.0001.
  • a threshold degree of change in the methylation state of a prognostic or diagnostic biomarker disclosed herein can be established, and the degree of change in the methylation state of the biomarker in a biological sample is simply compared to the threshold degree of change in the methylation state.
  • a preferred threshold change in the methylation state for biomarkers provided herein is about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 50%, about 75%, about 100%, and about 150%.
  • a “nomogram” can be established, by which a methylation state of a prognostic or diagnostic indicator (biomarker or combination of biomarkers) is directly related to an associated disposition towards a given outcome. The skilled artisan is acquainted with the use of such nomograms to relate two numeric values with the understanding that the uncertainty in this measurement is the same as the uncertainty in the marker concentration because individual sample measurements are referenced, not population averages.
  • a control sample is analyzed concurrently with the biological sample, such that the results obtained from the biological sample can be compared to the results obtained from the control sample.
  • standard curves can be provided, with which assay results for the biological sample may be compared. Such standard curves present methylation states of a biomarker as a function of assay units, e.g., fluorescent signal intensity, if a fluorescent label is used. Using samples taken from multiple donors, standard curves can be provided for control methylation states of the one or more biomarkers in normal tissue, as well as for “at-risk” levels of the one or more biomarkers in tissue taken from donors with lung cancer.
  • markers can be carried out separately or simultaneously with additional markers within one test sample. For example, several markers can be combined into one test for efficient processing of a multiple of samples and for potentially providing greater diagnostic and/or prognostic accuracy.
  • one skilled in the art would recognize the value of testing multiple samples (for example, at successive time points) from the same subject.
  • Such testing of serial samples can allow the identification of changes in marker methylation states over time. Changes in methylation state, as well as the absence of change in methylation state, can provide useful information about the disease status that includes, but is not limited to, identifying the approximate time from onset of the event, the presence and amount of salvageable tissue, the appropriateness of drug therapies, the effectiveness of various therapies, and identification of the subject's outcome, including risk of future events.
  • biomarkers can be carried out in a variety of physical formats.
  • the use of microtiter plates or automation can be used to facilitate the processing of large numbers of test samples.
  • single sample formats could be developed to facilitate immediate treatment and diagnosis in a timely fashion, for example, in ambulatory transport or emergency room settings.
  • the subject is diagnosed as having lung cancer if, when compared to a control methylation state, there is a measurable difference in the methylation state of at least one biomarker in the sample.
  • the subject can be identified as not having lung cancer, not being at risk for the cancer, or as having a low risk of the cancer.
  • subjects having lung cancer or risk thereof can be differentiated from subjects having low to substantially no cancer or risk thereof.
  • those subjects having a risk of developing lung cancer can be placed on a more intensive and/or regular screening schedule.
  • those subjects having low to substantially no risk may avoid being subjected to screening procedures, until such time as a future screening, for example, a screening conducted in accordance with the present technology, indicates that a risk of lung cancer has appeared in those subjects.
  • detecting a change in methylation state of the one or more biomarkers can be a qualitative determination or it can be a quantitative determination.
  • the step of diagnosing a subject as having, or at risk of developing, lung cancer indicates that certain threshold measurements are made, e.g., the methylation state of the one or more biomarkers in the biological sample varies from a predetermined control methylation state.
  • the control methylation state is any detectable methylation state of the biomarker.
  • the predetermined methylation state is the methylation state in the control sample.
  • the predetermined methylation state is based upon and/or identified by a standard curve. In other embodiments of the method, the predetermined methylation state is a specifically state or range of state. As such, the predetermined methylation state can be chosen, within acceptable limits that will be apparent to those skilled in the art, based in part on the embodiment of the method being practiced and the desired specificity, etc.
  • a sample from a subject having or suspected of having lung cancer is screened using one or more methylation markers and suitable assay methods that provide data that differentiate between different types of lung cancer, e.g., non-small cell (adenocarcinoma, large cell carcinoma, squamous cell carcinoma) and small cell carcinomas.
  • suitable assay methods that provide data that differentiate between different types of lung cancer, e.g., non-small cell (adenocarcinoma, large cell carcinoma, squamous cell carcinoma) and small cell carcinomas.
  • marker ref # AC27 FIG. 2 ; PLEC
  • marker ref # AC23 FIG.
  • marker ref # LC2 ( FIG. 3 ; DOCK2)), which is more highly methylated in large cell carcinomas than in any other sample type
  • marker ref # SC221 ( FIG. 4 ; ST8SIA4), which is more highly methylated in small cell carcinomas than in any other sample type
  • marker ref # SQ36 ( FIG. 5 , DOK1), which is more highly methylated in squamous cell carcinoma than in than in any other sample type.
  • Methylation markers selected as described herein may be used alone or in combination (e.g., in panels) such that analysis of a sample from a subject reveals the presence of a lung neoplasm and also provides sufficient information to distinguish between lung cancer type, e.g., small cell carcinoma vs. non-small cell carcinoma.
  • a marker or combination of markers further provide data sufficient to distinguish between adenomcarcinomas, large cell carcinomas, and squamous cell carcinomas; and/or to characterize carcinomas of undetermined or mixed pathologies.
  • methylation markers or combinations thereof are selected to provide a positive result (i.e., a result indicating the presence of lung neoplasm) regardless of the type of lung carcinoma present, without differentiating data.
  • circulating epithelial cells representing metastatic tumor cells
  • Cristofanilli M et al. (2004) N Engl J Med 351:781-791
  • Hayes D F et al. (2006) Clin Cancer Res 12:4218-4224
  • Budd G T et al., (2006) Clin Cancer Res 12:6403-6409
  • Moreno J G et al.
  • embodiments of the present disclosure provide compositions and methods for detecting the presence of metastatic cancer in a subject by identifying the presence of methylated markers in plasma or whole blood.
  • the following provides exemplary method for DNA isolation prior to analysis, and an exemplary QUARTS assay, such as may be used in accordance with embodiments of the technology.
  • Application of QUARTS technology to DNA from blood and various tissue samples is described in this example, but the technology is readily applied to other nucleic acid samples, as shown in other examples.
  • genomic DNA may be isolated from cell conditioned media using, for example, the “Maxwell® RSC ccfDNA Plasma Kit (Promega Corp., Madison, Wis.). Following the kit protocol, 1 mL of cell conditioned media (CCM) is used in place of plasma, and processed according to the kit procedure. The elution volume is 100 ⁇ L, of which 70 ⁇ L are generally used for bisulfite conversion.
  • CCM cell conditioned media
  • Samples are mixed using any appropriate device or technology to mix or incubate samples at the temperatures and mixing speeds essentially as described below.
  • a Thermomixer Eppendorf
  • An exemplary desulfonation is as follows:
  • the converted DNA is then used in a detection assay, e.g., a pre-amplification and/or flap endonuclease assays, as described below.
  • a detection assay e.g., a pre-amplification and/or flap endonuclease assays, as described below.
  • the QUARTS technology combines a polymerase-based target DNA amplification process with an invasive cleavage-based signal amplification process.
  • the technology is described, e.g., in U.S. Pat. Nos. 8,361,720; 8,715,937; 8,916,344; and 9,212,392, each of which is incorporated herein by reference.
  • Fluorescence signal generated by the QUARTS reaction is monitored in a fashion similar to real-time PCR and permits quantitation of the amount of a target nucleic acid in a sample.
  • An exemplary QUARTS reaction typically comprises approximately 400-600 n mol/L (e.g., 500 n mol/L) of each primer and detection probe, approximately 100 n mol/L of the invasive oligonucleotide, approximately 600-700 n mol/L of each FRET cassette (FAM, e.g., as supplied commercially by Hologic, Inc.; HEX, e.g., as supplied commercially by BioSearch Technologies; and Quasar 670, e.g., as supplied commercially by BioSearch Technologies), 6.675 ng/ ⁇ L FEN-1 endonuclease (e.g., Cleavase® 2.0, Hologic, Inc.), 1 unit Taq DNA polymerase in a 30 ⁇ L reaction volume (e.g., GoTaq® DNA polymerase, Promega Corp., Madison, Wis.), 10 m mol/L 3-(n-morpholino) propanesulfonic acid (MOPS), 7.5 m mol
  • Stage Temp/Time # of Cycles Denaturation 95° C./3′ 1 Amplification 1 95° C./20′′ 10 67° C./30′′ 70° C./30′′ Amplification 2 95° C./20′′ 37 53° C./1′ 70° C./30′′ Cooling 40° C./30′′ 1 Multiplex Targeted Pre-amplification of Large-Volume Bisulfite-Converted DNA
  • a large volume of the treated DNA may be used in a single, large-volume multiplex amplification reaction.
  • DNA is extracted from a cell lines (e.g., DFCI032 cell line (adenocarcinoma); H1755 cell line (neuroendocrine), using, for example, the Maxwell Promega blood kit # AS1400, as described above.
  • the DNA is bisulfate converted, e.g., as described above.
  • a pre-amplification is conducted, for example, in a reaction mixture containing 7.5 mM MgCl 2 , 10 mM MOPS, 0.3 mM Tris-HC1, pH 8.0, 0.8 mM KC1, 0.1 ⁇ g/ ⁇ L BSA, 0.0001% Tween-20, 0.0001% IGEPAL CA-630, 250 ⁇ M each dNTP, oligonucleotide primers, (e.g., for 12 targets, 12 primer pairs/24 primers, in equimolar amounts (including but not limited to the ranges of, e.g., 200-500 nM each primer), or with individual primer concentrations adjusted to balance amplification efficiencies of the different target regions), 0.025 units/ ⁇ L HotStart GoTaq concentration, and 20 to 50% by volume of bisulfate-treated target DNA (e.g., 10 ⁇ L of target DNA into a 50 ⁇ L reaction mixture, or 50 ⁇ L of target DNA into a 125 ⁇ L reaction mixture).
  • Stage Temp/Time #of Cycles Pre-incubation 95° C./5′ 1 Amplification 1 95° C./30′′ 10 64° C./30′′ 72° C./30′′ Cooling 4° C./Hold 1
  • aliquots of the pre-amplification reaction are diluted to 500 ⁇ L in 10 mM Tris, 0.1 mM EDTA, with or without fish DNA. Aliquots of the diluted pre-amplified DNA (e.g., 10 ⁇ L) are used in a QUARTS PCR-flap assay, e.g., as described above. See also U.S. Patent Appl. Ser. No. 62/249,097, filed Oct. 30, 2015; application Ser. No. 15/335,096, filed Oct. 26, 2016, and PCT/US16/58875, filed Oct. 26, 2016, each of which is incorporated herein by reference in its entirety for all purposes.
  • RRBS Reduced Representation Bisulfite Sequencing
  • FIGS. 2-5 RRBS data for different lung cancer tissue types is shown in FIGS. 2-5 . Based on the criteria above, the markers shown in the table below were selected and QuARTS flap assays were designed for them, as shown in FIG. 1 .
  • 264 tissue samples were obtained from various commercial and non-commercial sources (Asuragen, BioServe, ConversantBio, Cureline, Mayo Clinic, M D Anderson, and PrecisionMed), as shown below in Table 2.
  • Tissue sections were examined by a pathologist, who circled histologically distinct lesions to direct the micro-dissection.
  • Total nucleic acid extraction was performed using the Promega Maxwell RSC system.
  • Formalin-fixed, paraffin-embedded (FFPE) slides were scraped and the DNA was extracted using the Maxwell® RSC DNA FFPE Kit (#AS1450) using the manufacturer's procedure but skipping the RNase treatment step. The same procedure was used for FFPE curls.
  • FFPE Maxwell® RSC DNA FFPE Kit
  • a modified procedure using the lysis buffer from the RSC DNA FFPE kit with the Maxwell® RSC Blood DNA kit (#AS1400) was utilized omitting the RNase step. Samples were eluted in 10 mM Tris, 0.1 mM EDTA, pH 8.5 and 10 uL were used to setup 6 multiplex PCR reactions.
  • LOC100129726 SPOCK2, TSC22D4, PARP15, MAX.chr8.145105646-145105653, ST8SIA1_22, ZDHHC1, BIN2_Z, SKI, DNMT3A, BCL2L11, RASSF1, FERMT3, and BTACT.
  • MATK MATK, SHOX2, BCAT1, SUCLG2, BIN2, PRKAR1B, SHROOM1, S1PR4, NFIX, and BTACT.
  • Each multiplex PCR reaction was setup to a final concentration of 0.2 ⁇ M reaction buffer, 0.2 ⁇ M each primer, 0.05 ⁇ M Hotstart Go Taq (5U/ ⁇ L), resulting in 40 ⁇ L, of master mix that was combined with 10 ⁇ L of DNA template for a final reaction volume of 50 ⁇ L.
  • the thermal profile for the multiplex PCR entailed a pre-incubation stage of 95° for 5 minutes, 10 cycles of amplification at 95° for 30 seconds, 64° for 30 seconds, 72° for 30 seconds, and a cooling stage of 4° that was held until further processing.
  • the PCR product was diluted 1:10 using a diluent of 20 ng/ ⁇ L of fish DNA (e.g., in water or buffer, see U.S. Pat. No. 9,212,392, incorporated herein by reference) and 10 ⁇ L of diluted amplified sample were used for each QuARTS assay reaction.
  • Each QuARTS assay was configured in triplex form, consisting of 2 methylation markers and BTACT as the reference gene.
  • markers that were selected based on RRBS criteria with ⁇ 0.5% methylation in normal tissue and >10% methylation in cancer tissue were included. This resulted in 51 markers for further analysis.
  • the sensitivities for the 51 markers are shown below.
  • Combinations of markers may be used to increase specificity and sensitivity.
  • a combination of the 8 markers SLC12A8, KLHDC7B, PARP15, OPLAH, BCL2L11, MAX.chr12.526, HOXB2, and EMX1 resulted in 98.5% sensitivity (134/136 cancers) for all of the cancer tissues tested, with 100% specificity.
  • markers are selected for sensitive and specific detection associated with a particular type of lung cancer tissue, e.g., adenocarcinoma, large cell carcinoma, squamous cell carcinoma, or small cell carcinoma, e.g., by use of markers that show sensitivity and specificity for particular cancer types or combinations of types.
  • This panel of methylated DNA markers assayed on tissue achieves extremely high discrimination for all types of lung cancer while remaining negative in normal lung tissue and benign nodules.
  • Assays for this panel of markers can be also be applied to blood or bodily fluid-based testing, and finds applications in, e.g., lung cancer screening and discrimination of malignant from benign nodules.
  • markers in Example 2 From the list of markers in Example 2, 30 markers were selected for use in testing DNA from plasma samples from 295 subjects (64 with lung cancer, 231 normal controls. DNA was extracted from 2 mL of plasma from each subject and treated with bisulfite as described in Example 1. Aliquots of the bisulfite-converted DNA were used in two multiplex QuARTS assays, as described in Example 1. The markers selected for analysis are:
  • the target sequences, bisulfite converted target sequences, and the assay oligonucleotides for these markers were as shown in FIG. 1 .
  • the primers and flap oligonucleotides (probes) used for each converted target were as follows:
  • the B3GALT6 marker is used as both a cancer methylation marker and as a reference target. See U.S. Patent Application Ser. No. 62/364,082, filed Jul. 19, 2016, which is incorporated herein by reference in its entirety. ⁇ For zebrafish reference DNA see U.S. Patent Application Ser. No. 62/364,049, filed Jul. 19, 2016, which is incorporated herein by reference in its entirety.
  • the DNA prepared from plasma as described above was amplified in two multiplexed pre-amplification reactions, as described in Example 1.
  • the multiplex pre-amplification reactions comprised reagents to amplify the following marker combinations.
  • ZF_RASSF1-B3GALT6-BTACT ZBA Triplex
  • BARX1-SLC12A8-BTACT BAR2 Triplex
  • PARP15-MAX.chr8.124-BTACT PMA Triplex
  • SHOX2-ZDHHC1-BTACT SZA2 Triplex
  • BIN2_Z-SKI-BTACT BA Triplex
  • DBA Triplex DNMT3A-BCL2L11-BTACT
  • DBA Triplex TBX15-FERMT3-BTACT
  • PRKCB_28-SOBP-BTACT PSA2 Triplex
  • ZF_RASSF1-B3GALT6-BTACT ZBA Triplex
  • MAX.chr8.145-MAX_chr10.226-BTACT MMA2 Triplex
  • MAX.chr12.526-FLJ45983-BTACT MFA Triplex
  • HOXA9-EMX1-BTACT HEA Triplex
  • SP9-DMRTA2-BTACT SDA Triplex
  • OPLAH-CYP26C1-BTACT OPLAH-CYP26C1-BTACT
  • ZNF781-DLX4-BTACT ZDA Triplex
  • SUCLG2-KLHDC7B-BTACT SKA Triplex
  • SNA Triplex S1PR4-NFIX-BTACT
  • the dye reporters used on the FRET cassettes for each member of the triplexes listed above is FAM-HEX-Quasar670, respectively.
  • Plasmids containing target DNA sequences were used to calibrate the quantitative reactions.
  • a series of 10 ⁇ calibrator dilution stocks having from 10 to 10 6 copies of the target strand per ⁇ l in fish DNA diluent (20 ng/mL fish DNA in 10 mM Tris-HCl, 0.1 mM EDTA) were prepared.
  • a combined stock having plasmids that contain each of the targets of the triplex were used.
  • a mixture having each plasmid at 1 ⁇ 10 5 copies per ⁇ L was prepared and used to create a 1:10 dilution series. Strands in unknown samples were back calculated using standard curves generated by plotting Cp vs Log (strands of plasmid).
  • methylation markers are selected that exhibit high performance in detecting methylation associated with specific types of lung cancer.
  • a sample is collected, e.g., a plasma sample, and DNA is isolated from the sample and treated with bisulfite reagent, e.g., as described in Example 1.
  • the converted DNA is analyzed using a multiplex PCR followed by QuARTS flap endonuclease assay as described in Example 1, configured to provide different identifiable signals for different methylation markers or combinations of methylation markers, thereby providing data sets configured to specifically identify the presence of one or more different types of lung carcinoma in the subject (e.g., adenocarcinoma, large cell carcinoma, squamous cell carcinoma, and/or small cell carcinoma).
  • a report is generated indicating the presence or absence of an assay result indicative of the presence of lung carcinoma and, if present, further indicative of the presence of one or more identified types of lung carcinoma.
  • samples from a subject are collected over the course of a period of time or a course of treatment, and assay results are compared to monitor changes in the cancer pathology.
  • Marker and marker panels sensitive to different types of lung cancer find use, e.g., in classifying type(s) of cancer present, identifying mixed pathologies, and/or in monitoring cancer progression over time and/or in response to treatment.

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)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
US15/471,337 2016-05-05 2017-03-28 Detection of lung neoplasia by analysis of methylated DNA Active 2037-06-16 US10385406B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/471,337 US10385406B2 (en) 2016-05-05 2017-03-28 Detection of lung neoplasia by analysis of methylated DNA
US16/460,647 US11028447B2 (en) 2016-05-05 2019-07-02 Detection of neoplasia by analysis of methylated dna
US17/314,099 US20210262042A1 (en) 2016-05-05 2021-05-07 Detection and analysis of methylated dna

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662332295P 2016-05-05 2016-05-05
US201762462677P 2017-02-23 2017-02-23
US15/471,337 US10385406B2 (en) 2016-05-05 2017-03-28 Detection of lung neoplasia by analysis of methylated DNA

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/460,647 Continuation US11028447B2 (en) 2016-05-05 2019-07-02 Detection of neoplasia by analysis of methylated dna

Publications (2)

Publication Number Publication Date
US20170335401A1 US20170335401A1 (en) 2017-11-23
US10385406B2 true US10385406B2 (en) 2019-08-20

Family

ID=60203203

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/471,337 Active 2037-06-16 US10385406B2 (en) 2016-05-05 2017-03-28 Detection of lung neoplasia by analysis of methylated DNA
US16/460,647 Active US11028447B2 (en) 2016-05-05 2019-07-02 Detection of neoplasia by analysis of methylated dna
US17/314,099 Pending US20210262042A1 (en) 2016-05-05 2021-05-07 Detection and analysis of methylated dna

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/460,647 Active US11028447B2 (en) 2016-05-05 2019-07-02 Detection of neoplasia by analysis of methylated dna
US17/314,099 Pending US20210262042A1 (en) 2016-05-05 2021-05-07 Detection and analysis of methylated dna

Country Status (7)

Country Link
US (3) US10385406B2 (ko)
EP (1) EP3452616A4 (ko)
KR (1) KR102436270B1 (ko)
CN (2) CN109563546B (ko)
AU (2) AU2017260630B2 (ko)
CA (1) CA3022911A1 (ko)
WO (1) WO2017192221A1 (ko)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020112869A1 (en) * 2018-11-27 2020-06-04 Exact Sciences Development Company, Llc Characterizing methylated dna, rna, and proteins in the detection of lung neoplasia
US10704081B2 (en) 2015-10-30 2020-07-07 Exact Sciences Development Company, Llc Multiplex amplification detection assay
US11028447B2 (en) 2016-05-05 2021-06-08 Exact Sciences Development Company, Llc Detection of neoplasia by analysis of methylated dna
US11118228B2 (en) 2017-01-27 2021-09-14 Exact Sciences Development Company, Llc Detection of colon neoplasia by analysis of methylated DNA
US11193168B2 (en) 2017-12-13 2021-12-07 Exact Sciences Development Company, Llc Multiplex amplification detection assay II
US11345949B2 (en) 2016-07-19 2022-05-31 Exact Sciences Corporation Methylated control DNA
US11479823B2 (en) 2016-05-05 2022-10-25 Exact Sciences Corporation Detection of lung neoplasia by amplification of RNA sequences

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3126529B1 (en) 2014-03-31 2020-05-27 Mayo Foundation for Medical Education and Research Detecting colorectal neoplasm
KR20180081042A (ko) 2015-08-31 2018-07-13 메이오 파운데이션 포 메디칼 에쥬케이션 앤드 리써치 위 신생물 검출 방법
AU2018374176A1 (en) * 2017-11-30 2020-06-11 Exact Sciences Corporation Detecting breast cancer
EP3792363B1 (en) * 2018-01-23 2024-06-26 Excellen Medical Technology Co., Ltd. Method and kit for identifying lung cancer status
US20210388445A1 (en) * 2018-04-12 2021-12-16 Singlera Genomics, Inc. Compositions and methods for cancer and neoplasia assessment
CN111363812B (zh) * 2018-12-25 2023-12-19 广州康立明生物科技股份有限公司 基于dmrta2基因的肺癌诊断剂及试剂盒
KR102085669B1 (ko) * 2019-01-02 2020-03-06 한국 한의학 연구원 Cyp26c1 유전자의 메틸화 수준을 이용한 소혈관폐색증의 예측 또는 진단을 위한 정보제공방법 및 이를 위한 조성물
CN109504780B (zh) * 2019-01-21 2021-09-14 深圳市新合生物医疗科技有限公司 用于肺癌检测的DNA甲基化qPCR试剂盒及使用方法
EP3914733A4 (en) * 2019-01-24 2023-05-17 Mayo Foundation for Medical Education and Research ENDOMETRIAL CANCER DETECTION
CN113811622A (zh) * 2019-04-03 2021-12-17 梅约医学教育与研究基金会 在血浆中检测胰腺导管腺癌
WO2020214798A1 (en) * 2019-04-17 2020-10-22 The Brigham And Women's Hospital, Inc. Epigenetic signatures of alzheimer's disease
US11396679B2 (en) 2019-05-31 2022-07-26 Universal Diagnostics, S.L. Detection of colorectal cancer
US11001898B2 (en) 2019-05-31 2021-05-11 Universal Diagnostics, S.L. Detection of colorectal cancer
EP4022093A4 (en) * 2019-08-27 2024-05-22 Exact Sciences Corporation CHARACTERIZATION OF METHYLATED DNA, RNA AND PROTEINS IN SUBJECTS SUSPECTED TO HAVE LUNG NEOPLASIA
CN110632307A (zh) * 2019-10-25 2019-12-31 四川大学华西医院 Suclg2自身抗体检测试剂在制备肺癌筛查试剂盒中的用途
US11702704B2 (en) * 2019-10-31 2023-07-18 Mayo Foundation For Medical Education And Research Detecting ovarian cancer
US11898199B2 (en) 2019-11-11 2024-02-13 Universal Diagnostics, S.A. Detection of colorectal cancer and/or advanced adenomas
CN113637745B (zh) * 2020-04-27 2022-12-06 广州市基准医疗有限责任公司 用于检测肺结节良恶性的甲基化分子标记物或其组合和应用
CN113637746B (zh) * 2020-04-27 2022-09-20 广州市基准医疗有限责任公司 用于检测肺结节良恶性的甲基化分子标记物或其组合和应用
CN111676286B (zh) * 2020-05-29 2023-04-14 武汉爱基百客生物科技有限公司 肺癌血浆游离dna甲基化检测用的多重pcr引物系统、检测方法及应用
WO2022002424A1 (en) 2020-06-30 2022-01-06 Universal Diagnostics, S.L. Systems and methods for detection of multiple cancer types
EP3945135A1 (en) * 2020-07-27 2022-02-02 Les Laboratoires Servier Biomarkers for diagnosing and monitoring lung cancer
CN113355414B (zh) * 2021-06-04 2023-05-05 武汉艾米森生命科技有限公司 食管癌检测试剂盒及其应用
WO2024096536A1 (ko) * 2022-10-31 2024-05-10 주식회사 지씨지놈 폐암 진단용 dna 메틸화 마커 및 이의 용도

Citations (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4965188A (en) 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US5011769A (en) 1985-12-05 1991-04-30 Meiogenics U.S. Limited Partnership Methods for detecting nucleic acid sequences
US5124246A (en) 1987-10-15 1992-06-23 Chiron Corporation Nucleic acid multimers and amplified nucleic acid hybridization assays using same
US5288609A (en) 1984-04-27 1994-02-22 Enzo Diagnostics, Inc. Capture sandwich hybridization method and composition
US5338671A (en) 1992-10-07 1994-08-16 Eastman Kodak Company DNA amplification with thermostable DNA polymerase and polymerase inhibiting antibody
WO1995000669A1 (en) 1993-06-22 1995-01-05 Pharmacia Biotech Ab Parallel primer extension approach to nucleic acid sequence analysis
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
US5409818A (en) 1988-02-24 1995-04-25 Cangene Corporation Nucleic acid amplification process
WO1995015373A2 (en) 1993-11-30 1995-06-08 Mcgill University Inhibition of dna methyltransferase
US5494810A (en) 1990-05-03 1996-02-27 Cornell Research Foundation, Inc. Thermostable ligase-mediated DNA amplifications system for the detection of genetic disease
US5508169A (en) 1990-04-06 1996-04-16 Queen's University At Kingston Indexing linkers
US5624802A (en) 1987-10-15 1997-04-29 Chiron Corporation Nucleic acid multimers and amplified nucleic acid hybridization assays using same
US5639611A (en) 1988-12-12 1997-06-17 City Of Hope Allele specific polymerase chain reaction
US5660988A (en) 1993-11-17 1997-08-26 Id Biomedical Corporation Cycling probe cleavage detection of nucleic acid sequences
WO1997046705A1 (en) 1996-06-03 1997-12-11 The Johns Hopkins University School Of Medicine Methylation specific detection
US5710264A (en) 1990-07-27 1998-01-20 Chiron Corporation Large comb type branched polynucleotides
US5773258A (en) 1995-08-25 1998-06-30 Roche Molecular Systems, Inc. Nucleic acid amplification using a reversibly inactivated thermostable enzyme
US5786146A (en) 1996-06-03 1998-07-28 The Johns Hopkins University School Of Medicine Method of detection of methylated nucleic acid using agents which modify unmethylated cytosine and distinguishing modified methylated and non-methylated nucleic acids
US5792614A (en) 1994-12-23 1998-08-11 Dade Behring Marburg Gmbh Detection of nucleic acids by target-catalyzed product formation
US5846717A (en) 1996-01-24 1998-12-08 Third Wave Technologies, Inc. Detection of nucleic acid sequences by invader-directed cleavage
US5851770A (en) 1994-04-25 1998-12-22 Variagenics, Inc. Detection of mismatches by resolvase cleavage using a magnetic bead support
US5882867A (en) 1995-06-07 1999-03-16 Dade Behring Marburg Gmbh Detection of nucleic acids by formation of template-dependent product
WO1999028498A2 (de) 1997-11-27 1999-06-10 Epigenomics Gmbh Verfahren zur herstellung komplexer dna-methylierungs-fingerabdrücke
US5914230A (en) 1995-12-22 1999-06-22 Dade Behring Inc. Homogeneous amplification and detection of nucleic acids
US5958692A (en) 1994-04-25 1999-09-28 Variagenics, Inc. Detection of mutation by resolvase cleavage
US5965408A (en) 1996-07-09 1999-10-12 Diversa Corporation Method of DNA reassembly by interrupting synthesis
US5985557A (en) 1996-01-24 1999-11-16 Third Wave Technologies, Inc. Invasive cleavage of nucleic acids
US5994069A (en) 1996-01-24 1999-11-30 Third Wave Technologies, Inc. Detection of nucleic acids by multiple sequential invasive cleavages
US6013170A (en) 1997-06-12 2000-01-11 Clinical Micro Sensors, Inc. Detection of analytes using reorganization energy
US6063573A (en) 1998-01-27 2000-05-16 Clinical Micro Sensors, Inc. Cycling probe technology using electron transfer detection
US6110684A (en) 1998-02-04 2000-08-29 Variagenics, Inc. Mismatch detection techniques
US6150097A (en) 1996-04-12 2000-11-21 The Public Health Research Institute Of The City Of New York, Inc. Nucleic acid detection probes having non-FRET fluorescence quenching and kits and assays including such probes
US6183960B1 (en) 1995-11-21 2001-02-06 Yale University Rolling circle replication reporter systems
US6221583B1 (en) 1996-11-05 2001-04-24 Clinical Micro Sensors, Inc. Methods of detecting nucleic acids using electrodes
US6235502B1 (en) 1998-09-18 2001-05-22 Molecular Staging Inc. Methods for selectively isolating DNA using rolling circle amplification
US6251594B1 (en) 1997-06-09 2001-06-26 Usc/Norris Comprehensive Cancer Ctr. Cancer diagnostic method based upon DNA methylation differences
WO2002070755A2 (en) 2000-11-15 2002-09-12 Third Wave Technologies, Inc. Fen endonucleases
WO2005023091A2 (en) 2003-09-05 2005-03-17 The Trustees Of Boston University Method for non-invasive prenatal diagnosis
WO2005038051A2 (en) 2003-10-09 2005-04-28 Epigenomics Ag Improved bisulfite conversion of dna
US20050214926A1 (en) * 2004-02-20 2005-09-29 Ralf Zielenski Adsorption of nucleic acids to a solid phase
US20070202525A1 (en) 2006-02-02 2007-08-30 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US7662594B2 (en) 2002-09-20 2010-02-16 New England Biolabs, Inc. Helicase-dependent amplification of RNA
US20110160446A1 (en) * 2008-05-30 2011-06-30 Qiagen Gmbh Method for isolating short-chain nucleic acids
WO2012155072A2 (en) 2011-05-12 2012-11-15 Exact Sciences Corporation Isolation of nucleic acids
US8361720B2 (en) 2010-11-15 2013-01-29 Exact Sciences Corporation Real time cleavage assay
WO2013116375A1 (en) 2012-01-30 2013-08-08 Exact Sciences Corporation Modification of dna on magnetic beads
US8715937B2 (en) 2010-11-15 2014-05-06 Exact Sciences Corporation Mutation detection assay
US8808990B2 (en) 2011-05-12 2014-08-19 Exact Sciences Corporation Serial isolation of multiple DNA targets from stool
US8916344B2 (en) 2010-11-15 2014-12-23 Exact Sciences Corporation Methylation assay
US9096893B2 (en) 2008-03-15 2015-08-04 Hologic, Inc. Methods for analysis of nucleic acid molecules during amplification reactions
US9212392B2 (en) 2012-09-25 2015-12-15 Exact Sciences Corporation Normalization of polymerase activity
US20160010081A1 (en) 2014-07-11 2016-01-14 Exact Sciences Corporation Systems, methods and kits for extracting nucleic acid from digestive fluid
US20160168643A1 (en) * 2014-12-12 2016-06-16 Exact Sciences Corporation Compositions and methods for performing methylation detection assays
US20160194721A1 (en) 2014-12-12 2016-07-07 Exact Sciences Corporation Compositions and methods for detecting epithelial cell dna
US20170121704A1 (en) 2015-10-30 2017-05-04 Exact Sciences Corporation Isolation and detection of dna from plasma
US9657511B2 (en) 2008-03-12 2017-05-23 Masonite Corporation Impact resistant door skin, door including the same, and method of manufacturing an impact resistant door skin from a pre-formed door skin
WO2017192221A1 (en) 2016-05-05 2017-11-09 Exact Sciences Corporation Detection of lung neoplasia by analysis of methylated dna
US20180143198A1 (en) * 2014-12-26 2018-05-24 Peking University Method for detecting differentially methylated cpg islands associated with abnormal state of human body

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0874B2 (ja) 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 遺伝子発現を検出および変調するヌクレアーゼ耐性、ピリミジン修飾オリゴヌクレオチド
US6872816B1 (en) 1996-01-24 2005-03-29 Third Wave Technologies, Inc. Nucleic acid detection kits
EP0637965B1 (en) 1991-11-26 2002-10-16 Isis Pharmaceuticals, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
ATE154029T1 (de) 1993-03-30 1997-06-15 Sanofi Sa 7-deazapurin modifizierte oligonukleotide
WO1994024144A2 (en) 1993-04-19 1994-10-27 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US6395524B2 (en) 1996-11-27 2002-05-28 University Of Washington Thermostable polymerases having altered fidelity and method of identifying and using same
US6329178B1 (en) 2000-01-14 2001-12-11 University Of Washington DNA polymerase mutant having one or more mutations in the active site
US6605451B1 (en) 2000-06-06 2003-08-12 Xtrana, Inc. Methods and devices for multiplexing amplification reactions
US7087414B2 (en) 2000-06-06 2006-08-08 Applera Corporation Methods and devices for multiplexing amplification reactions
KR100870486B1 (ko) 2000-09-01 2008-12-11 에피제노믹스 아게 게놈 디엔에이 시료의 서열 콘택스트 5'-씨피쥐-3'에서 특정 시토신의 메틸화 정도 측정방법
US7482118B2 (en) 2001-11-15 2009-01-27 Third Wave Technologies, Inc. Endonuclease-substrate complexes
JP2006508662A (ja) 2002-12-04 2006-03-16 アプレラ コーポレイション ポリヌクレオチドの多重増幅
CA2543033A1 (en) 2003-10-16 2005-04-28 Third Wave Technologies, Inc. Direct nucleic acid detection in bodily fluids
WO2005042713A2 (en) 2003-10-28 2005-05-12 The Johns Hopkins University Quantitative multiplex methylation-specific pcr
EP2395098B1 (en) 2004-03-26 2015-07-15 Agena Bioscience, Inc. Base specific cleavage of methylation-specific amplification products in combination with mass analysis
EP1761782A2 (en) 2004-06-18 2007-03-14 Roche Diagnostics GmbH Use of protein cbp2 as a marker for colorectal cancer
US20070048748A1 (en) 2004-09-24 2007-03-01 Li-Cor, Inc. Mutant polymerases for sequencing and genotyping
MX2007005364A (es) 2004-11-03 2008-01-22 Third Wave Tech Inc Ensayo de deteccion de una sola etapa.
JP2008518610A (ja) 2004-11-03 2008-06-05 アルマック ダイアグノスティックス リミテッド トランスクリプトームマイクロアレイ技法およびそれを使用する方法
ES2382746T3 (es) 2005-04-15 2012-06-13 Epigenomics Ag Método para determinar la metilación de ADN en muestras de sangre u orina
US20090203011A1 (en) 2007-01-19 2009-08-13 Epigenomics Ag Methods and nucleic acids for analyses of cell proliferative disorders
KR101509049B1 (ko) * 2007-04-16 2015-04-06 한국생명공학연구원 위암 특이적 유전자의 조기 탐지를 위한 메틸화 바이오마커
ES2570828T3 (es) 2007-06-08 2016-05-20 Epigenomics Ag Método de análisis de metilación
US9428746B2 (en) 2007-10-31 2016-08-30 Akonni Biosystems, Inc. Method and kit for purifying nucleic acids
US20110318738A1 (en) 2008-12-23 2011-12-29 University Of Utah Research Foundation Identification and regulation of a novel dna demethylase system
EP2233590A1 (en) * 2009-01-28 2010-09-29 AIT Austrian Institute of Technology GmbH Methylation assay
WO2011139920A2 (en) 2010-04-29 2011-11-10 Life Technologies Corporation Methylation-specific competitive allele-specific taqman polymerase chain reaction (cast-pcr)
US20130022974A1 (en) * 2011-06-17 2013-01-24 The Regents Of The University Of Michigan Dna methylation profiles in cancer
WO2014082067A1 (en) * 2012-11-26 2014-05-30 The Johns Hopkins University Methods and compositions for diagnosing and treating gastric cancer
CN105378107A (zh) 2013-03-14 2016-03-02 雅培分子公司 多重甲基化-特异性扩增系统和方法
US10253358B2 (en) 2013-11-04 2019-04-09 Exact Sciences Development Company, Llc Multiple-control calibrators for DNA quantitation
US10138524B2 (en) 2013-12-19 2018-11-27 Exact Sciences Development Company, Llc Synthetic nucleic acid control molecules
EP3126529B1 (en) 2014-03-31 2020-05-27 Mayo Foundation for Medical Education and Research Detecting colorectal neoplasm
US10184154B2 (en) * 2014-09-26 2019-01-22 Mayo Foundation For Medical Education And Research Detecting cholangiocarcinoma
SI3408407T1 (sl) 2016-01-29 2021-04-30 Epigenomics Ag Postopki za odkrivanje CPG metilacije iz tumorja pridobljene DNA v vzorcih krvi
AU2017281099A1 (en) 2016-06-21 2019-01-03 The Wistar Institute Of Anatomy And Biology Compositions and methods for diagnosing lung cancers using gene expression profiles
CA3049459A1 (en) 2017-01-27 2018-08-02 Exact Sciences Development Company, Llc Detection of colon neoplasia by analysis of methylated dna
US10648025B2 (en) 2017-12-13 2020-05-12 Exact Sciences Development Company, Llc Multiplex amplification detection assay II
CN113423410A (zh) 2018-11-27 2021-09-21 精密科学发展有限责任公司 在肺肿瘤检测中表征甲基化dna、rna和蛋白质

Patent Citations (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5288609A (en) 1984-04-27 1994-02-22 Enzo Diagnostics, Inc. Capture sandwich hybridization method and composition
US4683202B1 (ko) 1985-03-28 1990-11-27 Cetus Corp
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5011769A (en) 1985-12-05 1991-04-30 Meiogenics U.S. Limited Partnership Methods for detecting nucleic acid sequences
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683195B1 (ko) 1986-01-30 1990-11-27 Cetus Corp
US4965188A (en) 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US5124246A (en) 1987-10-15 1992-06-23 Chiron Corporation Nucleic acid multimers and amplified nucleic acid hybridization assays using same
US5624802A (en) 1987-10-15 1997-04-29 Chiron Corporation Nucleic acid multimers and amplified nucleic acid hybridization assays using 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
US5409818A (en) 1988-02-24 1995-04-25 Cangene Corporation Nucleic acid amplification process
US5639611A (en) 1988-12-12 1997-06-17 City Of Hope Allele specific polymerase chain reaction
US5508169A (en) 1990-04-06 1996-04-16 Queen's University At Kingston Indexing linkers
US5494810A (en) 1990-05-03 1996-02-27 Cornell Research Foundation, Inc. Thermostable ligase-mediated DNA amplifications system for the detection of genetic disease
US5849481A (en) 1990-07-27 1998-12-15 Chiron Corporation Nucleic acid hybridization assays employing large comb-type branched polynucleotides
US5710264A (en) 1990-07-27 1998-01-20 Chiron Corporation Large comb type branched polynucleotides
US5338671A (en) 1992-10-07 1994-08-16 Eastman Kodak Company DNA amplification with thermostable DNA polymerase and polymerase inhibiting antibody
WO1995000669A1 (en) 1993-06-22 1995-01-05 Pharmacia Biotech Ab Parallel primer extension approach to nucleic acid sequence analysis
US5660988A (en) 1993-11-17 1997-08-26 Id Biomedical Corporation Cycling probe cleavage detection of nucleic acid sequences
WO1995015373A2 (en) 1993-11-30 1995-06-08 Mcgill University Inhibition of dna methyltransferase
US5958692A (en) 1994-04-25 1999-09-28 Variagenics, Inc. Detection of mutation by resolvase cleavage
US5851770A (en) 1994-04-25 1998-12-22 Variagenics, Inc. Detection of mismatches by resolvase cleavage using a magnetic bead support
US6121001A (en) 1994-12-23 2000-09-19 Dade Behring Marburg Gmbh Detection of nucleic acids by target-catalyzed product formation
US6110677A (en) 1994-12-23 2000-08-29 Dade Behring Marburg Gmbh Oligonucleotide modification, signal amplification, and nucleic acid detection by target-catalyzed product formation
US5792614A (en) 1994-12-23 1998-08-11 Dade Behring Marburg Gmbh Detection of nucleic acids by target-catalyzed product formation
US5882867A (en) 1995-06-07 1999-03-16 Dade Behring Marburg Gmbh Detection of nucleic acids by formation of template-dependent product
US5773258A (en) 1995-08-25 1998-06-30 Roche Molecular Systems, Inc. Nucleic acid amplification using a reversibly inactivated thermostable enzyme
US6210884B1 (en) 1995-11-21 2001-04-03 Yale University Rolling circle replication reporter systems
US6183960B1 (en) 1995-11-21 2001-02-06 Yale University Rolling circle replication reporter systems
US5914230A (en) 1995-12-22 1999-06-22 Dade Behring Inc. Homogeneous amplification and detection of nucleic acids
US6090543A (en) 1996-01-24 2000-07-18 Third Wave Technologies, Inc. Cleavage of nucleic acids
US5985557A (en) 1996-01-24 1999-11-16 Third Wave Technologies, Inc. Invasive cleavage of nucleic acids
US5994069A (en) 1996-01-24 1999-11-30 Third Wave Technologies, Inc. Detection of nucleic acids by multiple sequential invasive cleavages
US6001567A (en) 1996-01-24 1999-12-14 Third Wave Technologies, Inc. Detection of nucleic acid sequences by invader-directed cleavage
US5846717A (en) 1996-01-24 1998-12-08 Third Wave Technologies, Inc. Detection of nucleic acid sequences by invader-directed cleavage
US6150097A (en) 1996-04-12 2000-11-21 The Public Health Research Institute Of The City Of New York, Inc. Nucleic acid detection probes having non-FRET fluorescence quenching and kits and assays including such probes
US5786146A (en) 1996-06-03 1998-07-28 The Johns Hopkins University School Of Medicine Method of detection of methylated nucleic acid using agents which modify unmethylated cytosine and distinguishing modified methylated and non-methylated nucleic acids
WO1997046705A1 (en) 1996-06-03 1997-12-11 The Johns Hopkins University School Of Medicine Methylation specific detection
US5965408A (en) 1996-07-09 1999-10-12 Diversa Corporation Method of DNA reassembly by interrupting synthesis
US6221583B1 (en) 1996-11-05 2001-04-24 Clinical Micro Sensors, Inc. Methods of detecting nucleic acids using electrodes
US7037650B2 (en) 1997-06-09 2006-05-02 University Of Southern California Cancer diagnostic method based upon DNA methylation differences
US6251594B1 (en) 1997-06-09 2001-06-26 Usc/Norris Comprehensive Cancer Ctr. Cancer diagnostic method based upon DNA methylation differences
US6013170A (en) 1997-06-12 2000-01-11 Clinical Micro Sensors, Inc. Detection of analytes using reorganization energy
US6248229B1 (en) 1997-06-12 2001-06-19 Clinical Micro Sensors, Inc. Detection of analytes using reorganization energy
WO1999028498A2 (de) 1997-11-27 1999-06-10 Epigenomics Gmbh Verfahren zur herstellung komplexer dna-methylierungs-fingerabdrücke
US6063573A (en) 1998-01-27 2000-05-16 Clinical Micro Sensors, Inc. Cycling probe technology using electron transfer detection
US6110684A (en) 1998-02-04 2000-08-29 Variagenics, Inc. Mismatch detection techniques
US6235502B1 (en) 1998-09-18 2001-05-22 Molecular Staging Inc. Methods for selectively isolating DNA using rolling circle amplification
WO2002070755A2 (en) 2000-11-15 2002-09-12 Third Wave Technologies, Inc. Fen endonucleases
US7662594B2 (en) 2002-09-20 2010-02-16 New England Biolabs, Inc. Helicase-dependent amplification of RNA
WO2005023091A2 (en) 2003-09-05 2005-03-17 The Trustees Of Boston University Method for non-invasive prenatal diagnosis
WO2005038051A2 (en) 2003-10-09 2005-04-28 Epigenomics Ag Improved bisulfite conversion of dna
US20050214926A1 (en) * 2004-02-20 2005-09-29 Ralf Zielenski Adsorption of nucleic acids to a solid phase
US20070202525A1 (en) 2006-02-02 2007-08-30 The Board Of Trustees Of The Leland Stanford Junior University Non-invasive fetal genetic screening by digital analysis
US9657511B2 (en) 2008-03-12 2017-05-23 Masonite Corporation Impact resistant door skin, door including the same, and method of manufacturing an impact resistant door skin from a pre-formed door skin
US9096893B2 (en) 2008-03-15 2015-08-04 Hologic, Inc. Methods for analysis of nucleic acid molecules during amplification reactions
US20110160446A1 (en) * 2008-05-30 2011-06-30 Qiagen Gmbh Method for isolating short-chain nucleic acids
US8361720B2 (en) 2010-11-15 2013-01-29 Exact Sciences Corporation Real time cleavage assay
US8715937B2 (en) 2010-11-15 2014-05-06 Exact Sciences Corporation Mutation detection assay
US8916344B2 (en) 2010-11-15 2014-12-23 Exact Sciences Corporation Methylation assay
US8808990B2 (en) 2011-05-12 2014-08-19 Exact Sciences Corporation Serial isolation of multiple DNA targets from stool
US9000146B2 (en) 2011-05-12 2015-04-07 Exact Sciences Corporation Isolation of nucleic acids
WO2012155072A2 (en) 2011-05-12 2012-11-15 Exact Sciences Corporation Isolation of nucleic acids
US9163278B2 (en) 2011-05-12 2015-10-20 Exact Sciences Corporation Isolation of nucleic acids
US9169511B2 (en) 2011-05-12 2015-10-27 Exact Sciences Corporation Isolation of nucleic acids
WO2013116375A1 (en) 2012-01-30 2013-08-08 Exact Sciences Corporation Modification of dna on magnetic beads
US9315853B2 (en) 2012-01-30 2016-04-19 Exact Sciences Corporation Modification of DNA on magnetic beads
US9212392B2 (en) 2012-09-25 2015-12-15 Exact Sciences Corporation Normalization of polymerase activity
US20160010081A1 (en) 2014-07-11 2016-01-14 Exact Sciences Corporation Systems, methods and kits for extracting nucleic acid from digestive fluid
US20160168643A1 (en) * 2014-12-12 2016-06-16 Exact Sciences Corporation Compositions and methods for performing methylation detection assays
US20160194721A1 (en) 2014-12-12 2016-07-07 Exact Sciences Corporation Compositions and methods for detecting epithelial cell dna
US20180143198A1 (en) * 2014-12-26 2018-05-24 Peking University Method for detecting differentially methylated cpg islands associated with abnormal state of human body
US20170121704A1 (en) 2015-10-30 2017-05-04 Exact Sciences Corporation Isolation and detection of dna from plasma
US20170121757A1 (en) 2015-10-30 2017-05-04 Exact Sciences Corporation Multiplex amplification detection assay
WO2017075061A1 (en) 2015-10-30 2017-05-04 Exact Sciences Corporation Multiplex amplification detection assay and isolation and detection of dna from plasma
WO2017192221A1 (en) 2016-05-05 2017-11-09 Exact Sciences Corporation Detection of lung neoplasia by analysis of methylated dna

Non-Patent Citations (70)

* Cited by examiner, † Cited by third party
Title
Antequera et al., High levels of de novo methylation and altered chromatin structure at CpG islands in cell lines. Cell. Aug. 10, 1990;62(3):503-14.
Ballabio, et al., Screening for steroid sulfatase (STS) gene deletions by multiplex DNA amplification, Human Genetics, 1990, 84(6): 571-573.
Barnay, Genetic disease detection and DNA amplification using cloned thermostable ligase, Proc. Natl. Acad. Sci USA, 1991, 88:189-93.
Budd et al., Circulating tumor cells versus imaging-predicting overall survival in metastatic breast cancer. Clin Cancer Res. Nov. 1, 2006;12(21):6403-9.
Budd et al., Circulating tumor cells versus imaging—predicting overall survival in metastatic breast cancer. Clin Cancer Res. Nov. 1, 2006;12(21):6403-9.
Bustin, Absolute quantification of mRNA using real-time reverse transcription polymerase chain reaction assays, J. Molecular Endocrinology, 2000, 25:169-193.
Carvalho et al., Genome-wide DNA methylation profiling of non-small cell lung carcinomas. Epigenetics Chromatin. Jun. 22, 2012;5(1):9.
Ceska et al., Structure-specific DNA cleavage by 5′ nucleases. Trends Biochem Sci. Sep. 1998;23(9):331-6.
Chamberlain et al., Deletion screening of the Duchenne muscular dystrophy locus via multiplex DNA amplification, Nucleic Acids Research, 1988, 16(23):11141-11156.
Cohen et al., Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. Jul. 1, 2008;26(19):3213-21.
Cristofanilli et al., Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. Aug. 19, 2004;351(8):781-91.
Don et al., ‘Touchdown’ PCR to circumvent spurious priming during gene amplification, Nucleic Acids Research, 1991, 19(14):4008.
Don et al., 'Touchdown' PCR to circumvent spurious priming during gene amplification, Nucleic Acids Research, 1991, 19(14):4008.
Eads et al., CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltransferase overexpression. Cancer Res. May 15, 1999;59(10):2302-6.
Feil et al., Methylation analysis on individual chromosomes: improved protocol for bisulphite genomic sequencing. Nucleic Acids Res. Feb. 25, 1994;22(4):695-6.
Frommer et al., A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci U S A. Mar. 1, 1992;89(5):1827-31.
Gonzalgo et al., Identification and characterization of differentially methylated regions of genomic DNA by methylation-sensitive arbitrarily primed PCR. Cancer Res. Feb. 15, 1997;57(4):594-9.
Gonzalgo et al., Rapid quantitation of methylation differences at specific sites using methylation-sensitive single nucleotide primer extension (Ms-SNuPE). Nucleic Acids Res. Jun. 15, 1997;25(12):2529-31.
Grafstrom et al., The characteristics of DNA methylation in an in vitro DNA synthesizing system from mouse fibroblasts. Nucleic Acids Res. Apr. 25, 1985;13(8):2827-42.
Grigg et al., Sequencing 5-methylcytosine residues in genomic DNA. Bioessays. Jun. 1994;16(6):431-6.
Grigg, Sequencing 5-methylcytosine residues by the bisulphite method. DNA Seq. 1996;6(4):189-98.
Gu et al., Genome-scale DNA methylation mapping of clinical samples at single-nucleotide resolution. Nat Methods. Feb. 2010;7(2):133-6.
Guilfoyle et al., Ligation-mediated PCR amplification of specific fragments from a class-II restriction endonuclease total digest, Nucleic Acids Research, 1997, 25:1854-1858.
Hall et al., Sensitive detection of DNA polymorphisms by the serial invasive signal amplification reaction, PNAS, 2000, 97:8272.
Hayden et al., Multiplex-Ready PCR: A new method for multiplexed SSR and SNP genotyping, BMC Genomics, 2008, 9:80.
Hayes et al., Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. Jul. 15, 2006;12(14 Pt 1):4218-24.
Hecker et al., High and low annealing temperatures increase both specificity and yield in touchdown and stepdown PCR, Biotechniques, 1996, 20(3):478-485.
Herman et al., Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 1996; 93: 9821-9826.
Higuchi et al., A general method of in vitro preparation and specific mutagenesis of DNA fragments: study of protein and DNA interactions, Nucleic Acids Research, 1988, 16(15):7351-7367.
Higuchi et al., Simultaneous amplification and detection of specific DNA sequences, Biotechnology, 1992, 10:413-417.
Higuchi et al.,Kinetic PCR analysis: real-time monitoring of DNA amplification reactions, Biotechnology, 1993, 11:1026-1030.
International Search Report and Written Opinion for PCT/US2017/024468, dated Sep. 1, 2017, 17 pages.
Kaiser et al., A comparison of eubacterial and archaeal structure-specific 5′-exonucleases. J Biol Chem. Jul. 23, 1999;274(30):21387-94.
Kalinina et al., Nanoliter scale PCR with TaqMan detection, Nucleic Acids Research, 1997, 25:1999-2004.
Kneip, C. et al., SHOX2 DNA Methylation Is a Biomarker for the Diagnosis of Lung Cancer in Plasma, J. Thoracic Oncol., vol. 6, pp. 1632-1638 (Year: 2011). *
Kober et al., Methyl-CpG binding column-based identification of nine genes hypermethylated in colorectal cancer. Mol Carcinog. Nov. 2011;50(11):846-56.
Kuppuswamy et al., Single nucleotide primer extension to detect genetic diseases: experimental application to hemophilia B (factor IX) and cystic fibrosis genes. Proc Natl Acad Sci U S A. Feb. 15, 1991;88(4):1143-7.
Liu et al., Flap endonuclease 1: a central component of DNA metabolism. Annu Rev Biochem. 2004;73:589-615.
Lyamichev et al.,Polymorphism identification and quantitative detection of genomic DNA by invasive cleavage of oligonucleotide probes, Nat. Biotech., 1999, 17:292-296.
Martin et al., Genomic sequencing indicates a correlation between DNA hypomethylation in the 5′ region of the pS2 gene and its expression in human breast cancer cell lines. Gene. May 19, 1995;157(1-2):261-4.
Meissner et al., Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. Oct. 13, 2005;33(18):5868-77.
Moreno et al., Circulating tumor cells predict survival in patients with metastatic prostate cancer. Urology. Apr. 2005;65(4):713-8.
Nyce et al., Variable effects of DNA-synthesis inhibitors upon DNA methylation in mammalian cells. Nucleic Acids Res. May 27, 1986;14(10):4353-67.
Olek et al., A modified and improved method for bisulphite based cytosine methylation analysis. Nucleic Acids Res. Dec. 15, 1996;24(24):5064-6.
Olek et al., The pre-implantation ontogeny of the H19 methylation imprint. Nat Genet. Nov. 1997;17(3):275-6.
Olivier, The Invader assay for SNP genotyping, Mutat Res. Jun. 3, 2005;573(1-2):103-10.
Orpana, Fluorescence resonance energy transfer (FRET) using ssDNA binding fluorescent dye, Biomol Eng. Apr. 2004;21(2):45-50.
Pantel et al., Detection, clinical relevance and specific biological properties of disseminating tumour cells. Nat Rev Cancer. May 2008;8(5):329-40.
Ponomaryova et al., Potentialities of aberrantly methylated circulating DNA for diagnostics and post-treatment follow-up of lung cancer patients. Lung Cancer. Sep. 2013;81(3):397-403.
Ramsahoye et al., Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc Natl Acad Sci U S A. May 9, 2000;97(10):5237-42.
Rein et al., Identifying 5-methylcytosine and related modifications in DNA genomes. Nucleic Acids Res. May 15, 1998;26(10):2255-64.
Roux, Using mismatched primer-template pairs in touchdown PCR, Biotechniques, 1994, 16(5):812-814.
Sadri et al., Rapid analysis of DNA methylation using new restriction enzyme sites created by bisulfite modification. Nucleic Acids Res. Dec. 15, 1996;24(24):5058-9.
Salomon et al., Methylation of mouse DNA in vivo: di- and tripyrimidine sequences containing 5-methylcytosine. Biochim Biophys Acta. Apr. 15, 1970;204(2):340-51.
Schmidt, B. et al., SHOX2 DNA Methylation is a Biomarker for the Diagnosis of Lung Cancer Based on Bronchial Aspirates, BMC Cancer, vol. 10:600, pp. 1-9 (Year: 2010). *
Schouten et al., Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification, Nucleic Acids Research, 2002, 30(12): e57.
Selvin, Fluorescence resonance energy transfer, 1995, Methods Enzymol. 1995;246:300-34.
Singer-Sam et al., A quantitative Hpall-PCR assay to measure methylation of DNA from a small number of cells. Nucleic Acids Res. Feb. 11, 1990;18(3):687.
Singer-Sam et al., A sensitive, quantitative assay for measurement of allele-specific transcripts differing by a single nucleotide. PCR Methods Appl. Feb. 1992;1(3):160-3.
Stryer, Fluorescence energy transfer as a spectroscopic ruler, Annu Rev Biochem. 1978;47:819-46.
Szabo et al., Allele-specific expression and total expression levels of imprinted genes during early mouse development: implications for imprinting mechanisms. Genes Dev. Dec. 15, 1995;9(24):3097-108.
Toyota et al., Identification of differentially methylated sequences in colorectal cancer by methylated CpG island amplification. Cancer Res. May 15, 1999;59(10):2307-12.
Triglia et al., A procedure for in vitro amplification of DNA segments that lie outside the boundaries of known sequences, Nucleic Acids Res., 1988, 16:8186.
Vogelstein et al., Digital PCR, PNAS, 1999, 96: 9236-41.
Woodcock et al., The majority of methylated deoxycytidines in human DNA are not in the CpG dinucleotide. Biochem Biophys Res Commun. Jun. 15, 1987;145(2):888-94.
Xiong et al., COBRA: a sensitive and quantitative DNA methylation assay. Nucleic Acids Res. Jun. 15, 1997;25(12):2532-4.
Yamada, H. et al., Fluorometric Identification of 5-Methylcytosine Modification in DNA: Combination of Photosensitized Oxidation and Invasive Cleavage, Bioconjugate Chem., vol. 19, pp. 20-23 (Year: 2008). *
Zeschnigk et al., Imprinted segments in the human genome: different DNA methylation patterns in the Prader-Willi/Angelman syndrome region as determined by the genomic sequencing method. Hum Mol Genet. Mar. 1997;6(3):387-95.
Zou et al., Sensitive quantification of methylated markers with a novel methylation specific technology. Abstract D-144, Clin Chem 2010;56(6)Suppl:A199.
Zou, H. et al., Quantification of Methylated Markers with a Multiplex Methylation-Specific Technology, Clin. Chem., vol. 58, pp. 375-383 (Year: 2012). *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10704081B2 (en) 2015-10-30 2020-07-07 Exact Sciences Development Company, Llc Multiplex amplification detection assay
US10822638B2 (en) 2015-10-30 2020-11-03 Exact Sciences Development Company, Llc Isolation and detection of DNA from plasma
US11299766B2 (en) 2015-10-30 2022-04-12 Exact Sciences Corporation Multiplex amplification detection assay
US11674168B2 (en) 2015-10-30 2023-06-13 Exact Sciences Corporation Isolation and detection of DNA from plasma
US11028447B2 (en) 2016-05-05 2021-06-08 Exact Sciences Development Company, Llc Detection of neoplasia by analysis of methylated dna
US11479823B2 (en) 2016-05-05 2022-10-25 Exact Sciences Corporation Detection of lung neoplasia by amplification of RNA sequences
US11345949B2 (en) 2016-07-19 2022-05-31 Exact Sciences Corporation Methylated control DNA
US11118228B2 (en) 2017-01-27 2021-09-14 Exact Sciences Development Company, Llc Detection of colon neoplasia by analysis of methylated DNA
US11193168B2 (en) 2017-12-13 2021-12-07 Exact Sciences Development Company, Llc Multiplex amplification detection assay II
WO2020112869A1 (en) * 2018-11-27 2020-06-04 Exact Sciences Development Company, Llc Characterizing methylated dna, rna, and proteins in the detection of lung neoplasia

Also Published As

Publication number Publication date
US11028447B2 (en) 2021-06-08
CN116064796A (zh) 2023-05-05
AU2017260630B2 (en) 2023-07-13
US20170335401A1 (en) 2017-11-23
CA3022911A1 (en) 2017-11-09
CN109563546B (zh) 2022-09-09
CN109563546A (zh) 2019-04-02
WO2017192221A1 (en) 2017-11-09
EP3452616A1 (en) 2019-03-13
AU2023248106A1 (en) 2023-11-30
KR20190004768A (ko) 2019-01-14
EP3452616A4 (en) 2020-01-22
AU2017260630A8 (en) 2019-03-21
AU2017260630A1 (en) 2018-11-22
KR102436270B1 (ko) 2022-08-25
US20210262042A1 (en) 2021-08-26
US20190330702A1 (en) 2019-10-31

Similar Documents

Publication Publication Date Title
US11028447B2 (en) Detection of neoplasia by analysis of methylated dna
US20220056526A1 (en) Detection of colon neoplasia by analysis of methylated dna
AU2017318700B2 (en) Detecting hepatocellular carcinoma
US20220136058A1 (en) Characterizing methylated dna, rna, and proteins in the detection of lung neoplasia
US20220403471A1 (en) Characterizing methylated dna, rna, and proteins in subjects suspected of having lung neoplasia
US20220340959A1 (en) Methylated control dna
JP7512278B2 (ja) 肺腫瘍の検出におけるメチル化dna、rna、及びタンパク質の特性決定

Legal Events

Date Code Title Description
AS Assignment

Owner name: EXACT SCIENCES CORPORATION, WISCONSIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ALLAWI, HATIM;LIDGARD, GRAHAM P.;GIAKOUMOPOULOS, MARIA;REEL/FRAME:041777/0207

Effective date: 20160607

Owner name: MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AHLQUIST, DAVID A.;MAHONEY, DOUGLAS;TAYLOR, WILLIAM R.;SIGNING DATES FROM 20170323 TO 20170327;REEL/FRAME:041777/0325

AS Assignment

Owner name: EXACT SCIENCES DEVELOPMENT COMPANY, LLC, WISCONSIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:EXACT SCIENCES CORPORATION;REEL/FRAME:044119/0001

Effective date: 20171001

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

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

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

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: EXACT SCIENCES CORPORATION, WISCONSIN

Free format text: MERGER;ASSIGNOR:EXACT SCIENCES DEVELOPMENT COMPANY, LLC;REEL/FRAME:058738/0465

Effective date: 20220101

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4