WO2016109712A1 - Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus - Google Patents

Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus Download PDF

Info

Publication number
WO2016109712A1
WO2016109712A1 PCT/US2015/068131 US2015068131W WO2016109712A1 WO 2016109712 A1 WO2016109712 A1 WO 2016109712A1 US 2015068131 W US2015068131 W US 2015068131W WO 2016109712 A1 WO2016109712 A1 WO 2016109712A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
sequences
methylation
sample
seq
Prior art date
Application number
PCT/US2015/068131
Other languages
French (fr)
Inventor
Sanford D. Markowitz
Joseph E. WILLIS
Helen Moinova
Thomas LAFRAMBOISE
Omar DE LA CRUZ CABRERA
Amitabh Chak
Original Assignee
Case Western Reserve University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Case Western Reserve University filed Critical Case Western Reserve University
Priority to US15/540,956 priority Critical patent/US20190136325A1/en
Priority to CA2972782A priority patent/CA2972782A1/en
Priority to EP15876289.8A priority patent/EP3240912A4/en
Priority to AU2015374019A priority patent/AU2015374019B2/en
Publication of WO2016109712A1 publication Critical patent/WO2016109712A1/en
Priority to AU2022201153A priority patent/AU2022201153A1/en
Priority to US18/381,926 priority patent/US20240068045A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • 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
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • 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
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]
    • 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
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • 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
    • C12Q2600/112Disease subtyping, staging or classification
    • 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
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/16Primer sets for multiplex assays

Definitions

  • the present disclosure is based in part on the discovery of particularuman genomic DNA regions (also referred to herein as informative loci or patches) in which the cytosines within CpG dinucleotides are differentially methylated in esophageal neoplasia compared to normal human tissues.
  • the method comprises assaying for the presence of differentially methylated genomic loci in a tissue sample or a bodily fluid sample from a subject.
  • tissue sample may he obtained from biopsies of the gastrointestinal tract, including hut not limited to the esophagus and stomach, Tissue samples may be obtained as a biopsy, or as a swab or brushing of the gastrointestinal tract (e.g., esophagus or stomach), or other organs believed to contain cancerous cells or tissues.
  • Exemplary bodily fluids include blood, serum, plasma, saliva, spit, esophageal washing, a blood-derived fraction, or urine
  • the method involves methylation-sensitive restriction enzyme(s)
  • the method involves methylation-sensitive PGR.
  • the method involves restriction enzynie/metbylation-sensitive PCR.
  • the method comprises reacting DNA from the sample with a chemical compound that converts non-methylated cytosine bases (also called "con version-sensitive" cytosines), but not methylated cytosine bases, to a different nucleotide base.
  • the chemical compound is sodium bisulfite, which converts unmethylated cytosine bases to uracil.
  • the compound-converted DNA is then amplified using a methylation-sensitive polymerase chain reaction (MSP) employing primers that amplify the compound-converted DNA template if cytosine bases within CpG dinucleotides of the DNA from the sample are methylated, Production of a PCR product indicates that the subject has cancer or precancerous adenomas, in some embodiments, compound-converted DNA is amplified by bisulfite specific methylation indifferent PCR primers and methylation of the parental DNA template is inferred by DNA sequence analysis of the bisulfite converted and amplified product. Other methods for assaying for the presence of methylated DNA are known in the art. In certain embodiments, the method optionally comprises detecting the presence or absence of a somatic mutation in TP53.
  • MSP methylation-sensitive polymerase chain reaction
  • the present invention provides a detection method of prognosis of a neoplasia (or metaplasia) (e.g.. upper gastrointestinal neoplasia, esophageal neoplasia) in a subject known to have or suspected of having neoplasia (or metaplasia),
  • the neoplasia is cancer.
  • Such method comprises assaying for the presence of methylated informative loci in a tissue sample or bodily fluid from the subject. In certain cases, it is expected that detection of methylated informative loci in a tissue sample or bodily fluid from the subject is indicative of an advanced state of cancer (e.g., gastrointestinal cancer such as esophageal cancer).
  • detection of methylated informative loci in a tissue sample or sample from other bodily fluids may be indicative of a cancer that will respond to therapeutic agents that demethylate DNA or reactivate expression of genes located within methylated informative loci
  • the method optionally comprises detecting the presence or absence of somatic mutation(s) in TP53.
  • detection of somatic mutaiion(s) in TP53 in a tissue sample or bodily fluid from the subject is indicative of an advanced state of cancer (e.g., gastrointestinal cancer such as esophageal cancer).
  • detection of somatic mutation(s) in TP53 in a tissue sample or sample from other bodily fluids may be indicative of a cancer that will respond to therapeutic agents that demethylate DNA or reactivate expression of genes located within methylated informative loci.
  • the present invention pro vides a method of monitoring over time the status of neoplasia (e.g., gastrointestinal neoplasia such as esophageal neoplasia) in a subject.
  • neoplasia e.g., gastrointestinal neoplasia such as esophageal neoplasia
  • the neoplasia is a cancer.
  • the neoplasia is a metaplasia such as Barrett's esophagus.
  • the method optionally comprises detecting the presence or absence of somatic mutation(s) in TP53 in combination with detecting the methylation status of any of the informative loci disclosed herein.
  • detection of the appearance of somatic mutation(s) in TPS 3 over time in a tissue .sample or bodily fluid from the subject is indicative of neoplastic progression
  • detection of a disappearance of a somatic mutation(s) in TP53 over time in a tissue sample or sample from other bodily fluids may be indicative of neoplastic regression.
  • the present invention provides a method of e valuating therapy in a subject having cancer or suspected of having neoplasia (e.g., gastrointestinal neoplasia such as esophageal neoplasia),
  • the neoplasia is a cancer.
  • the neoplasia is a metaplasia such as Barrett's esophagus.
  • the method optionally further comprises detecting the presence or absence of somatic mutation(s) in TP53.
  • the present invention also relates to oligonucleotide primer sequences for use in assays (e.g., methylation-sensitive PGR assays or Hpall assays) designed to detect the methyiation status of the informative methylated genomic loci.
  • assays e.g., methylation-sensitive PGR assays or Hpall assays
  • the present invention also provides a method of inhibiting or reducing growth of neoplasia cells (e.g., gastrointestinal neoplasia such as esophageal neoplasia).
  • neoplasia cells e.g., gastrointestinal neoplasia such as esophageal neoplasia.
  • the neoplasia is a cancer.
  • the neoplasia is a metaplasia such as Barrett's esophagus.
  • the disclosure provides for a method of delecting the presence of an esophageal neoplasia or metaplasia in the esophagus in a human subject, comprising: a) obtaining a human sample; and b) assaying the sample for the presence of methyiation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1
  • the disclosure provides for a method of detecting an esophageal neoplasia or metaplasia in the esophagus, comprising: a) obtaining a human sample; and b) assaying said sample for the presence of DNA methyiation by assay in a bisulfite converted DNA for retention of a eytosine base at any of the Y posi tions present in one or more of the nucleotide sequences having at least 90% identical to the sequence of any one or more of SEQ ID NOs: 857-1284, 1713-2140, 3425-3852, 4281-4708, 5927-6321, 6717-711 1, 7559-7584, 761 1 - 7636, 7715-7740, 7767-7792, 7867-7890, 7915-7938, 7991-8004, 8019-8032, 8075-8088, 8103-81 16, 8157-8169, 8183-8195, 82
  • the disclosure provides for method of detecting an esophageal neoplasia, comprising: a) obtaining a human sample; and b) assaying the sample for the presence of methyl atlon within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447- 8818, 9563-9934, 10679-10972, 11561-11662, 1 1867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or complements or fragments thereof, in some embodiments, the disclosure provides for a method of detecting an esophageal neoplasia, comprising: a
  • the sample is assayed for the presence of DNA methylation by assay in a bisulfite converted DNA for retention of a cytosine base at any of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of SEQ ID NOs: 1 1663-
  • the subject has been assayed for having a somatic mutation in TPS 3, in some embodiments, the esophageal neoplasia or metaplasia in the esophagus is selected from the group consisting of Barrett's esophagus (BE), and/or, Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia (HGD), and/or esophageal adenocarcinoma (EAC), In some embodiments, the sample is obtained from a subject suspected of having or is known to have an esophageal neoplasia or metaplasia in the esophagus, in some embodiments, the assay is methylation-specific PGR.
  • BE Barrett's esophagus
  • HCD high grade dysplasia
  • EAC esophageal adenocarcinoma
  • the sample is obtained from a subject suspected of having or is
  • the methods disclosed herein further comprise: a) treating DNA from the sample with a compound that converts a non-methylated cytosine base in the DNA to a different base; b) amplifying a region of the compound converted nucleotide sequence with a forward primer and a reverse primer; and c) analyzing the meihylation patterns of the nucleotide sequences.
  • the methods disclosed herein further comprise: a) treating DNA from the sample with a compound that converts a non-methylated cytosine base in the DNA to a different base: b) amplifying a region of the compound converted nucleotide sequence with a forward primer and a reverse primer; and c) detecting the presence and/or amount of the amplified product.
  • the compound used to treat DNA is a bisulfite compound.
  • the assay comprises using a
  • the methylation-sensitive restriction enzyme is selected from HpaII SmaI, Saell, Eagl, BstUI, and BssHII.
  • the sample is a bodily fluid selected from the group consisting of blood, saliva, spit, an esophageal washing, serum, plasma, a blood-derived fraction, stool, urine, and a colonic effluent.
  • the sample is derived from a tissue, In some embodiments, the sample is a biopsy. In some embodiments, the sample is a brushing.
  • the disclosure provides for a method of monitoring over time an esophageal neoplasia or metaplasia in the esophagus comprising: a) detecting the presence of meihylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - ⁇ 8424, or complements or fragments thereof:, or complements or fragments thereof; from a sample from a subject
  • the disclosure provides for a method of monitoring over time an esophageal neoplasia or metaplasia in the esophagus comprising: a) detecting the meihylation status of DNA by assay in a bisulfite converted DNA for retention of a cytosine base at one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 857-1284, .1713-2140, 3425-3852, 4281-4708, 5927-6321 , 6717-71 1 1 , 7559-7584, 761 1-7636, 7715-7740, 7767- 7792, 7867-7890, 7915-7938, 7991 -8004, 8019-8032, 8075-8088, 8103-8116, 8157-8169, 8183-8195, 8223-8236, 8265-8278,
  • the disclosure provides for a method of monitoring over time an esophageal neoplasia comprising: a) de tecting the presence of methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 1 1561 -11662, 1 1 867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 1258142586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or complements or fragments thereof; from a sample from a subject for a first time: and h) detecting the level of methylation of the nucleotide sequence in a sample from the same subject at a
  • the disclosure provides for a method of monitoring over time an esophageal neoplasia comprising: a) detecting the methylation status of a DNA molecule by assay in a bisulfite converted DNA for retention of a cytosine base of one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 8819-9004, 9191-9376, 9935-10120, 10307- 10492, 10973-1 1 1 19, 1 1267-1 1413, 11663-1 1713, 1 1765-11815, 1 1969-12019, 12071-12121 , 12267-12313, 12361-12407, 12467-12472, 12479-12484, 12503-12508, 12515-12520, 12539-12544, 12551 -12556, 12569-12574, 12587-12592, 12605-12610
  • the disclosure provides for a method of treating a subject having a metaplasia in the esophagus or neoplasia, comprising the step of treating the subject with chemotherapy , radia tion therapy and/or with the resection of the neoplasia or metaplasia; and/or with ablation of the neoplasia or metaplasia; wherein the subject has been determined to have methylation in a sequence tha t is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 ⁇ ⁇ 8306, 8335 - ⁇ 8348, 8405 -
  • the disclosure provides for a method of treating a subject having a metaplasia in the esophagus or neoplasia, comprising the step of treating the subject with chemotherapy, radiation therapy and/or with the resection of the neoplasia or metaplasia; and/or with ablation of the neoplasia or metplasia; wherein the subject has been determined to have DNA methylation by assay in a bisulfite converted DNA for retention of a cytosine base of one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 857-1284, 1713-2140, 3425-3852, 4281-4708, 5927-6321 , 6717-71 1 1 , 7559-7584, 761 1-7636, 7715-7740, 7767- 7792, 7867-7890, 7915-7938
  • the disclosure provides for a method of treating a subject having an esophageal neoplasia, comprising the step of treating the subject with chemotherapy, radiation therapy, ablation, and/or with neoplasia resection; wherein the subject has been determined to have methylation methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 1 1561-11662, 1 1867- 1 1968, 12173-12266, 12455-12466, 12491 -12502, 12527- 12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or complements or fragments thereof.
  • the disclosure provides for
  • the disclosure provides for a bisulfite converted sequence having at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136,
  • the disclosure provides for a panel of bisulfite converted sequences selected from the sequences having at least 90% identity to any one or more of SEQ IX) NOs: 857 -2568, 3425-5136, 5927-7506, 7559 - 7662, 7715 - 7818, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434, and/or fragments thereof, and/or the reverse complements thereof including all unique fragments of these sequences and their reverse complements, in some embodiments, the disclosure provides for a panel of bisulfite converted sequences selected from the sequences having at least 90% identity to any one or more of SEQ IX) NOs: 857 -2568, 3425-5136, 5927-7506, 7559 - 7662
  • the disclosure provides for an oligonucleotide primer that hybridizes to any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136, 5927-7506, 7559 - 7662, 7715 - 7S1 8, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434.
  • the primer is any of the primers having at least 90% identity to any one or more of SEQ ID NOs: 8377-8404 or 8435-8444, or fragments or complements thereof. In some embodiments, the primers are combined as forward and reverse primers for PGR amplification of any of the bisulfite converted sequences having at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136,
  • one of the primers in the primer pairs has a nucleotide sequence that is at least 90% identity to any one or more of SEQ ID NOs: 8377-8404 or 8435-8444, or fragments or complements thereof.
  • the disclosure provides for a panel of primer pairs selected from any of the primer pairs disclosed herein.
  • the panel corresponds to a combination of primer pairs for amplifying any of the combinations of SEQ ID NOs: 8307-8334, 8349- 8376, 8410-8419 or 8425-8434.
  • the disclosure provides for a bisulfite converted sequence having at least 90% identity to any one or more of SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 11560; 1 1663 - 11866, 1 1969 - 12172, 12267 - 12454; 12467 - 12490, .12503 - 12526, 12539 - 12562, 12569 - 12580, 12587 - 12598, 12605 - 12616, 12623 - 12634, 12650 - 12655, and 12659 - 12664, and the reverse complements thereof, including all unique fragments of these sequences and their reverse complements.
  • the disclosure provides for a pane! of bisulfite converted sequences selected from any of these sequences.
  • the disclosure provides for an oligonucleotide primer that hybridizes to any of these sequences.
  • the panel corresponds to any combination of any of the sequences of SEQ ID NOs: 12605-12616, 12623-12634, 12650- 12655, or 12659-12664.
  • the disclosure provides for primers having at least 90% identity to any one or more of SEQ ID NOs: 12635-12646, and 12665-12670, or fragments or complements thereof.
  • the primers are combined as forward and reverse primers for PGR amplification of any of the bisulfite converted sequences having at least 90% identity to any one or more of SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 11560; 1 1663 - 11866, 1 1969 - 12172, 12267 - 12454; 12467 - ⁇ 12490, 12503 - 12526, 12539 - 12562, 12569 - 12580, 12587 - 12598, 12605 - 12616, 12623 - 12634, 12650 - 12655, and 12659 - 12664, and the reverse complements thereof, including all unique fragments of these sequences and their reverse complements, in some embodiments, one of the primers in the primer pairs has a nucleotide sequence that is at least 90% identity to any one or more of SEQ ID NOs: 12635-12646, and 12665
  • the disclosure provides for a panel of primer pairs selected from any of the primer pairs disclosed herein.
  • the panel corresponds to the combination of primer pairs for amplifying any combination of any of the sequences of SEQ ID NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664.
  • the disclosure provides for a method of selecting an individual to undergo a diagnostic procedure to determine the presence of Barrett's esophagus, of Barrett's esophagus with low grade dysplasia, of Barrett's esophagus with high grade dysplasia, or of esophageal adenocarcinoma, by obtaining a biological sample from an individual , and determining the presence in DNA from that sample of DNA methylation present in any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420
  • the method further comprises determining the presence of a somatic mutation in TP53, In some embodiments, DNA methylation is detected by cutting one of the DNA sequences with a methylation sensitive restriction enzyme. In some embodiments, DNA methylation is detected by bisulfite converting DNA from the sample and detecting the presence of any of the bisulfite converted DNA sequences ha ving at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136, 5927-7506, 7559 - 7662, 7715 - 7818, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434, and/or fragments thereof and/or the reverse complements thereof including all unique fragments of these sequences and their reverse complement
  • the bisulfite converted sequences are detected using any of: DNA sequencing, next generation sequencing, methylation specific PGR, methylation specific PGR combined with a fluorogenic hybridization probe, and real time methylation specific PGR.
  • the bisulfite converted sequences are detected using PGR amplification employing a PGR primer or primer pair comprising the nucleotide sequence of any of SEQ ID NOs: 8377-8404 or 8435-8444, in some embodiments, the method further comprises the step of determining the nucleotide sequence of the bisulfite converted sequences.
  • the percent of the target sequence that is methylated in any of the individual target sequences is greater than 1%, or greater than 5%, or greater than 10%.
  • the biological sample is a tissue sample, including a sample of the esophagus.
  • the tissue sample is a biopsy or a brushing, including a biopsy or a brushing of the esophagus.
  • the biological sample is a body fluid, in some embodiments, the body fluid is blood, saliva, spit or an esophageal washing,
  • the disclosure provides for a method of selecting an individual to undergo a diagnostic procedure to determine presence of Barrett's esophagus with low grade dysplasia, of Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma; or to undergo a treatment for for Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia or for esophageal adenocarcinoma; or to undergo enhanced surveillance for the development of Barrett's esophagus with low grade dysplasia, of Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma, by obtaining a biological sample irorn an individual, and determining the presence in DNA from that sample of methylation present in any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818,
  • the disclosure provides for a method of treating a subject having Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia or esophageal adenocarcinoma, comprising the step of treating the subject with chemotherapy, radiation therapy, and/or with resection of an esophageal lesion or with ablation of an esophageal lesion, wherein the subject has been determined to have methylation at any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818, 9563- 9934, 10679-10826, 1 1561 -11662, 1 1867-1 1968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658.
  • the disclosure provides for a method of selecting an individual to undergo enhanced surveillance for the development of Barren's esophagus with high grade dysplasia or of esophageal adenocarcinoma, wherein the subject has been determined to have rnethylation at any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818, 9563-9934, 10679-10826, 1 1561-11662, 11867- 11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658.
  • the methods disiosed herein fuxther comprises determining the presence of a somatic mutation in TP53.
  • DNA rnethylation is detected by cutting one of the DNA sequences with a rnethylation sensitive restriction enzyme, in some embodiments, DNA rnethylation is detected by bisulfite converting DNA from the sample and detecting the presence of any of the bisulfite converted DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 1 1560: 1 1663 - 11866, 11969 - 12172, 12267 - 12454; 12467 - 12490, 12503 - 12526, 12539 - 12562, 12569 - 12580, 12587 - 12598, 12605 - 12616, 12623 - 12634, 12650 - 12655, and 12659 -
  • the bisulfite converted sequences or the presence of unconverted cytosine residues at any of the Y bases of these sequence or panels are detected using any of: DNA sequencing, next generation sequencing, rnethylation specific PGR, rnethylation specific PGR combined with a iluorogemc hybridization probe, and real time rnethylation specific PGR.
  • the bisulfite converted sequences or the presence of unconverted cytosine residues in the bisulfite converted sequences are detected using PGR amplification employing any of the PGR primers or primer pairs disclosed herein.
  • the percent of the target sequence that is methylated in any of the individual target sequences is greater than 1%, or greater than 5%, or greater than 10%.
  • the biological sample is a tissue sample, including a sample of the esophagus.
  • the tissue sample is a biopsy or a brushing, including a biopsy or brushing of the esophagus, i n some embodiments, the biological sample is a body fluid, in some embodiments, the body fluid is blood, saliva, spit or an esophageal washing, in some embodiments, the individual is known to have Barrett's esophagus, In some embodiments, the individual is known to have Barrett's esophagus with low grade dysplasia,
  • the disclosure provides for a method of determining the response of an individual with esophageal cancer to therapy by detection in a body fluid of methylation in any of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 11561-1 1662, 1 1867- 1 1968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581
  • any of the methods disclosed herein further comprise detecting the presence of a somatic mutation in TP53.
  • the DNA methylation is detected by bisulfite converting DNA from a body fluid and detecting the presence of any of the bisulfite converted DNA sequences disclosed herein.
  • the disclosure provides for a method of monitoring over time an esophageal neoplasia comprising: a) detecting in a sample from a subject for a first time (i) the methylation status of one or more of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 -- 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 1 1561-1 1662, 1 1867-11968, 12173-12266, 12455-12466, 12491-125
  • the neoplasia progression is indicated by: (i) the sample taken at the first time lacking methylation in Up] 5-1, Up35-1, Up35-2, Up3, Up27, and Up10, and lacking a somatic mutation in TP53; and (ii) the sample taken at a later time having methylation in any of Up15-1 , Up35 ⁇ I, Up35-2, Up3, Up27, Up10, or having a somatic mutation in TP53.
  • the biological sample is a tissue sample, including a sample of the esophagus.
  • the tissue sample is a biopsy or a brushing, including a biopsy or a brushing of the esophagus.
  • the biological sample is a body fluid, in some embodiments, the body fluid is blood, saliva, spit or an esophageal washing.
  • the disclosure provides for a method of detecting the presence of an esophageal neoplasia in a human subject, comprising: a) obtaining a human sample; and b) detecting the methylation status of one or more of the DNA sequences ha ving at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679- 10826, 1 1561 -11662, 11867-1 1968, 12173-12266, 12455-124
  • the method further comprises the step of detecting the somatic mutation status of TPS 3 from that sample.
  • the TPS 3 somatic mutation is detected at a later time than the detection of the .methylation status of any of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 1 1561-11662, 1 1867-1 1968, 12173-12266, 12455-12466, 12491-12502, 12527
  • the presence of methylation at any one of Up15-1 , Up.35 ⁇ l , Up35-2, Up3, Up27, and Up 10; or the presence of a somatic mutation in TPS 3 is indicative of esophageal adenocarcinoma, of Barrett's esophagus with low grade dysplasia, or of Barrett's esophagus with high grade dysplasia.
  • the presence of methylation at any one of Up15-1 , Up35-I, Up35 ⁇ 2, Up3, Up27, or Up 10, is indicative of esophageal adenocarcinoma, of Barrett's esophagus with low grade dysplasia, or of Barrett's esophagus with, high grade dysplasia.
  • the biological sample is a tissue sample, including a sample of the esophagus, in some embodiments, the tissue sample is a biopsy or a brushing, including a. biopsy or a brushing of the esophagus, in some embodiments, the biological sample is a body fluid, in some embodiments, the body fluid is blood, saliva, spit or an esophageal washing.
  • the disclosure provides for a bisulfite-converted nucleotide sequence comprising the bisulfite-converted nucleotide sequence of any one of the following: Up3, Up10, Up1 5-1, Up1 5-2, Up20-1 , Up20-2, Up20-2, Up27, Up35-1, Up35-2, SqBE2, SqBES, SqBE7, SqBE9, SqBE1 O, SqBE1 l-1, SqBE1 l-2, SqBE13, SqBE14-2, SqBE1S, SqBE16-l, SqBE16-2, SqBE17-l, SqBE1 S, 3qBE22 ⁇ l, SqBE22-2 or SqBE23.
  • the bisulfite-converted nucleotide sequence of claim 98 wherein the sequence comprises the bisulfite-converted nucleotide sequence of any one of the follo wing: Up3, Up10, Up1 5-1 ⁇ Up15 ⁇ 2, Up20-1, Up20-2, Up20-2, Up27, Up35-1, or Up35-2.
  • the bisulfite-converted nucleotide sequence of claim 98 wherein the sequence comprises the bisulfite-converted nucleotide sequence of any one of the following: SqBE2, SqBES, SqBE7, SqBE9, SqBE1O, SqBEI 1-1, SqBE1 1-2, SqBEB, SqBE14-2, SqBE1S, SqBE16-l, SqBE16-2, SqBE174, SqBE1 S, SqBE22-l, SqBE22-2 or SqBE23.
  • the disclosure provides for a bisulfite-converted nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8307-8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8410, 841 1, 8412,
  • the sequence comprises a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8307- 8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371 -8376, 8410, 841 1, 8412, 8414,
  • the sequence comprises a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664.
  • the sequence comprises a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: 12652, 12655, 12661 , and 12664.
  • Figure 1 shows the sensitivity (percentage of samples detected), the specificity
  • an element means one element or more than one element.
  • adenoma is used herein to describe any precancerous neoplasia or benign tumor of epithelial tissue, for example, a precancerous neoplasia of the gastrointestinal tract, pancreas, and/or the bladder.
  • blood-derived fraction refers to a component or components of whole blood.
  • Whole blood comprises a liquid portion (i.e., plasma) and a solid portion (i.e., blood cells).
  • the liquid and solid portions of blood are each comprised of multiple components; e.g., different proteins in plasma or different cell types in the solid portion.
  • One of these components or a mixture of any of these components is a blood-derived fraction as long as such fraction is missing one or more components found in whole blood.
  • esphophagus is intended to encompass the upper portion of the digestive system spanning from the back of the oral cavity, passing downwards through the rear part of the mediastinum, through the diaphragm and into the stomach.
  • esophageal cancer is used herein to refer to any cancerous neoplasia of the esophagus.
  • Barrett's esophagus refers to an abnormal change (metaplasia) in the cells of the lower portion of the esophagus. Barrett's is characterized the finding of intestinal metaplasia in the esophagus,
  • a "brushing" of the esophagus may be obtained using any of the means known in the art.
  • a brushing is obtained by contacting the esophagus with a brush, a sponge, a balloon, or with any other device or substance that contacts the esophagus and obtains an esophageal sample.
  • test compound test compound
  • agent agent
  • molecule molecule
  • peptides nucleic acids
  • carbohydrates small organic molecules
  • natural product extract libraries any other molecules (including, hut not limited to, chemicals, metals, and organometallic compounds)
  • compound-converted DNA refers to DNA that has been treated or reacted with a chemical compound that, converts unmethylated C bases in DNA to a different nucleotide base.
  • a chemical compound that converts unmethylated C bases in DNA to a different nucleotide base.
  • one such compound is sodium bisulfite, which converts unmethylated C to U. If DNA that contains conversion-sensitive cytosme is treated with sodium bisulfite, the compound-converted DNA will contain U in place of C, If the DNA which is treated with sodium bisulfite contains only methylcytosine, the compound-converted DNA will not contain uracil in place of the methylcytosine,
  • de-methylating agent refers agents that restore activity and/or gene expression of target genes silenced by niethylation upon treatment with the agent.
  • examples of such agents include without limitation 5-azacytidme and 5-aza-2'-deoxycytidine.
  • detection is used herein to refer to any process of observing a marker, or a change in a marker (such as for example the change in the niethylation state of the .marker), in a biological sample, whether or not the marker or the change in the marker is actually detected.
  • the act of probing a sample for a marker or a change in the marker is a “detection” even if the marker is determined to be not present or below the level of sensitivity.
  • Detection may be a quantitative, semi-quantitative or non-quantitative
  • differentiated nucleotide sequence refers to a region of a genomic loci that is found to be methylated in a in cancer tissues or cell lines, but. not methylated in the normal tissues or cell lines,
  • neoplasia refers to an abnormal growth of tissue.
  • the term “neoplasia” may be used to refer to cancerous and non-cancerous tumors, as well as to Barrett's esophagus (which may also be referred to herein as a metaplasia) and Barrett's esophagus with dysplasia.
  • the Barrett's esophagus with dysplasia is Barrett's esophagus with high grade dysplasia
  • the Barrett's esophagus with dysplasia is Barrett's esophagus with low grade dysplasia.
  • the neoplasia is a cancer (e.g. , esophageal adenocarcinoma).
  • a cancer e.g. , esophageal adenocarcinoma.
  • Gastrointestinal neoplasia refers to neoplasia of the upper and lower gastrointestinalact.
  • the upper gastrointestinal tract includes the ophagus, stomach, and duodenum; the lower gastrointestinal tract includes the remainder of e small intestine and all of the large intestine.
  • non-neoplastic refers to a subject or particular cell or tissue that is devoid (at least to the limit of detection)f a disease condition, such as a neoplasia.
  • Homology or “identity” or “similarity” refers to sequence similarity between twoeptides or between two nucleic acid molecules. Homology and identity can each beetermined by comparing a position in each sequence which may be aligned for purposes ofomparison. When an equivalent position in the compared sequences is occupied by the samease or amino acid, men the molecules are identical at that position; when the equivalent siteccupied by the same or a similar amino acid residue (e.g., similar in steric and/or electronicature), then the molecules can be referred to as homologous (similar) at that position.
  • homologous similar
  • xpression as a percentage of homology/similarity or identity refers to a function of theumber of identical or similar amino acids at positions shared by the compared sequences.
  • a quence which is "unrelated or “non-homologous" shares, in some embodiments, less than0% identity, and in particular embodiments, less than 25% identity with a sequence of the esent invention. In comparing two sequences, the absence of residues (amino acids orucleic acids) or presence of extra residues also decreases the identity and
  • the term "homology” describes a mathematically based comparison of sequencemilarities which is used to identify genes or proteins with similar functions or motifs.
  • Theucleic acid and protein sequences of the present invention may be used as a "query quence" to perform a search against public databases to, for example, identify other family embers, related sequences or homologs. Such searches can be performed using the
  • Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res.
  • identity means the percentage of identical nucleotide or amino acid residues at corresponding positions in two or more sequences when the sequences are aligned to maximize sequence matching, i.e., taking into account gaps and insertions. Identity can be readily calculated by known methods, including but not limited to those described in
  • Methods to determine identity are designed to give the largest match between the sequences tested. Moreover, methods to determine identity are codified in publicly available computer programs. Computer program methods to determine identity between two sequences include, but are not limited to, the GCG program package (Devereux, J., et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, and FAST A (Altschul, S. F. et al., J. Moke. Biol.
  • BLAST X program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S., et al, NCBI NLM NIH Bethesda, Md.
  • isolated refers to molecules in a form which does not occur in nature.
  • isolated nucleic acid is meant to include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state.
  • MSP methylation-specific PGR
  • methylation-specific PGR refers to a polymerase chain reaction in which amplification of the compound-converted template sequence is performed.
  • Two sets of primers are designed for use in MSP. Each set of primers comprises a forward primer and a reverse primer.
  • One set of primers, called methylation-specific primers will amplify the compound-converted template sequence if C bases in CpG dinucleotides within the DNA are methylated.
  • Another set of primers will amplify the compound-converted template sequences if C bases in CpG dinucleotides within the DNA are not methylated.
  • nucleic acid refers to polynucleotides such as
  • RNA deoxyribonucleic acid
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • the term should also be understood to include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single- stranded (such as sense or antisense) and double-stranded polynucleotides.
  • operably linked when describing the relationship between two DNA regions simply means that they are functionally related to each other.
  • a promoter or other transcriptional regulatory sequence is operably linked to a coding sequence if it controls the transcription of the coding sequence.
  • proteins and “polypeptides” are used interchangeably herein.
  • sample includes any material that is obtained or prepared for detection of a molecular marker or a change in a molecular marker such as for example the methylation state, or any material that is contacted with a detection reagent or detection device for the purpose of detecting a molecular marker or a change in the molecular marker.
  • obtaining a sample includes directly retrieving a sample from a subject to be assayed, or directly retrieving a sample from a subject to be stored and assayed at a later time.
  • a sample may be obtained via a second party. That is, a sample may be obtained via, e.g., shipment, from another individual who has retrieved the sample, or otherwise obtained the sample.
  • a “subject” is any organism of interest, generally a mammalian subject, such as a mouse, and in particular embodiments, a human subject,
  • the term “specifically hybridizes” refers to the ability of a nucleic acid probe/primer of the invention to hybridize to at least 12, 15, 20, 25, 30. 35, 40, 45, 50 or 100 consecutive nucleotides of a target sequence, or a sequence complementary thereto, or naturally occurring mutants thereof, such that it has, in some embodiments, less than 15%, less than 10%, or less than 5% background hybridization to a cellular nucleic acid (e.g., mRNA or genomic DNA) other than the target gene.
  • a variety of hybridization conditions may be used to detect specific hybridization, and the stringency is determined primarily by the wash stage of the hybridization assay. Generally high temperatures and low salt
  • Low stringency hybridization is achieved by washing in, for example, about 2.0 x SSC at 50 °C, and high stringency is achieved with about 0,2 x SSC at 50 °C. Further descriptions of stringency are provided below.
  • the term "substantial sequence identity” means that two peptide sequences, when optimally aligned such as by the programs GAP or BESTFIT using default gap, share at least 90 percent sequence identity, in some embodiments, at least 95 percent sequence identity, or at least 99 percent sequence identity or more, in some embodiments, residue positions which are not identical differ by conservative amino acid substitutions.
  • substitution of amino acids having similar chemical properties such as charge or polarity is not likely to affect the properties of a protein. Examples include glutamine for asparagine or glutamic acid for aspartic acid.
  • an "informative loci" as used herein refers to any of the nucleic acid sequences disclosed herein that may have altered (e.g. , increased) methylation in a sample (e.g., an esophageal tissue sample) from a subject having Barrett's esophagus and/or an esophageal neoplasia as compared to the methylation patterns of the corresponding nucleic acid sequence in a sample from a healthy control subject.
  • a sample e.g., an esophageal tissue sample
  • Barrett's esophagus and/or an esophageal neoplasia as compared to the methylation patterns of the corresponding nucleic acid sequence in a sample from a healthy control subject.
  • Up3 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12563, 12581, 12599, 12617, or fragments or reverse complements thereof.
  • the Up3 sequence refers to a bisuliite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%>, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12569, 12587, 12605 or 12623, or fragments or reverse complements thereof, in some embodiments, the Up3 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12575, 12593, 1261 1 or 12629, or fragments or reverse complements thereof, hi some embodiments, the Up3 sequence may he amplified using primers comprising the sequence of SEQ ID NOs: 12635 and/or 12641
  • Up 10 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%*, 92%, 93%), 94%, 95%, 96%, 97%, 98%, 99% or
  • the Up10 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12570, 12588, 12606 or 12624, or fragments or reverse complements thereof
  • the Up 10 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12576, 12594, 12612 or 12630, or fragments or reverse complements thereof.
  • the Up10 sequence may be
  • Up1 5-1 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%,, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12565, 12583, 12601 or 12619, or fragments or reverse complements thereof.
  • the Up15-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12571, 12589, 12607 or 12625, or fragments or reverse complements thereof.
  • the Up15-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 32577, 12595, 12613 or 12631, or fragments or reverse complements thereof.
  • the Up 15-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12637 and/or 12643, or fragments or reverse complements thereof
  • Up 15-2 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12565, 12583, 12647 or 12656, or fragments or reverse complements thereof
  • the Up] 5-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12571, 12589, 12650 or 12659, or fragments or reverse complements thereof.
  • the Up] 5-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12577, 12595. 12653 or 12662, or fragments or reverse complements thereof.
  • the Up15-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12665 and/or 12668, or fragments or reverse complements thereof.
  • Up20-1 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12566, 12584, 12602 or 12620, or fragments or reverse complements thereof.
  • the Up20-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12572, 12590.
  • the Up20-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12578, 12596,
  • the ⁇ 20-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12638 and/or 12644, or fragments or reverse complements thereof.
  • Up20-2 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12566, 12584, 12648 or 12657, or fragments or reverse complements thereof
  • the Up20-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12572, 12590, 12651 or 12660, or fragments or reverse complements thereof
  • the Up20-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 9
  • Up27 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or
  • the Up27 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12573, 12591, 12609 or 12627, or fragments or reverse complements thereof.
  • the Up27 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12579, 12597,
  • the Up27 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12639 and/or 12645, or fragments or reverse complements thereof,
  • Up35-1 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12568, 12586, 12604 or 12622, or fragments or reverse complements thereof.
  • the Up35-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%», 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12574, 12592, 12610 or 12628, or fragments or reverse complements thereof.
  • the Up35 ⁇ l sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12580, 12598, 12616 or 126.34, or fragments or reverse complements thereof.
  • the Up35-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12640 and/or 12646, or fragments or reverse complements thereof.
  • Up35-2 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12568, 12586, 12649 or 12658, or fragments or reverse complements thereof.
  • the Up35-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12574, 12592, 12652 or 12661 , or fragments or reverse complements thereof.
  • the Up35-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ I D NO: 12580, 12598, 12655 or 12664, or fragments or reverse complements thereof
  • the Up35-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12667 and/or 12670, or fragments or reverse complements thereof.
  • SqBE 2 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8209, 8251 , 8293 or 8335, or fragments or reverse complements thereof.
  • the SqBE 2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ I D NO: 8223, 8265, 8307 or 8349, or fragments or reverse complements thereof.
  • the SqBE 2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%», 85%, 90%, 91%, 92%, 93%, 94%>, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8237, 8279,
  • the SqBE 2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8377 and/or 8391, or fragments or reverse complements thereof.
  • SqBE 5" refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8210, 8252, 8294 or 8336, or fragments or reverse complements thereof.
  • the SqBE 5 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8224, 8266, 8308 or 8350, or fragments or reverse complements thereof.
  • the SqBE 5 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8238, 8280,
  • the SqBE 5 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8378 and/or 8392, or fragments or reverse complements thereof.
  • SqBE 7 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 821 1 , 8253, 8295 or 8337, or fragments or reverse complements thereof.
  • the SqBE 7 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%.
  • the SqBE 7 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8239, 8281, 8323 or 8365, or fragments or reverse complements thereof.
  • the SqBE 7 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8379 and/or 8393, or fragments or reverse complements thereof. sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%. 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8212, 8254, 8296 or 8338, or fragments or reverse complements thereof, hi some embodiments, the SqBE 9 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%.
  • the SqBE 9 sequence refers to a hi sulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%), 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8240, 8282, 8324 or 8366, or fragments or reverse complements thereof.
  • the SqBE 9 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8380 and/or 8394, or fragments or reverse complements thereof.
  • SqBE 10 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8213, 8255, 8297 or 8339, or fragments or reverse complements thereof.
  • the SqBE 10 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8227, 8269, 831 1 or 8353, or fragments or reverse complements thereof.
  • the SqBE S O sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85% ⁇ , 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8241 , 8283, 8325 or 8367, or fragments or reverse complemenis (.hereof. Irs some embodiments, the SqBE 10 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8381 and/or 8395, or fragments or reverse complements thereof
  • SqBE 1 1-1 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8214, 8256, 8298 or 8340, or fragments or reverse complements thereof.
  • the SqBE 1 1 -1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%», 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8228, 8270, 8312 or 8354, or fragments OF reverse complements thereof.
  • the SqBE 1 1 -1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8242, 8284,
  • the SqBE 1 1 -1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs:
  • SqBE 1 1 -2 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8214, 8256, 8405 or 8420, or fragments or reverse complements thereof, in some embodiments, die SqBE- 3 1 -2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8228, 8270, 8410 or 8425, or fragments or reverse complements thereof.
  • the SqBE 1 1-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8242, 8284, 8415 or 8430, or fragments or reverse complements thereof.
  • the SqBE 1 1 -2 sequence may be amplified using primers comprising the sequence of SEQ I D NOs: 8435 and/or 8440, or fragmenis or reverse complements thereof.
  • SqBE 13 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8215, 8257, 8299 or 8341 , or fragments or reverse complements thereof.
  • the SqBE 13 sequence refers to a bisulfite, converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%.
  • the SqBE 13 sequence refers to a. bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8243, 8285,
  • the SqBE 13 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8383 and/or 8397, or fragments or reverse complements thereof,
  • SqBE 14-2 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8216, 8258, 8406 or 8421 , or fragments or reverse complements thereof.
  • the SqBE 14-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8230, 8272, 841 1 or 8426, or fragments or reverse complements thereof.
  • the SqBE 14-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8244, 8286, 8416 or 8431, or fragments or reverse complements thereof.
  • the SqBE 14-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8436 and/or 8441 , or fragments or reverse complements thereof
  • SqBE 15 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8217, 8259, 8301 or 8343, or fragments or reverse complements thereof
  • the SqBE 15 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8231 , 8273, 8315 or 8357, or fragments or reverse complements thereof.
  • the SqBE 15 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8245, 8287, 8329 or 8371 , or fragments or reverse complements thereof.
  • the SqBE 15 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8385 and/or 8399, or fragments or reverse complements thereof.
  • SqBE 16-1 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ I D NO: 8218, 8260, 8302 or 8344, or fragments or reverse complements thereof.
  • the SqBE 16-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8232, 8274, 8316 or 8358, or fragments or reverse complements thereof.
  • die SqBE 16-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8246, 8288, 8330 or 8372, or fragments or reverse complements thereof.
  • the SqBE 16-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8386 and/or 8400, or fragments or reverse complements thereof.
  • SqBE 16-2 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 821 8, 8260, 8407 or 8422, or fragments or reverse complements thereof.
  • the SqBE 16-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8232, 8274, 8412 or 8427, or fragments or reverse complements thereof.
  • the SqBE 16-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8246, 8288, 8417 or 8432, or fragments or reverse complements thereof.
  • the SqBE 16-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8246, 8288, 8417 or 8432, or fragments or reverse complements thereof.
  • the SqBE 16-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a
  • 16- 2 sequence may be amplified using primers comprising the sequence of S EQ ID NOs: 8437 and/or 8442, or fragments or reverse complements thereof.
  • SqBE 17-1 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8219, 8261 , 8303 or 8345, or fragments or reverse complements thereof.
  • the SqBE 17-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8233, 8275, 8317 or 8359, or fragments or reverse complements thereof.
  • the SqBE 17-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8247, 8289,
  • 17- 1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8387 and/or 8401, or fragments or reverse complements thereof,
  • SqBE 18 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8220, 8262, 8304 or 8346, or fragments or reverse complements thereof.
  • the SqBE 18 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8234, 8276, 8318 or 8360, or fragments or reverse complements thereof.
  • the SqBE 18 sequence refers to a bisulfite converted product, of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 9(5%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8248, 8290,
  • the SqBE 18 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8388 and/or 8402, or fragments or reverse complements thereof.
  • SqBE 22-1 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%. 95%, 96%, 97%, 98%,, 99% or 100% identity to the sequence of SEQ ID NO: 8221 , 8263, 8305 or 8347, or fragments or reverse complements thereof.
  • the SqBE 22-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at.
  • the SqBE 22-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ I ' D NO: 8249, 8291 , 8333 or 8375, or fragments or reverse complements thereof, in some embodiments, the SqBE 22-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8389 and/or 8403, or fragments or reverse complements thereof
  • SqBE 22-2 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8221 , 8263, 8409 or 8424, or fragments or reverse complements thereof.
  • the SqBE 22-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8235, 8277, 8414 or 8429, or fragments or reverse complements thereof.
  • the SqBE 22-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8249, 829 i ,
  • the SqBE 22-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8439 and/or 8444, or fragments or reverse complements thereof.
  • SqBE 23 refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%», 95%, 96%, 97%, 98% ? 99% or 100% identity to the sequence of SEQ ID NO: 8222, 8264, 8306 or 8348, or fragments or reverse complements thereof.
  • the SqBE 23 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%>, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8236, 8278, 8320 or 8362, or fragments or reverse complements thereof.
  • the SqBE 23 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8250, 8292, 8334 or 8376, or fragments or reverse complements thereof.
  • the SqBE 23 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8390 and/or 8404, or fragments or reverse complements thereof.
  • any of the nucleotide sequences disclosed herein contain one or more "Y" positions. Cytosine residues that may be methylated or umiiethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a € (if methylated), are designated with a ' ⁇ ," In some embodiments, a parental nucleotide sequence is fully unmethylated if the sequence comprises a T at every Y position following bisulfite conversion.
  • a parental nucleotide sequence is fully methylated if the sequence comprises a C at every Y position following bisulfite conversion, in some embodiments, a parental nucleotide sequence is partially methylated if the sequence comprises at least one C at a Y position and at least one T at a Y position of the sequence following bisulfite conversion. In some embodiments, the bisulfite converted sequences disclosed herein comprise at least one C at a Y position and at least one T at a Y position, i.e., the parental sequence is partially methylated.
  • This disclosure is based at least in part on the recognition that differential methylation of particular genomic loci may be indicative of neoplasia of the upper gastrointestinal tract, e.g., esophagus.
  • the present findings demonstrate that methylation at these genomic loci may be a useful biomarker of neoplasia in the upper gastrointestinal tract.
  • the present findings further demonstrate that the status of methylation at these genomic loci used in combination with the status of somatic mutation(s) in TP53 may be a highly sensitive and specific biomarker of neoplasia in the upper gastrointestinal tract.
  • neoplasias may develop through one of at least three different pathways, termed chromosomal instability, microsatellite instability, and the CpG island methyiator phenotype (CIMP). Although there is some overlap, these pathways tend to present somewhat different biological behavior, By understanding the pathway of tumor
  • This disclosure is based, at least in part on the recognition that certain target genes may be silenced or inactivated by the differential raethylation of CpG islands in the 5 s flanking or promoter regions of the target gene, CpG islands are clusters of cytosine- guanosine residues in a DNA sequence, which are prominently represented in the 5-flanking region or promoter region of about half the genes in our genome.
  • this application is based at least in part on the recognition that differential methylation of particular genomic loci may be indicative of neoplasia of the upper gastrointestinal tract including, but not limited to, esophageal neoplasia,
  • this disclosure is based, at least in part, on the recognition that somatic mutations in TP53 (e.g. , any of the somatic TP53 mutations disclosed herein), in combination with methylation of certain informative loci as disclosed herein, may serve as useful indicators of neoplasia, including esophageal neoplasia (e.g., esophageal adenocarcinoma).
  • somatic mutations in TP53 e.g. , any of the somatic TP53 mutations disclosed herein
  • methylation of certain informative loci as disclosed herein may serve as useful indicators of neoplasia, including esophageal neoplasia (e.g., esophageal adenocarcinoma).
  • the TPS 3 somatic mutation is any of the TP53 mutations disclosed herein, in certain embodiments, the TP53 somatic mutation is any nonsynonymous somatic mutation known in the art, in certain embodiments, the TP53 somatic mutation is any one or more mutation at any one or more amino acid residue corresponding to amino acid residue 72, 105, 108, 1 10, 1 13, 124, 127, 132, 144, 152, 163, 175, 183, 194, 213, 214, 218, 232, 234, 248, 265, 273, 278, 306, 337, 347, or 639 of SEQ ID NO: 16205.
  • the TP53 somatic mutation is any one or more mutation selected from the group consisting of: Leu194Arg, GlylOSAsp, Arg273His, Tyrl 63His, Ile232Thr, Arg213Ter, Arg273His,
  • the TP53 mutation is any one or more mutation at any one or more nucleotide position corresponding to nucleotide position 108, 215, 314, 338, 380, 395, 430, 455, 487, 524, 548, 581 , 637, 639, 641 , 653, 695, 700, 743, 818, 832, 916, 1010, or 1039 of SEQ ID NO: 16206.
  • NM 000546.5 is as follows:
  • Esophageal adenocarcinoma has steadily increased in incidence over recent decades. With an 85% mortality rate this cancer is the most, rapidly increasing cause of cancer mortality from solid tumors in the American population.
  • BE Barrett's esophagus
  • BACs develop in patients without prior symptoms, and current approaches of endoscopic screening of individuals with persistent symptoms of gastroesophageal reflux disease, combined with longitudinal screening of those found to ha ve BE, have accordingly not had significant impact on reducing deaths from EACs.
  • gastrointestinal neoplasia e.g., neoplasia of the upper gastrointestinal tract
  • present systems for screening for esophageal neoplasia are deficient for a variety of reasons, including a lack of specificity and/or sensitivity (e.g., barium swallow) or a high cost and intensive use of medical resources (e.g., upper endoscopy or CT scan).
  • detecting esophageal neoplasia may select the patient to undergo therapies that include, but are not limited, to resection of the neoplasia (via endoscopic resection or surgical resection), ablation of the neoplasia, chemotherapy, or radiation therapy.
  • therapies include, but are not limited, to resection of the neoplasia (via endoscopic resection or surgical resection), ablation of the neoplasia, chemotherapy, or radiation therapy.
  • patients who have received surgical and/or pharmaceutical therapy for esophageal cancer may experience a relapse. It would be advantageous to have an alternative system for determining whether such patients have a recurrent or relapsed neoplasia of the upper gastrointestinal tract.
  • an alternative diagnostic system would facilitate monitoring an increase, decrease or persistence of neoplasia of the upper gastrointestinal tract in a patient known to have such a neoplasia.
  • a patient undergoing chemotherapy may be monitored to assess the effectiveness of the therapy.
  • III Methvlation of fefomiative Loci as .Disease Bioroarkers.
  • the present disclosure relates at least in part to the identification of genomic loci whose altered DNA methylation is indicative of the presence of esophageal neoplasias and/or metaplasias that include Barrett's esophagus (BE) and/or esophageal adenocarcinoma (EAC),
  • the Barrett's esophagus is associated with dysplasia.
  • the dysplasia is high-grade dysplasia, in some embodiments, the dysplasia is low-grade dysplasia, In some embodiments, the methylation patterns of the informative loci as disclosed herein are determined in a sample taken from a subject as described herein and may be used to distinguish between subjects having Barrett's esophagus and subjects having high grade dysplasia and/or low grade dysplasia and/or esophageal adenocarcinoma.
  • any of the nucleotide sequences disclosed herein, or fragments or reverse complements thereof may contain one or more "Y" residues, Cytosine residues that may be methylated or unmethyiaied, and hence may be bisulfite converted to T (if unmethyiaied) or remain as a C (if methylated), are designated with a ' ⁇ .”
  • one or more of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) designates a methylated C.
  • one or more of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) designates an unmethyiaied C.
  • At least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, I I , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) correspond to methylated C residues. In some embodiments, at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues in any of t he sequences disclosed herein (or fragments or reverse complements thereof) correspond to unmethyiaied C residues.
  • At least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) correspond to methylated C residues. In some embodiments, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) are correspond to unmethyiaied C residues, in some embodiments, any of the sequences disclosed herein (or fragments or reverse complements thereof) is bisulfite-converied. In some embodiments, at least 1 , 2, 3, 4.
  • Y residues in any of the bisulfite-converted sequences disclosed herein correspond to C residues.
  • at least. 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues in any of the bisulfite-converted sequences disclosed herein (or fragments or reverse complements thereof) correspond to T residues.
  • an informative loci in a subject is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g. , Barrett's esophagus) or neoplasia ⁇ e.g..
  • Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia
  • esophageal cancer such as esophageal adenocarcinoma
  • the loci is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% methylated
  • a DNA sample from a subject is treated with bisulfite, and the resulting bisulfite sequence
  • the sequence corresponds to any of the nucleotide sequences disclosed herein comprising a "Y" nucleotide.
  • the sequence is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g.
  • esophageal cancer such as esophageal adenocarcinoma
  • a DNA sample from a subject is treated with bisulfite, and the resulting bisulfite sequence corresponds to any of the nucleotide sequences disclosed herein comprising a ' ⁇ " nucleotide, in some embodiments, if at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues of the bisulfite-converted sequence have a C, the sequence is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g.
  • Barrett's esophagus or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma).
  • neoplasia e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia
  • esophageal cancer such as esophageal adenocarcinoma
  • the disclosure provides for informative loci that may be used to assess whether a subject (e.g.
  • a human has or is prone to developing a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal
  • a metaplasia in the esophagus e.g., Barrett's esophagus
  • neoplasia e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia
  • esophageal cancer such as esophageal
  • one or more informative loci may be used for determining whether a subject has or is likely to develop, a metaplasia (e.g., Barrett's esophagus), in some embodiments, one or more informative loci, as defined herein, may be used for determining whether a subject has or is likely to develop, a neoplasia (e.g., Barrett's esophagus with high grade dysplasia, or an esophageal cancer such as esophageal adenocarcinoma).
  • a metaplasia e.g., Barrett's esophagus
  • a neoplasia e.g., Barrett's esophagus with high grade dysplasia, or an esophageal cancer such as esophageal adenocarcinoma
  • one or more informative loci may be used to distinguish between whether a subject has a metaplasia in the esophagus (e.g., Barrett's esophagus) or an esophageal neoplasia (e.g., Barrett's esophagus with high grade dysplasia, or an esophageal cancer such as esophageal adenocarcinoma).
  • a metaplasia in the esophagus e.g., Barrett's esophagus
  • an esophageal neoplasia e.g., Barrett's esophagus with high grade dysplasia, or an esophageal cancer such as esophageal adenocarcinoma
  • the informative loci include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1-428, 2569-2996, 5137-5531 , 7507-7532, 7663-7668, 7819-7842, 7963-7976, 8047-8060, 8131- 8143, 8209-8222, 8293-8306, 8405-8409, 8447-8632, 9563-9748, 10679-10825, 11561- 1161 1 , 1 1867-1 1917, 12173-12219, 12455-12460, 12491-12496, 12527-12532, 12563-12568, 12599-12604, 12647-12649, 12671 -12907, 14093-14329, 15515
  • the informative loci are associated with increased methyl ation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject, in some embodiments, the informative loci that are associated with increased methylation in both Barrett ' s esophagus and esophageal
  • adenocarcinoma samples include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%», 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1 -428, 2569-2996, 5137- 5531, 7507-7532, 7663-7668, 7819-7842, 7963-7976, 8047-8060, 8131-8143, 8209-8222, 8293-8306, or 8405-8409, or fragments or complements thereof.
  • the informative loci thai are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 7963-7976, 8047- 8060, or 8131-8143, or fragments or complements thereof,
  • the informative loci are associated with increased methylation in an esophageal adenocarcinoma sample and/or a Barrett's with low grade or high grade dysplasia as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia.
  • the informative loci that, are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 8447- 8632, 9563-9748, 10679-10825, 1 1561 -1 161 1 , 1 1867-1 1917, 12173-12219, 12455-12460, 12491-12496, or 12527-12532, or fragments or complements thereof, in particular embodiments, the informative loci that are associated with increased methylation in Barrett's with low grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 9
  • the informative loci that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 12455-12460, 12491-12496, or 12527-12532, or fragments or complements thereof, in some embodiments, the informative loci are associated with reduced methylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus.
  • the informative loci thai are associated with reduced methylation in an esophageal adenocarcinoma sample include any ⁇ one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 12671-12908, 14093- 14329, 15515-15537, 15653-15692, 15893-15932, 16133-16137, 16163-16165, 16181 -16183, or 16199, or fragments or complements thereof.
  • the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 16163- 16165, 16181-16183, or 16199, or fragments or complements thereof.
  • the informative loci or amplicon of the informative loci are treated with an agent, such as bisulfite.
  • the informative loci include sequences that have been treated with bisulfite.
  • the disclosure provides for bisulfite control sequences of any of the plus DNA strands disclosed herein.
  • the disclosure provides for bisulfite-treated unme thy Sated sequences of any of the plus DNA strands disclosed herein.
  • the bisulfite-converted plus-strand control DNA sequences include any one or more having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 857-1284, 3425-3852, 5927-6321 , 7559-7584, 7715-7740, 7867-7890, 7991-8004, 8075-8088, 8157-8169, 8223-8236, 8307-8320, 8410-8414, 8819-9004, 9935-10120, 10973- 1 1 1 19, 1 1663-1 1713, 1 1969-12019, 12267-12313, 12467-12472, 12503-12508, or 12539- 12544, 12569-12574, 12605-12610, 12650-12652, or fragments or complements thereof.
  • the bisulfite-converted plus-strand control DNA sequences include any one or more having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 7991 -8004, 8075-8088, 8157-8169, 8223-8236, 8307-8320, 8410-8414, 12467-12472, 12503-12508, or 12539-12544, 12569- 12574, 12605-12610.
  • the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject, in some embodiments, the disclosure provides for bisuifiie-treated mtmefhylated sequences of any of the plus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject.
  • the bisulfite converted sequences of any of the plus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples are selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 857-1284, 3425-3852, 5927-6321, 7559-7584, 7715-7740, 7867- 7890, 7991-8004, 8075-8088, 8157-8169, 8223-8236, 8307-8320, or 8410-8414, or fragments or complements thereof.
  • the bisulfite converted sequences of any of the plus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisulfite- converted methylated plus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of: SEQ ID NOs: 7991-8004, 8075-8088, 8157-8169, 8223-8236, 8307-8320, or 8410-8414, or fragments or complements thereof, in some embodiments, the disclosure provides for bisulfite-treatecl unmethylated sequences of any of the plus DNA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasi
  • the bisulfite converted sequences of any of the plus DNA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more bisulfite-converted methylated plus-strand DNA sequences selected from the group consisting having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 8819-9004, 9935-10120, 10973-1 1 1 19, 1 1663-1 1713, 11969-12019, 12267-12313, 12467-12472, 12503-12508,12539-12544, 12569-12574, 12605- 12610, or 12650-12652, or fragments or complements thereof, in particular embodiments, the bisulfite converted sequences of any of the plus DMA strands that are
  • the informative loci are associated with reduced meihylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus.
  • the disclosure provides for methylated control sequences of the plus DNA strand that are associated with reduced meihylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus.
  • the methylated control sequences of any of the plus DNA strands that are associated with reduced meihylation in an esophageal adenocarcinoma sample include any one or more bisulfite-con verted methylated plus-strand DN A sequences selected from the group consisting having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 13145-13381, 14567-14803, 15561-15583, 15733-15772, 15973-16012, 16143-16147, 16169-16171, 16187-16189 or 16201 , or fragments or complements thereof.
  • meihylation in an esophageal adenocarcinoma sample include any one or more bisulfite- converted methylated plus-strand DNA sequences selected from the group consisting having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 16169-16171 , 16187-16189 or 16201 , or fragments or complements thereof,
  • the informative loci or amplicon of the informative loci are treated with an agent, such as bisulfite
  • the informative loci include sequences that have been treated with bisulfite
  • the informative loci include methylated nucleic acid sequences that have been treated with bisulfite
  • the bisulfite-converted methylated plus-strand DNA sequences have at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1285-1712, 3853-4280, 6322-6716, 7585-7610, 7741-7766, 7891- 7914, 8005-8018, 8089-8102, 8170-8182, 8237-8250, 8321-8334, 8415-841 9, 9005-9190, 10121-10306, 1 1 120-1 1266, 11714-1 1764, 12020-120
  • the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject. In some embodiments, the informative loci that are associated with increased methy!ation in both Barrett's esophagus and esophageal
  • adenocai'cinoma samples include any one or more bisulfite-converted methylated plus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1285-1712, 3853-4280, 6322-6716, 7585-7610, 7741-7766, 7891 -7914, 8005- 801 8, 8089-8102, 8170-8182, 8237-8250, 8321 -8334, or 8415-8419, or fragments or complements thereof.
  • the informative loci that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisulfite-converted methylated plus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to: SEQ ID NOs: 8005-8018, 8089- 8102, 8170-81 82, 8237-8250, 8321-8334, or 8415-8419, or fragments or complements thereof.
  • the informative loci are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia
  • the informative loci that are associated with increased methvlation in an esophageal adenoearcmoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more bisulfite-converted methylated plus-strand DMA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%., 98%, 99% or 100% identity to SEQ ID NOs: 9005-9190, 10121-10306, 11 120-11266, 1 1714-1 1764, 12020-12070, 12314-12360, 124
  • adenocarcinoma sample include any one or more bisulfite-converted methylated plus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 13382-13618, 14804-15040, 15584-15606, 15773-15812, 16013-16052, 16148-16152, 16172-16174, 16190-16192 or 16202.
  • the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 16172-16174, 16190-16192 or 16202, or fragments or complements thereof.
  • the informative loci include sequences associated with any of the minus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 429-856, 2997- 3424, 5532-5926, 7533-7558, 7689-7714, 7843-7866, 7977-7990, 8061 -8074, 8144-8156, 8251-8264, 8335-8348, 8420-8424, 8633-8818, 9749-9934, 10826-10972, 1 1612-1 1662, 11918-1 1968, 12220-12266, 12461-12466, 12497-12502, 12533-12538, 12581-12586, 12617-12622, 12656-12658, 12909-13144, 14330-14566, 15538-15560, 156
  • the informative loci include sequences associated with any of the minus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%», 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 7977-7990, 8061-8074, 8144-8156, 8251 -8264, 8335-8348, 8420-8424, 12461-12466, 12497-12502, 12533-12538, 12581-12586, 12617-12622, 12656-12658, 16166-16168, 16184-16186 or 16200, or fragments or complements thereof.
  • the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject.
  • the informative loci that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include sequences associated with any one or more of the minus strand DN A sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 429-856, 2997-3424, 5532-5926, 7533-7558, 7689- 7714, 7843-7866, 7977-7990, 8061-8074, 8144-8156, 8251-8264, 8335-8348, 8420-8424, or fragments or
  • the informative loci that are associated with increased methylation in both Barrett ' s esophagus and esophageal adenocarcinoma samples include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%., 96 ( 1 ⁇ 2., 97%, 98%), 99% or 100% identity to any of SEQ ID NOs: 8251-8264, 8335-8348, 8420- 8424, or fragments or complements thereof.
  • the informative loci are associated with, increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia
  • the informative ioci that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%., 97%, 98%», 99% or 100% identity to any of SEQ I D NOs: 8633-8818, 9749- 9934, 10826-10972, 1 1612-1 1662, 1 1918-1 1968, 12220-12266, 12461-12466, 12497-12502,
  • the informative loci are associated with reduced methylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus.
  • the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 12909-13144, 14330-14566, 15538-15560, 15693-15732, 15933- 15972, 16138-16142, 16166-16168, 16184-16186 or 16200, or fragments or complements thereof, in particular embodiments, the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of
  • the informative loci or amplicon of the informative Soei are treated with an agent, such as bisulfite.
  • the informative loci include sequences that have been treated with bisulfite.
  • the disclosure provides for bisulfite control sequences of any of the minus DNA strands disclosed herein, in some embodiments, the disclosure provides for hisulfite-treated sequences of any of the minus DNA strands disclosed herein, in some embodiments, the bisulfite-eonverted minus-strand control DNA sequences include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs; 1713-2140, 4281 -4708, 6717-71 1 1 , 761 1 -7636, 7767-7792, 7915-7938, 8019- 8032, 8103-81 16, 8183-8195
  • the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject
  • the disclosure provides for bisulfite-treated sequences of any of the minus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject.
  • the unmethylated sequences of any of the minus DNA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93% s , 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 12479- 12484, 12515-12520, 12551-12556, 12587-12592, 12623-12628, or 12659-12661, or fragments or complements thereof, in some embodiments, the informative loci are associated with reduced methylalion in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus.
  • the disclosure provides for methylated control sequences of the minus DNA strand that are associated with reduced methylalion in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus
  • the methylated control sequences of any of the minus DNA strands that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more bisulfite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87% s , 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 13619-13855, 15041 -15277, 15607-15629, 15813-15852, 16053-16092, 16153-161 57, 16175-1 6177
  • the informative loci or amplicon of the informative loci are treated with an agent, such as bisulfite.
  • the informative loci include sequences that have been treated with bisulfite.
  • the informative loci include methylated nucleic acid sequences that have been treated with bisulfite.
  • the bisuifite-converted methylated minus-strand DNA sequences include any one or more of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%.
  • the bisuifite- converted methylated minus-strand DNA sequences include any one or more of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%>, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 8033-8046, 81 17-81 30, 8196-8208, 8279-8292, 8363-8376, 8430-8434, 12485-12490, 12521-12526, 12557-12562, 12593-12598, 12269-12634, or 12662-12664, or fragments or complements thereof.
  • the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject.
  • the informative loci that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisuifite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 2141 -2568, 4709-5136, 71 ⁇ 2- 7506, 7637-7662, 7793-7818, 7939-7962, 8033-8046, 81 17-8130, 8196-8208, 8279-8292, 8363
  • the i.nfonnaiive loci that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisuifite-converted methylated minus-strand DN A sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to: SEQ ID NOs: 8033-8046, 81 17-8130, 8196-8208, 8279-8292, 8363-8376, or 8430 ⁇ 8434,or fragments or complements thereof.
  • the informative loci are associated with increased meihylation in an esophageal adenocarcinoma sample or a Barrett's with high grade dysplasia sample as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia. In some embodiments, the informative loci that are associated with increased methylation in an esophageal
  • adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more bisuifite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%*, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 9377-9562, 10493- 10678, 11414-1 1560, 11816-1 1866, 12122-12172, 12408-12454, 12485-12490, 12521 -12526, 12557-12562, 12593-12598, 12269-12634, or 12662-12664, or fragments or complements thereof.
  • the informative loci that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences of sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%», 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 12485-12490, 12521 -12526, 12557-12562, 12593-12598, 12269-12634, or 12662-12664, or fragments or complements thereof, in some embodiments, the informative loci are associated with reduced methylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus.
  • the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more bisuifite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 13856-14092, 15278-15514, 15630-15652, 15853-15892, 16093-16132, 16158-16162, 16178-16180. .16196-16198, or 16204, or fragments or complements thereof.
  • the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%. 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 16178-16180, 161 96-16198, or 16204, or fragments or complements thereof.
  • the disclosure provides for a bisuifite-converted nucleotide sequence comprising the bisulfite-conveited nucleotide sequence of any one of the following: Up3, Up10, Up15-1, Up1 5-2, Up20-L Up20-2, Up20-2, Up27, Up35 ⁇ L Up35-2, SqBE2, SqBE5, SqBE7, SqBE9, SqBE1O, SqBE1 l-I , SqBE1 I-2, SqBE13, SqBE14-2, SqBE1S, SqBE16-l, SqBE16-2, SqBE17-l, SqBE18, SqBE22-l , SqBE22-2 or SqBE23.
  • the sequence comprises the bisuS4.000-converted nucleotide sequence of any one of the following: Up3, Up 10, Up15-1. Up15-2, Up20-1 , Up2G-2, Up20-2, Up27, Up35-1, or Up35 ⁇ 2.
  • the sequence comprises the hisuliite-converted nucleotide sequence of any one of the following: SqBE2, SqBE5, SqBE7, SqBE9, SqBE1O, SqBE1 l-1, SqBE1 l ⁇ 2, SqBE13, SqBE14-2, SqBE1 S, SqBE16-l, SqBE16-2, SqBE17 ⁇ l , SqBE1 S, SqBE22-l , SqBE22-2 or SqBE23.
  • the disclosure provides for a panel of any of the sequences disclosed herein, in some embodiments, the panel comprises any of the following
  • the disclosure provides for a method of detecting the methylation status of the sequences in any of the panels disclosed herein. In some embodiments, the disclosure provides for a method of detecting the methylation status of the sequences in any of the panels disclosed herein, and further comprises detecting the mutation status of p53. In particular embodimenis, the disclosure provides for a method of a) detecting the methylation status of a panel comprising the sequences of Up-3 and Up35-2, and b) further detecting the mutation status of TP53,
  • the disclosure provides for a method of detecting the methylation status of any of the loci disclosed herein, and further comprises detecting the methylation status of vimentin.
  • the vimentin methylation is detected in a manner consistent with that described in Li et al, (Li M, et al, (2009) Sensitive digital quantification of D ' NA methylation in clinical samples. Nat Biotechnol 27(91:858-863).
  • the vimentin methylation patterns are determined in a nucleotide sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NO: 16207 or 16208.
  • the methylation patterns are determined in any of the following nucleic acid sequence combinations: a) vimentin and SQBE5; b) vimentin and SQBE7, c) vimentin and SQBE 16, d) vimentin and SQBE 17 or e) vimentin and SQBE18.
  • the disclosure provides for a nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:8209-8222, 8251-8264, 8293-8306, 8335-8348, 8405-8409, 8420-8424, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or fragments and/or reverse complements thereof.
  • the disclosure provides for a nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: 12563-12568, 12581- 12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or fragments and/or reverse complements thereof.
  • the disclosure provides for a nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:8209- 8222, 8251-8264, 8293-8306, 8335-8348, 8405-8409, or 8420-8424, or fragments and/or reverse complements thereof.
  • the disclosure provides for a bisuiilte-con verted nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences; SEQ ID NOs: 8307-8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8411, 8412, 8414, 8416, 8417, 8419, 8426, 8427, 8429, 8431 , 8432, 8434, 1260542616, 12623-12634, 12650- 12655, or 12659-12664, or fragments and/or reverse complements thereof.
  • the sequence comprises a sequence having at least 80%, 85%», 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 1G0% identity to any of the following sequences: SEQ ID NOs; 8307-8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8411 , 8412, 8414, 8416, 8417, 8419, 8426, 8427, 8429, 8431, or 8432, 8434.
  • SEQ ID NOs SEQ ID NOs
  • 8307-8313 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8411 , 8412, 8414, 8416, 8417, 8419, 8426, 8427, 8429, 8431, or 8432, 84
  • the sequence comprises a sequence having at. least 80%, 85%, 90%, 91 %. 92%, 93%, 94%, 95%, 96%», 97%, 98%, 99%. or 100% identity to any of the following sequences: SEQ ID NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664,
  • the disclosure provides for a bisuifite-eonverted nucleotide sequence comprising a sequence having at least 80%, 85%, 90%., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8223-8250, 8265-8292, 12569-12580, or 12587-12598, or fragments and/or reverse complements thereof.
  • the sequence comprises a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences; SEQ ID NOs: 8223-8250 or 8265-8292.
  • the sequence comprises a sequence having at least 80%. 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 12569-12580 or 12587-12598,
  • the sample for use in any of the methods disclosed herein is a tissue sample taken from the subject.
  • the sample is tissue sample from the esophagus, in some embodiments, the sample is a biopsy or a brushing. In some embodiments, the sample is a biopsy or brushing of the esophagus.
  • the sample is a body fluid. In some embodiments, the body fluid is blood, serum, saliva, spit, stool, urine or an esophageal washing.
  • the present disclosure contemplates methods of selecting an individual to undergo a diagnostic procedure to determine the presence of Barrett's esophagus, Barrett's esophagus with dysplasia (e.g., Barrett's esophagus with low-grade or high-grade dysplasia), or of esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA meihylatiors in at least one of any of the sequences disclosed herein.
  • a diagnostic procedure to determine the presence of Barrett's esophagus, Barrett's esophagus with dysplasia (e.g., Barrett's esophagus with low-grade or high-grade dysplasia), or of esophageal adenocarcinoma
  • the disclosure provides for a method of selecting a subj ect for monitoring of esophageal neoplasia, wherein the presence of DNA methylation in at least one of any of the sequences disclosed herein is detected in a sample from the subject, in some embodiments, detection is achieved by any one or more of DNA sequencing, next generation sequencing, methylation specific PGR, methylation specific PGR combined with a fluorogenic hybridization probe, real time methylation specific PGR. or hybridization to an array. In some embodiments, the detection in the sample is indicative that the subject is at high risk of progression to esophageal neoplasia (e.g., esophageal cancer).
  • the subject is monitored by endoscopy.
  • a sample from a subject in which DNA methylation of at least one of any of the sequences disclosed herein is detected is indicative that the subject should be administered a particular treatment.
  • the treatment is selected from the group consisting of endoscopic removal or ablation of an esophageal neoplasia, and/or surgery, radiation, or chemotherapy treatment of esophageal adenocarcinoma.
  • the sequence is any one or more sequence selected from the group consisting of a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 1- 856, 2569-3424, 5137-5926; 7507-7558, 7663-7714, 7819-7866, 7963-7990, 8047-8074, 8131-8156, 8209-8222, 8251-8264, 8293-8306, 8335- 8348, 8405-8409, and 8420-8424, or fragments or complements thereof.
  • the present disclosure also contemplates methods of selecting an individual to undergo a diagnostic procedure to determine presence of Barrett's esophagus with low-grade dysplasia, Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA meihylation in at least one of any of the sequences disclosed herein, in some embodiments, the sequence is any one or more sequence selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: : SEQ ID NOs: 8447 - 8818, 9563 - 9934, 10679 - 10972, 11561 - 11662, 1 1867 - 1 1968, 12173 - 12266, 12455 - 12466, 12491
  • determining the status of methylation of chromosomal loci e.g., Up15-L Up35-1 , Up35-2, Up3, Up27, and Up 10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated.
  • the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample.
  • determining the status of methylation of chromosomal loci e.g. , Up15-1 , Up35-1 , Up35-2, Up3, Up27, and Up 10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample.
  • the method optionally further comprises detecting the presence or absence of a somatic mutation in TP53.
  • the TP53 somatic mutation is any one or more mutation at any one or more amino acid residue corresponding to amino acid residue 72, 105, 108, 1 10, 113, 124, 127, 132, 144, 152, 163, 175, 183, 194, 213, 214, 218, 232, 234, 248, 265, 273, 278, 306, 337, 347, or 639 of SEQ ID NO: 16205.
  • the TP53 somatic mutation is any non-synonomous somatic mutation known in the art.
  • the TP53 somatic mutation is any one or more mutation selected from the group consisting of: Leul94Arg, GlylOSAsp, Arg273His, Tyrl63His, i.!e232Thr, Arg213Ter, Arg273His, Arg248Gln, Argl75His, Argl 1 OdelinsGlnSer, Serl 83Ter, Arg24SGln,
  • the TP53 mutation is any one or more non-synonymous somatic mutation at any one or .more nucleotide position corresponding to nucleotide position 108, 215, 314, 338, 380, 395, 430, 455. 487, 524, 548, 581, 637, 639, 641 , 653, 695, 700, 743, 818, 832, 916, 1010, or 1039 of SEQ ID NO: 16206.
  • the present disclosure also contemplates methods of selecting an individual to undergo a treatment for Barrett's esophagus, Barrett's esophagus with low grade dysplasia, Barrett ' s esophagus with high grade dysplasia or for esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA raethylation in at least one of any of the sequences disclosed herein.'
  • the sequence is any one or more sequence selected from the group consisting of sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926: 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 -
  • the present disclosure further contemplates that the method may further comprise determining the status of somatic rnutation(s) in TP53 in the sample. For example, a method comprising determining the status of methylation of chromosomal loci e.g., Upi 5-1 , Up35-1 , Up35 ⁇ 2, Up3, Up27, and Up1 O in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated.
  • the present disclosure also contemplates methods of selecting an individual to undergo a treatment for Barrett's esophagus, Barrett's esophagus with low-grade dysplasia, Barrett's esophagus with high grade dysplasia or for esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA methylation in at least one of any of the sequences disclosed herein.
  • the sequence is any one or more sequence selected from the group consisting of sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8447 - 8818, 9563 - 9934, 10679 - 10972, 11561 - 1 1662, 1 1867 - 1 1968, 12173 - 12266, 12455 - 12466, 12491 - 12502, 12527 - 12538, 12563 - 12568, 12581 - 12586, 12599 - 12604, 12617 - 12622, 12647 - 12649, or 12656 - 12658, or fragments or complements thereof.
  • the present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP 53 in the sample. For example, a
  • determining the status of methylation of chromosomal loci e.g., Up15-1, Up35 ⁇ l, Up35 ⁇ 2, Up3, Up27, and Up 10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated.
  • the present disclosure also contemplates methods of selecting an individual to undergo enhanced surveillance for the development of Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA methylation in at least one of any of the sequences disclosed herein, in some embodiments, the sequence is any one or more sequence selected from the group consisting of a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 8447 - 8818, 9563 - ⁇ 9934, 10679 - 10972, 1 1561 - 1 1662, 1 1867 - 1 1968, 12173 - 12266, 12455 - 12466, 12491 -
  • the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample.
  • a method comprising determining the status of methylation of chromosomal loci e.g., Up15-1, Up35-1, Up35-2, Up3, Up27, and Up10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated.
  • the present disclosure also contemplates methods of determining the response of an individual with esophageal cancer to therapy by obtaining a biological sample from an individual with esophageal cancer, and determining the presence of methylation in at least one of any of the sequences disclosed herein.
  • the sequence is any one or more sequence selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%o identity to any of the following sequences:: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926: 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 81 56, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, 8447 - 8818, 9563 - 9934, 10679 ⁇ 10972; SEQ ID NOs: 1 1561 - 1 1662, 1 1867 - 1 1968, 12173 - 12266;
  • an increase in levels of methyiation over time is indicative of disease progression and a need for a change in therapy (such as modifying the dosing regime of an exiting therapy, or administering a new therapeutic(s) either alone or in combination with the existing therapy), and an absence of increase in levels of methyiation over time or decrease in levels of inethylation over time is indicative that a change in therapy is not required.
  • the present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample.
  • a method comprising determining the status of methyiation of chromosomal loci e.g., Up1 5-1 , Up35-1 , Up35-2, Up3, Up27, and Up 10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated.
  • the present disclosure also contemplates method of distinguishing EAC and/or low/high grade dysplasia from BE by obtaining a biological sample from an individual, and determining in the sample the presence of DNA methyiation in at least one of any of the sequences disclosed herein.
  • the sequence is any one or more sequence selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96% , 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8447 - 8818, 9563 - 9934, 10679 - 10972; SEQ ID NOs: 1 1561 - 11662, 1 1867 - 1 1968, 12173 - 12266; SEQ ID NOs: 12455 - 12466, 12491 - 12502, 12527 - 12.538, 12563 - 12568, 12581 - 12586, 12599 - 12604, 12617 - 12622, .12647 - 12649, and 12656 - 12658, or fragments or complements thereof.
  • the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample.
  • a method comprising determining the status of methylation of chromosomal loci e.g., Up15-1 , Up35 ⁇ l, Up35-2, Up3, Up27, and Up1 O in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated, in certain embodiments, the absence of methylation at Up] 5-1 , Up35-1, Up35-2, Up3, Up27rada and Up10; and the absence of a somatic mutation in TP53 may be indicative of non-dysplastic Barrel's esophagus.
  • the presence of methylation at any one of Up] 5-1 , Up35-1 , Up35-2. Up3. Up27, and Up10; or the presence of a somatic mutation in TPS 3 may be indicative of esophageal adenocarcinoma or of Barrett's with high grade dysplasia,
  • the present disclosure also contemplates method of distinguishing EAC and/or low/high grade dysplasia from BE by obtaining a biological sample from an individual, and determining in the sample the presence of DNA methylation in at least one of any of the sequences disclosed herein, in some embodiments, the sequence is any one or more sequence selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%>, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8447 - 8818, 9563 - 9934, 10679 - 10972; SEQ ID NOs: 11561 - 11662, 1 1867 - 1 1968, 12173 - 12266; SEQ ID NOs: 12455 - 12466, 12491 - 12502, 12527 - 12538, 12563 - 12568, 12581 - 12586, 12599 -
  • the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample.
  • determining the status of somatic mutation(s) in TP53 in the sample For example, a method comprising determining the status of meihylation of chromosomal loci e.g., Up1 5-1 , Up35-1, Up35-2, Up3, Up27, and Up10 in a sample; and determining the presence or absence of somatic mutation(s) in TPS 3 in the sample is contemplated.
  • the absence of methylation at Up1 S-1 , Up35-1, Up35-2, Up3, Up27, and Up] 0; and the absence of a somatic mutation in TP53 may be indicative of non-dysplastic Barret's esophagus, in certain embodiments, the presence of methylation at any one of Up15-1, Up35-1, Up35-2, Up3, Up27, and Up1O; or the presence of a somatic mutation in TPS 3 may be indicative of esophageal adenocarcinoma.
  • the present disclosure also contemplates a method of monitoring the progression (or regression) of esophageal neoplasias over time, The method involves detecting the methylation status of one or more nucleotide sequences selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, and 8420 - 8424, 8447 - 8818, 9563 - 99
  • neoplasia regression may be indicated by the absence of methylation in the nucleotide sequence taken at a later time and the presence of methyl ati on in the nucleotide sequence taken at the first time
  • neoplasia progression may be indicated by the presence of methylation in the nucleotide sequence taken at a later time and the absence of methylation in the nucleotide sequence taken at the first time.
  • the present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP53 in the samples.
  • neoplastic regression may be indicated by the presence of methylated chromosomal loci e.g..
  • neoplastic progression may be indicated by the presence of unmethylated chromosomal loci e.g., unmethylated Up15-1 , Up35-1 , Up35-2, Up3, Up27, and Up1 O; and the absence of somatic mutation(s) in TPS 3 in a first sample; and the presence of methylated chromosomal loci e.g., methylated Up15-i, Up35-1, Up35-2, Up3, Up27, and/or Up1 O, or the presence of a somatic mutation in TPS 3 in a later sample.
  • unmethylated chromosomal loci e.g., unmethylated Up15-1 , Up35-1 , Up35-2, Up3, Up27, and Up1 O
  • somatic mutation in TPS 3 in a later sample.
  • the present disclosure also provides sequences that will hybridize under highly stringent conditions to the nucleotide sequences of any one or more of SEQ ID NOs: 1 -8444 and 8447-16204, or fragments or complements thereof.
  • appropriate stringency conditions which promote DNA hybridization can be varied.
  • appropriate stringency conditions which promote DNA hybridization can be varied. For example, one could perform the hybridization at 6.0 x sodium chloride/sodium citrate (SSC) at about 45 °C, followed by a wash of 2.0 x SSC at SO °C.
  • SSC sodium chloride/sodium citrate
  • the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50 °C to a high stringency of about. 0,2 x SSC at 50 °C.
  • the temperature in die wash step can be increased from low stringency conditions at room temperature, about 22 °C, to high stringency conditions at about 65 °C, Both temperature and salt may be varied, or
  • the disclosure provides nucleic acids which hybridize under low stringency conditions of 6 x SSC at room temperature followed by a wash at 2 x SSC at room
  • the disclosure also provides the methylated forms of the nucleotide sequences of any one or more of SEQ ID NOs: 1 -8444 and 8447-16204, or fragments thereof, wherein the cytosine bases of the CpG islands present in the sequences are methylated.
  • the nucleotide sequences listed of any one or more of SEQ ID NOs: 1-8444 or 8447-16204 or fragments or complements thereof may be either in the methylated status (e.g.. as seen in neoplasias) or in the unmethylated status (e.g., as seen in normal cells).
  • the nucleotide sequences of the disclosure can be isolated, recombinant, and/or fused with a heterologous nucleotide sequence, or in a DNA library.
  • the present disclosure provides bisulfite-converted nucleotide sequences, for example, bisulfite-converted sequences selected from any of the sequences disclosed herein.
  • the sequence is selected from the group consisting of sequences having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 857 -2568, 3425-
  • the disclosure provides bisulfite-converted sequences selected from the group consisting of sequences having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 1 1560; 11663 - 1 1866, 1 1969 - 12172, 12267 -
  • the disclosure provides bisulfiie-con verted sequences selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 13145 - 14092, 14567 -15514, 15561 - 15652; 15733 - 15892, 15973 -- 16132, 16143 - 16162; 16169 - 16180, 16187 - 16198, and 16201 - 16204, and/or fragments thereof, and/or the reverse complements thereof.
  • a fragment of any of the nucleotide sequences disclosed herein may be of any length, so long as the meihylation status of that nucleotide sequence may be determined.
  • the nucleotide sequence is at least 10, 15, 25, 30, 40, 50, 60, 70, 80, 90, 100, 1 10, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 1200, 1400, 1500, 1700, or 2000 nucleotides in length.
  • the nucleotide sequence is at least 10-2000, 10-1000, 10-500, 10-200, 10-150, 10-100, 50-2000, 50-1000, 50-500, 50-200, 50-150, 50-100, 80-2000, 80-1000, 80-500, 80-150, 80-100, 100- 2000, 100-1000, 100-500, 100-200, or 100-150 nucleotides in length.
  • Such bisulfite-converted nucleotide sequences can be used for detecting the meihylation status, for example, by an MSP reaction or by direct sequencing (e.g., next generation sequencing). These bisulfite-converted sequences are also of use for designing primers for MSP reactions that specifically detect methylated or unmethylated nucleotide sequences following bisulfite conversion, In yet other embodiments, the bisulfite-converted nucleotide sequences of the disclosure also include nucleo tide sequences that will hybridize under highly stringent conditions to any nucleotide sequence of any one or more of SEQ ID NOs: 1-8444 and 8447-16204, or fragments or complements thereof.
  • the application provides methods for producing such bisulfite- converted nucleotide sequences, for example, the application provides methods for treating a nucleotide sequence with a bisulfite agent such that the unmethylated eytosine bases are converted to a different nucleotide base such as a uracil.
  • the application provides oligonucleotide primers for amplifying a region within the nucleic acid sequence of any one or more of SEQ ID NOs: 1-8444 and 8447-16204,
  • a pair of the oligonucleotide primers can be used in a detection assay, such as the Hpall assay.
  • primers used in an MSP reaction can specifically distinguish between methylated and non-methylated DNA.
  • the primers of the disclosure have sufficient length and appropriate sequence so as to provide specific initiation of amplification nucleic acids.
  • Primers of the disclosure are- designed to be "substantially" complementary to each strand of the nucleic acid sequence to be amplified.
  • the primer is selected from the group consisting of sequences having 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%», 97%, 98%, 99%, or 100% identity to any of SEQ ID NOs: 8377-8404, 8435-8446, 12635-12646, and 12665- 12670,
  • the primer comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 .1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, or 35 consecutive nucleotides of any of the primer sequences of SEQ ID NOs: 8377-8404, 8435- 8446, 12635-12646, and 12665-
  • exemplary primers include the sequences of any sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%», or 100% identity to any of SEQ ID NOs: 8377-8404, 8435-8446, 12635-12646, and 12665-12670, or fragments thereof it is understood that any primers that hybridize with the bisulfite-eonverted sequence of any one or more of SEQ ID NOs: 1-8444 and 8447-16204 are included within the scope of this disclosure and is useful in the method of the disclosure for detecting methylated nucleic acid, as described.
  • any primers that would serve to amplify a methyl atiori sensitive restriction site or sites within the differentially methylated region of the informative loci of any of the sequences of SEQ ID NOs: 1-8444 or 8447-16204, or fragments or complements thereof are included within the scope of this disclosure and is useful in the method of the disclosure for detecting nucleic methylated nucleic acid, as described.
  • oligonucleotide primers of the disclosure may be prepared by using any suitable method, such as conventional phosphotriester and phosphodiester methods or automated embodiments thereof.
  • dietbylphosphoramidites are used as starting materials and may be synthesized as described by Beaucage, et al. ⁇ Tetrahedron Letters, 22: 1859-1862, 1981).
  • One method of synthesizing oligonucleotides on a modified solid support is described in U.S. Patent No. 4,458,066,
  • the application provides assays and methods using any of the informative loci, or bisulfite converted methylated or unmethyiated sequences thereof! disclosed herein.
  • the informative loci comprise a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any of the sequences of SEQ ID NOs: 1-428, 2569-2996, 5137-5531 , 7507-7532, 7663- 7668, 7819-7842, 7963-7976, 8047-8060, 8131-8143, 8209-8222, 8293-8306, 8405-8409, 8447-8632, 9563-9748, 1067940825, 1 1561-1161 1 , 1 1867-1 191 7, 12173-12219, 12455-
  • the informative loci are used as molecular markers to distinguish between healthy cells and neoplastic cells (e.g., cancer cells).
  • the informative loci are used as molecular markers to distinguish between healthy cells and esophageal adenocarcinoma cells.
  • the informative loci are used as molecular markers to distinguish between Barrett's esophagus cells and cancer cells.
  • the informative loci are used as molecular markers to distinguish between Barrett's esophagus cells and esophageal adenocarcinoma eel is.
  • the application provides methods and assays using any of the informative loci comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any one or more of SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 1 1561-1 1662, 1 1867-1 1968, 12173-12266, 12455- 12466, 12491-12502, 12527-12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649, OR 12656-12658, or any fragments or complements thereof as markers that distinguish between healthy cells and neoplasia cells.
  • the application provides methods and assays using the informative loci comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any one or more of SEQ ID NOs: 1 -856, 2569-3424, 5137-5926, 7507-7558, 7663-7714, 7819- 7866, 7963-7990, 8047-8047, 8131-8156, 8209-8222, 8251-8264, 8293-8306, 8335-8348, 8405-8409, 8420-8424, 8447-8818, 9563-9934, 10679-10972, 11561-1 1662, 1 1867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-
  • a molecular marker of the invention is a differentially methylated sequence of an informative locus.
  • the application provides assays and methods using the informative loci comprising a sequence having at least.
  • the application provides methods and assays using the informative loci disclosed herein (e.g., chromosomal loci Up1 5-1, Up35-1, Up35-2, Up3, Up27, and Up 10) and the status of somatic mutation(s) in TP53 as markers that distinguish between healthy cells and cells derived from neoplasias of the upper
  • a molecular marker of the invention is a differentially methylated sequence of an informative locus.
  • the application provides assays for detecting differentially methylated nucleotide sequences.
  • a differentially methylated nucleotide sequence, in its methylated state can serve as a target for detection using various methods described herein and the methods that are well within the purview of the skilled artisan in view of the teachings of this application.
  • such methods for detecting methylated nucleotide sequences are based on treatment of genomic DNA with a chemical compound which converts non- methylated C, but not methylated C (i.e., 5mC), to a different nucleotide base.
  • a chemical compound which converts non- methylated C, but not methylated C i.e., 5mC
  • One such compound is sodium bisulfite (also referred to simply as "bisulfite” herein), which converts C, but not 5mC, to U.
  • bisulfite treatment of DNA are known in tire art (Herman, et ah, 1996, Proc Natl Acad Sei USA, 93:9821-6; Herman and Baylin, 1998, Current Protocols in Human Genetics, N, B, A. Dracopoli, ed., John Wiley & Sons, 2:10.6.1-10.6.10; U.S.
  • Patent No. 5,786,146 To illustrate, when a DNA molecule that contains unmethylated C nucleotides is treated with sodium bisulfite to become a compound-converted DNA, the sequence of that DNA is changed (C ⁇ U). Detection of the U in the convened nucleotide sequence is indicative of an unmethyiated C.
  • the different nucleotide base (e.g., U) present in compound-converted nucleotide sequences can subsequently be detected in a variety of ways.
  • the present invention provides a method of detecting U in compound-converted DNA sequences by using "methylation sensitive PCR" (MSP) (see, e.g., Herman, et a!,, 1996, Proc. Natl. Acad Sci. USA, 93:9821-9826; U.S. Patent No. 6,265,171 ; U.S. Patent No, 6,017,704; U.S. Patent No. 6,200,756).
  • MSP methylation sensitive PCR
  • one set of primers (i.e., comprising a forward and a reverse primer) amplifies the compound-converted template sequence if C bases in CpG dinucieotides within the DNA are methylated. This set of primers is called “methylation-specific primers.” Another set of primers amplifies the compound-converted template sequence if C bases in CpG dinucieotides within the 5' flanking sequence are not methylated. This set of primers is called "unmeihylation-specific primers.”
  • the reactions use the compound-converted DNA from a sample in a subject.
  • methylation- specific primers are used, in the case where C within CpG dinucieotides of the target sequence of the DNA are methylated, the methylaiion- specific primers will amplify the compound-converted template sequence in the presence of a polymerase and an MS P product will be produced. If C within CpG dinucieotides of the target sequence of the DNA is not methylated, the methylaiion-specific primers will not amplify the compound-converted template sequence in the presence of a polymerase and an MSP product will not be produced.
  • any of the bisulfite converted methylated sequences disclosed herein is used as a marker for a particular indication.
  • the reaction uses the compound-converted DNA from a sample in a subject and
  • unmethylation-speeific primers are used.
  • the unmethylation specific primers will amplify the compound-converted template sequence in the presence of a polymerase and an MSP product will be produced.
  • the unmethylation-specific primers will not amplify the compound- converted template sequence in the presence of a polymerase and an MSP product will not be produced.
  • a biologic sample will often contain a mixture of both neoplastic cells that give rise to a signal with methylation specific primers, and normal cellular elements that give rise to a signal with unmethylation-speeific primers.
  • the unmethylation specific signal is often of use as a control reaction, but does not in this instance imply the absence of neoplasia as indicated by the positive signal derived from reactions using the methylation specific primers.
  • any of the bisulfite converted unmethylated sequences disclosed herein are used as controls.
  • the unmethylated control sequences are any of the bisulfite converted sequences of SEQ ID NOs: 857-1284, 3425-3852, 5927-6321, 7559-7584, 7715-7740, 7867-7890, 7991-8004, 8075-8088, 8157- 8169, 8223-8236, 8307-8320, 8410-8414, 8819-9004, 9935-10120, 10973-1 1119, 1 1663- 1 1713, 1 1969-12019, 12267-12313, 12467-12472, 12503-12508, or 12539-12544, 12569- 12574, 12605-12610, 12650-12652,1713-2140, 4281-4708, 6717-71 1 1, 761 1-7636, 7767- 7792, 7915-7938, 8019-8032, 8103-81 16, 8183-8195, 8265-8278, 8349-8362, 8425
  • Primers for a MSP reaction are derived from the compound-converted template sequence.
  • "derived from” means that the sequences of the primers are chosen such that the primers amplify the compound-converted template sequence in a MSP reaction.
  • Each primer comprises a single-stranded DNA fragment which is at least 8 nucleotides in length. In some embodiments, the primers are less than 50 nucleotides in length, or in some embodiments, from 15 to 35 nucleotides in length.
  • the compound -converted template sequence can be either the Watson strand or the Crick strand of the double-stranded DNA that is treated with sodium bisulfite
  • the sequences of the primers is dependent upon whether the Watson or Crick compound-converted template sequence is chosen to be amplified in the MSP. Either the Watson or Crick strand can be chosen to he amplified.
  • the compound-converted template sequence, and therefore the product of the MSP reaction is, in some embodiments, between 20 to 3000 nucleotides in length, in other embodiments, the product of the MSP reaction is between 50 to 1000 nucleotides in length, in other embodiments, the product of the MSP reaction is between 50 to 500 nucleotides in length. In other embodiments, the product of the MSP reaction is between 80-150 nucleotides in length, in some embodiments, the product of the MSP reaction is at least 20, 30, 40, 50, 60, 70. 80, 90, 100, 1 10, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240 or 250 nucleotides in length. In some embodiments, the meihylation-specific primers result in an MSP product of a different length than the MSP product produced by the unmethylation- specific primers.
  • a variety of methods can be used to determine if an MSP product has been produced in a reaction assay.
  • One way to determine if an MSP product has been produced in the reaction is to analyze a portion of the reaction by agarose gel electrophoresis. For example, a horizontal agarose gel of from 0.6 to 2.0% agarose is made and a portion of the MSP reaction mixture is electrophoresed through the agarose gel. After electrophoresis, the agarose gel is stained with ethidium bromide. MSP products are visible when the gel is viewed during illumination with ultraviolet light. By comparison to standardized size markers, it is determined if the MSP product is of the correct expected size.
  • Real-time PGR utilizes a thermal cycler (i.e., an instrument thai provides the temperature changes necessary for the PGR reaction to occur) that incorporates a fiuorimeter (i.e. an instrument that measures
  • the real-time PGR reaction mixture also contains a reagent whose
  • incorporation into a product can be quantified and whose quantification is indicative of copy number of that sequence in the template.
  • One such reagent is a fluorescent dye, called SYBR Green I (Molecular Probes, Inc.; Eugene, Oregon) that preferentially binds double-siranded DNA and whose fluorescence is greatly enhanced by binding of double-stranded DNA.
  • a PGR reaction When a PGR reaction is performed in the presence of SYBR Green I, resulting DNA products bind SYBR Green I and fluorescence. The fluorescence is detected and quantified by the fiuorimeter. Such technique is particularly useful for quantification of the amount of the product in the PGR reaction. Additionally, the product from the PGR reaction may be quantitated in "real-time PGR" by the use of a variety of probes thai hybridize to the product including TaqMan probes and molecular beacons. Quantitation may be on an absolute basis, or may be relative to a constitutively methylated DNA standard, or may be relative to an unmethylated DNA standard. In one instance the ratio of methylated derived product to unmethylated derived product may be constructed.
  • Methods for detecting methylation of the DNA are not limited to MSP, and may cover any assay for detecting DNA methylation.
  • Another example method of detecting methylation of the DNA is by using "methylation-sensitive" restriction endonucleases. Such methods comprise treating the genomic DNA isolated from a subject with a methylation-sensitive restriction endonuclease and then using the restriction endonucSease-treated DNA as a template in a PGR reaction.
  • methylation-sensitive restriction endonucleases recognize and cleave a specific sequence within the DNA if C bases within the recognition sequence are not methylated, If C bases within the recognition sequence of the restriction endonuclease are methylated, the DNA will not be cleaved.
  • Examples of such methylation-sensitive restriction endonucleases include, but are not limited to Hpai l, SmaL SacII, Eagl, BstUi, and BssHIL
  • a recognition sequence for a methylation-sensitive restriction endonuclease is located within the template DNA, at a position between the forward and reverse primers used for the PGR reaction.
  • methylation of C bases can be determined by the absence or presence of a PGR product (Kane, et a!,, 1997, Cancer Res, 57:808-1 1). No sodium bisulfite is used in this technique.
  • modified MSP utilizes primers that are designed and chosen such that products of the MSP reaction are susceptible to digestion by restriction endonucleases, depending upon whether the compound-converted template sequence contains CpG dinueleotides or UpG ciinucleotides.
  • Yet other methods for detecting methylation of the DNA include the MS-SnuPE methods.
  • This method uses compound-converted DNA as a template in a primer extension reaction wherein the primers used produce a product, dependent upon whether the compound- converted template contains CpG dinueleotides or UpG dinueleotides (see e.g., Gonzalgo, et al confuse 1997, Nucleic Acids Res,, 25:2529-31).
  • COBRA i.e., combined bisulfite restriction analysis
  • methylation-sensitive restriction endonucleases recognize and cleave a specific sequence within the DNA if C bases within the recognition sequence are methylated. If C bases within the recognition sequence of the restriction endonuclease are not methylated, the DNA will not be cleaved.
  • the method utilizes methylation-sensitive restriction endonucleases.
  • Another exemplary method of detecting rnethylation of DNA requires hybridization of a compound converted DNA to an-ays that include probes that hybridize to sequences derived from a methylated template.
  • Another exemplary method of detecting rnethylation of DNA includes precipitation of methylated DNA with antibodies that bind meihylated DNA or with other proteins that hind methylated DNA, and then detection of DNA sequences in the precipitate.
  • the detection of DNA could be done by PGR based methods, by hybridization to arrays, or by other methods known to those skilled in the art.
  • Another exemplary method of detecting methylated DNA is bisulfite sequencing that involves amplification of a target region of bisulfite converted DNA using rnethylation indifferent PGR primers that amplify converted DNAs derived from both methylated and unmethyiated templates.
  • the rnethylation indifferent primers are often designed to be both rnethylation indifferent and bisulfite specific, i.e.
  • the amplified DN As then may be characterized by Next Generation Sequencing methods that allow each cytosine base in the original template to be assessed within each DNA sequence read for the presence of rnethylation (retention of cytosine) or the absence of rnethylation (conversion to thymidine).
  • the percent of rnethylation at each cytosine base in the original template can then be calculated by the percent of DNA reads in which the cytosine is preserved as cytosine versus is converted to thymidine.
  • the percent of rnethylation across a region of interest can be assessed by determining a rule for assessing the region as methylated or unmethyiated in an individual DNA read (i.e. determining a cutoff for methylation in the region that will categorize the region as "methylated"), and then determining the percent of DNA reads in which the region qualifies as methylated.
  • the disclosure provides methods that involve directly sequencing the product resulting from an MSP reaction to determine if the compound- converted template sequence contains CpG dinucleotides or UpG dinucleotides.
  • Molecular biology techniques such as directly sequencing a PGR product are well known in the art.
  • methylation of DNA may be measured as a percentage of total DNA.
  • High levels of methylation may be 1-100% methylation, for example, 1%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% methylation.
  • Low levels of methylation may be 0%-0.99% methylation, for example, 0%, 0.1 %, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%.
  • At least some normal tissues, for example, normal esophagus samples, may not have any detectable methylation.
  • any one of the informative loci disclosed herein may include nucleotide sequences that encode polypeptides that, for example, may function as a tumor suppressor gene. Accordingly, the application further provides methods for detecting such polypeptides in cell samples, in some embodiments, the disclosure provides detection methods by assaying such polypeptides so as to determine whether a patient has or does not have a disease condition. Further, such a disease condiiion may be characterized by decreased levels of such polypeptides. In certain embodiments, the disclosure provides methods for determining whether a patient is or is not likely to have cancer by detecting such polypeptides. In further embodiments, the disclosure provides methods for determining whether the patient is having a relapse or determining whether a patient's cancer is responding to treatment.
  • a protein is detected with an antibody.
  • an antibody-based detection assay involves bringing the sample and the antibody into contact so that the antibody has an opportunity to bind to proteins having the corresponding epitope.
  • an antibody-based, detection assay also typically involves a system for detecting the presence of antibody-epitope complexes, thereby achieving a detection of the presence of the proteins having the corresponding epitope.
  • Antibodies may be used in a variety of detection techniques, including enzyme-linked immunosorbent assays (ELISAs). immunoprecipitations, Western blots. Antibody-independent techniques for identifying a protein may also be employed. For example, mass spectroscopy, particularly coupled with liquid chromatography, permits detection and quantification of large numbers of proteins in a sample. Two-dimensional gel electrophoresis may also be used to identify proteins, and may be coupled with mass spectroscopy or other detection techniques, such as N-terminal protein sequencing, RNA aptamers with specific binding for the protein of interest may also be generated and used as a detection reagent. Samples should generally be prepared in a. manner that is consistent with the detection system to be employed. For example, a sample to be used in a protein detection system should generally be prepared in the absence of proteases.
  • a sample to be used in a nucleic acid detection system should generally be prepared in the absence of nucleases.
  • a sample for use in an antibody-based detection system will not be subjected to substantial preparatory steps.
  • urine may be used directly, as may saliva and blood, although blood will, in certain embodiments, be separated into fractions such as plasma and serum.
  • a method of the disclosure comprises detecting the presence of an informative loci-expressed nucleic acid, such as an mRNA, in a sample,
  • the method involves obtaining a quantitative measure of the informative loci-expressed nucleic acid in the sample, in view of this specification.
  • Nucleic acid detection systems generally involve preparing a purified nucleic acid fraction of a sample, and subjecting the sample to a direct detection assay or an amplification process followed by a detection assay.
  • Amplification may be achieved, for example, by polymerase chain reaction (PGR), reverse transcriptase (RT) and coupled RT- PCR.
  • PGR polymerase chain reaction
  • RT reverse transcriptase
  • Detection of a nucleic acid is generally accomplished by probing the purified nucleic acid fraction with a probe that hybridizes to the nucleic acid of interest, and in many instances, detection involves an amplification as well, Northern blots, dot blots, microarrays, quantitative PGR, and quantitative RT-PCR are all well known methods for detecting a nucleic acid in a sample.
  • the disclosure provides nucleic acid probes that bind specifically to an informative loci nucleic acid.
  • probes may be labeled with, for example, a fluorescent moiety, a radionuclide, an enzyme or an affinity tag such as a biotin moiety.
  • the TaqMan® system employs nucleic acid probes that are labeled in such a way that the fluorescent signal is quenched when the probe is free in solution and bright when the probe is incorporated into a larger nucleic acid.
  • immunoscintigraphy using monoclonal antibodies directed at the informative Iocs may be used to detect and/or diagnose a cancer.
  • monoclonal antibodies against the informative loci labeled with "Technetium, 11 'indium, 325 Iodine- may be effectively used for such imaging.
  • the amount of radioisotope to be administered is dependent upon the radioisotope.
  • Those having ordinary skill in the art can readily formulate the amount of the imaging agent to be administered based upon the specific activity and energy of a gi ven radionuclide used as the active moiety.
  • compositions according to the present invention useful as imaging agents comprising a targeting moiety conjugated to a radioactive moiety comprise 0.1-100 millicuries, in some embodiments 1 -10 millicuries, in some embodiments 2-5 millicuries, in some embodiments 1 -5 millicuries.
  • TP53 gene or protein sequence is determined and any change in the determined sequence relative to the wildtype sequence is detected.
  • the TP53 gene sequence is determined by PGR, RT-PCR, Northern Blot, Southern Blot, and/or in situ hybridization.
  • Another way to determine if TP53 contains a somatic mutation may involve the use of an antibody- based detection assay (e.g. ELISA, immunohistocheniistry, and/or Western Blot).
  • the antibody-based detection assay utilizes an antibody that binds to a mutant TP53 protein with a tighter affinity than it binds to a wildtype TP53 protein.
  • an antibody that binds to a mutant TP53 protein with a tighter affinity than it binds to a wildtype TP53 protein The skilled artisan will also readily appreciate methods of determining somatic mutations in TP53 based on the disclosures of US5843654, US5620848, EP0390323 and US5527676, ail of which are herein incorporated by reference in their entirely.
  • the disclosure provides for a device useful for detecting the methylaiion status of any of the informative loci, or fragments or complements thereof, disclosed herein, in some embodiments, the disclosure provides for a kit comprising components useful for detecting the methylaiion status of the informative loci, or fragments, or complements thereof, disclosed herein.
  • the kit comprises a swallowable balloon for collecting an esophageal sample from the subject.
  • the kit comprises any of the swallowable balloon devices disclosed in WO 2015/089422, which is incorporated herein in its entirety.
  • the present disclosure provides drug screening assays for identifying test compounds which potentiate the tumor suppressor function of polypeptides encoded by sequences located in the informative loci disclosed herein.
  • the assays detect test compounds which potentiate the expression level of polypeptides encoded by sequences located in the informative loci disclosed herein.
  • the assays detect test compounds which inhibit the methylaiion of DNA.
  • drag screening assays can be generated which detect test compounds on the basis of their ability to interfere with stability or function of polypeptides encoded by sequences located in the informative loci disclosed herein.
  • assay formats may be used and, in light of the present disclosure, those not expressly described herein will nevertheless be considered to be within the purview of ordinary skill in the art.
  • Assay fomiats can approximate such conditions as protein expression level, methylaiion status of nucleotide sequences, tumor suppressing activity, and may be generated in many different forms.
  • the disclosure provides assays including both cell-free systems and cell-based assays which utilize intact cells.
  • Compounds to be tested can be produced, for example, by bacteria, yeast or other organisms (e.g., natural products), produced chemically (e.g., small molecules, including peptidomimetics), or produced recombinantly.
  • the efficacy of the compound can be assessed by generating dose response curves from data obtained using various concentrations of the test compound.
  • a control assay can also be performed to provide a baseline for comparison. In the control assay, the formation of complexes is quantiiated in the absence of the test compound.
  • Assays of the present invention which are performed in cell-free systems, such as may he developed with purified or semi-purified proteins or with lysates, are often preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound.
  • the effects of cellular toxicity and/or bioavailability of the test compound can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an alteration of binding affinity with other proteins or changes in enzymatic properties of the molecular target,
  • test compounds identified from these assays may be used in a therapeutic method of treating cancer.
  • Still another aspect of the application provides transgenic non-human animals which express a gene located within any one of the informative loci disclosed herein, or which have had one or more of such genomic gene(s) disrupted in at. least one of the tissue or cell-types of the animal.
  • the application provides an animal model for cancer, which has a mis-expressed allele of a gene located within any one of the informative loci disclosed herein.
  • Such a mouse model can then be used to study disorders arising from mis-expression of genes located within any one of the informative loci disclosed herein.
  • target sequence refers to a nucleotide sequence that is genetically recombined by a recombinase, The target sequence is flanked by recombinase recognition sequences arid is generally either excised or inverted in cel ls expressing recombinase activity.
  • Recombinase catalyzed recombination events can be designed such that recombination of the target sequence results in either the activation or repression of expression of the polypeptides.
  • excision of a target sequence which interferes with the expression of a recombinant gene can he designed to acti vate expression of that gene.
  • the transgene can be made wherein the coding sequence of the gene is flanked recombinase recognition sequences and is initially transfeeted into cells in a 3' to 5' orientation with respect to the promoter element, in such an instance, inversion of the target sequence wall reorient the subject gene by placing the 5' end of the coding sequence in an orientation with respect to the promoter element, which aHow ? for promoter driven transcriptional activation.
  • Saccharomyces cerevislae (O'Gorraan et aL, (1991 ) Science 251 : 1351-1355; PCX publication WO 92/15694) can be used to generate in vivo site-specific genetic recombination systems.
  • Cre recombinase catalyzes the site-specific recombination of an intervening target sequence located between loxP sequences.
  • loxP sequences are 34 base pair nucleotide repeat sequences to which the Cre recombinase binds and are required for Cre recombinase mediated genetic recombination.
  • the orientation of loxP sequences determines whether the intervening target sequence is excised or inverted when Cre recombinase is present (Abremski et al, (1984) J Biol. Chem. 259:1509-1514); catalyzing the excision of the target sequence when the loxP sequences are oriented as direct repeats and catalyzes inversion of the target sequence when loxP sequences are oriented as inverted repeats.
  • the invention relates to a subject suspected of having or 1ms: a cancer, a metaplasia, or a neoplasia of the upper gastrointestinal tract (e.g., esophageal cancer).
  • a subject may be undergoing routine screening and may not necessarily be suspected of having such metaplasia or neoplasia.
  • the subject is a human subject, and the neoplasia is a neoplasia of the upper gastrointestinal tract, such as the esophagus, in some embodiments, the subject is a human subject, and the metaplasia is Barrett's esophagus.
  • detecting the methylation status of the nucleotide sequences by MSP can be used by itself, or in
  • TPS 3 a somatic mutation status of TPS 3 or other various assays, to improve the sensitivity and/or specificity for detecting, e.g., a neoplasia of the upper or lower gastrointestinal tract, in some embodiments, such detection is made at an early stage in the development of cancer, so that treatment is more likely to be effective.
  • an informative loci in a subject is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma) if the loci is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% methylated.
  • a DNA sample from a subject is treated with bisulfite, and the resulting bisulfite sequence
  • the sequence is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma).
  • a metaplasia in the esophagus e.g., Barrett's esophagus
  • neoplasia e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia
  • esophageal cancer such as esophageal adenocarcinoma
  • a DNA sample from a subject is treated with bisulfite, and die resulting bisulfite sequence corresponds to any of the nucleotide sequences disclosed herein comprising a "Y" nucleotide.
  • the sequence is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g.
  • Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia
  • esophageal cancer such as esophageal adenocarcinoma
  • a subject is determined to be prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma) if a certain number of "Y"
  • nucleotides in a bisulfite converted sequence are cytosines.
  • the certain number is at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues of the bisuifite-converted sequence.
  • the certain number is least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues of the bisuifite-converted sequence.
  • a subject is determined to be prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., esophageal cancer such as esophageal adenocarcinoma) (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) if a certain percentage of DNA molecules from a sample from a subject are determined to be "methylated," as defined herein.
  • a metaplasia in the esophagus e.g., Barrett's esophagus
  • neoplasia e.g., esophageal cancer such as esophageal adenocarcinoma
  • Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia
  • the certain percentage of DNA molecules is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the DNA molecules from the sample are determined to be "methylated.” In some embodiments, the percentage of methylated DNA molecules is determined using next- generation sequencing. Exemplary cut-offs of DNA methylation and DN A molecule percentages may be found in the Examples section provided herein.
  • assaying of a marker in a sample from a subject not known to have, e.g., a metaplasia or neoplasia of the upper gastrointestinal tract can be prognostic for the subject (i.e., indicating the probable course of the disease).
  • subjects having a predisposition to develop a metaplasia or neoplasia of the upper gastrointestinal tract may possess methylated nucleotide sequences.
  • Assaying of methylated informative loci in a sample from subjects either by itself, or in combination with assaying for somatic mutation(s) in TP53 can also be used to select a particular therapy or therapies which are particularly effective against, e.g., a neoplasia of the upper gastrointestinal tract in the subject, or to exclude therapies that are not likely to be effective.
  • Assaying of methylated informative loci in samples from subjects that are known to have, or io have had, a cancer is also useful.
  • the present methods can be used to identify whether therapy is effective or not for certain subjects.
  • One or more samples are taken from the same subject prior to and following therapy, and assayed for any one or more of the informative loci markers either by itself or themselves, or in combination with assaying for somatic mutation(s) in TP53.
  • a finding that an informative locus is methylated in the sample taken prior to therapy and absent (or at a lower level) after therapy may indicate that the therapy is effective and need not be altered.
  • the informative locus is methylated in the sample taken before therapy and in the sample taken after therapy, it may be desirable to alter the therapy to increase the likelihood that the cancer will be reduced in the subject.
  • the present method may obviate the need to perform more invasive procedures which are used to determine a patient's response to therapy.
  • the assays of the invention are useful for monitoring over time the status of a cancer associated with silencing of genes located in the informative loci disclosed herein.
  • a cancer for subjects in whom a cancer is progressing, there can be no DNA methyiation in some or all samples when the first sample is taken and then appear in one or more samples when the second sample is taken.
  • DNA methyiation may be present in one or a number of samples when the first sample is taken and then be absent in some or all of these samples when the second sample is taken.
  • Samples for use with the methods described herein may be essentially any biological material of interest.
  • a sample may be a bodily fluid sample from a subject, a tissue sample from a subject, a solid or semi-solid sample from a subject, a primary cell culture or tissue culture of materials derived from a subject, cells from a cell line, or medium or other extracellular material from a cell or tissue culture, or a xenograft (meaning a sample of a cancer from a first subject, e.g., a human, that has been cultured in a second subject, e.g., an immuno-compromised mouse).
  • a xenograft meaning a sample of a cancer from a first subject, e.g., a human, that has been cultured in a second subject, e.g., an immuno-compromised mouse.
  • sample as used herein is intended to encompass both a biological material obtained directly from a subject (which may be described as the primary sample) as well as any manipulated forms or portions of a primary sample.
  • a sample may also be obtained by contacting a biological material with an exogenous liquid, resulting in the production of a lavage liquid containing some portion of the contacted biological material.
  • sample is intended to encompass the primary sample after it has been mixed with one or more additive, such as preservatives, chelators, anti-clotting factors, etc.
  • a bodily fluid sample is a blood sample.
  • the term is a blood sample.
  • sample is intended to encompass not only the blood as obtained directly from the patient but also fractions of the blood, such as plasma, serum, cell fractions (e.g., platelets, erythrocytes, and lymphocytes), protein preparations, nucleic acid preparations, etc.
  • the bodily .fluid may be derived from the stomach, for example, gastric secretions, acid reflux, or vomit, in other embodiments, the bodily fluid may be a fluid secreted by the pancreas or bladder.
  • the body fluid may be saliva, spit, or an esophageal washing.
  • a tissue sample is a biopsy taken from the mucosa of the
  • tissue sample is the brushings from, e.g., the esophagus of a subject.
  • a subject is in some embodiments a human subject, bat it is expected that the molecular markers disclosed herein, and particularly their homologs from other animals, are of similar utility in other animals.
  • a biomarker described herein e.g., DNA raethylation or protein expression level
  • sample is intended to encompass that portion of biological material that is contacted with a reagent or device involved in the detection process.
  • DNA which is used as the template in an MSP reaction is obtained from a bodily fluid sample.
  • bodily fluids are blood, saliva, spit or an esophageal washing. Other body fluids can also be used. Because they can be easily obtained from a subject and can be used to screen for multiple diseases, blood or blood-derived fractions are especially useful.
  • Blood-derived fractions can comprise blood, serum, plasma, or other fractions.
  • a cellular fraction can be prepared as a "buffy coat" (i.e., leukocyte-enriched blood portion) by centrifuging 5 ml of whole blood for 10 min at 800 times gravity at room temperature. Red blood cells sedknent most rapidly and are present as the bottom-most fraction in the centrifuge tube.
  • the buffy coat is present as a thin creamy white colored layer on top of the red blood ceils.
  • the plasma portion of the blood forms a layer above the buffy coat.
  • Fractions from blood can also be isolated in a variety of other ways. One method is by taking a fraction or fractions from a gradient used in centrifugation to enrich for a specific size or density of cells.
  • DNA is then isolated from samples from the bodily fluids. Procedures for isolation of DNA from such samples are well known to those skilled in the art. Commonly, such DNA isolation procedures comprise lysis of any cells present in the samples using detergents, for example. After cell lysis, proteins are commonly removed from the DNA using various proteases, RNA is removed using RNase. The DNA is then commonly extracted with phenol, precipitated in alcohol and dissolved in an aqueous solution.
  • the disclosure provides for a method of determining whether a subject has any one or more of the methylated informative loci disclosed herein that are indicative of the presence of a metaplasia in the esophagus (e.g., Barrett ' s esophagus), wherein if the subject is determined to have a metaplasia in the esophagus (e.g., Barrett's esophagus), the subject is treated with an agent that treats the metaplasia in the esophagus (e.g. , Barrett's esophagus).
  • a metaplasia in the esophagus e.g., Barrett 's esophagus
  • the disclosure provides for a method of treating a subject determined to have a metaplasia in the esophagus (e.g., Barrett's esophagus), in some embodiments, the treatment of a metaplasia in the esophagus (e.g., Barrett's esophagus) encompasses administration of any one or more of the following compounds: proton pump inhibitors (PPIs) such as omeprazole (Prilosec, Zegerid), lansoprazole (Prevacid), pantoprazole (Protonix), rabeprazoie (AcipHex), esomeprazole (Nexium), dexlansoprazole (Dexllant).
  • PPIs proton pump inhibitors
  • Histamine H2 receptor blocking agents such as eimetidine (Tagamet), ranitidine (Zantac), famotidine (Pepcid) and nizatidine (Axid). Turns, Rolaids, or other quick-acting reflux medications. Prokinetic agents, or drugs thai help move food through the gastrointestinal tract more quickly, offer an attractive alternative either alone or in combination with acid inhibition,
  • the treatment of a metaplasia in the esophagus is endoscopic mucosal resection (EMR);
  • the disclosure provides for a method of determining whether a subject has any one or more of the methylated informative loci disclosed herein that are indicative of esophageal neoplasia (e.g., esophageal cancer), wherein if the subject is determined to have an esophageal neoplasia (e.g.
  • the subject is treated with an agent that treats the esophageal neoplasia (e.g., esophageal cancer).
  • the disclosure provides for a method of determining whether a subject has any one or more of the methylated informative loci disclosed herein in combination with any of the TP53 somatic mutations disclosed herein that are indicative of esophageal neoplasia (e.g., esophageal cancer), wherein if the subject is determined to have an esophageal neoplasia (e.g., esophageal cancer), the subject is treated with an agent that treats the esophageal neoplasia (e,g.
  • the disclosure provides for a method of treating a subject determined to have esophageal neoplasia (e.g., esophageal cancer), in some embodiments, the esophageal neoplasia is Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia (HGD) and/or esophageal adenocarcinoma (EAC). in some embodiments, the treatment of esophageal neoplasia (e.g., esophageal cancer) encompasses surgery (e.g.
  • the treatment of esophageal neoplasia encompasses administering one or more chemotherapeutic agent, such as any one or more therapeutic agent selected from the group consisting of: carboplatm and paelitaxel (Taxol®) (which may be combined with radiation): cispiatin and 5-fluorouracil (5 ⁇ FU) (often combined with radiation); ECF: epirubicine (Ellence®), cispiatin, and 5-FU (especially for gastroesophageal junction tumors); DCF: docetaxe!
  • chemotherapeutic agent such as any one or more therapeutic agent selected from the group consisting of: carboplatm and paelitaxel (Taxol®) (which may be combined with radiation): cispiatin and 5-fluorouracil (5 ⁇ FU) (often combined with radiation); ECF: epirubicine (Ellence®), cispiatin, and 5-FU (especially for gastroesophageal
  • Ramucirumab may be used to treat cancers that start at the gastroesophageal (GE) junction when they are advanced, in some embodiments, the treatment encompasses endoscopic treatments, such as endoscopic mucosal resection (EMR) followed by treatment with proton pump inhibitors, photodynarme therapy (PL)T); radiofrequency ablation (RFA); laser ablation; argon plasma coagulation (APC); electrocoagulation (electrofulguration); or esophageal stent.
  • EMR endoscopic mucosal resection
  • PL photodynarme therapy
  • RPA radiofrequency ablation
  • APC argon plasma coagulation
  • electrocoagulation electrocoagulation
  • treatment generally mean obtaining a desired pharmacologic and/or physiologic effect, and may also be used to refer to improving, alleviating, and/or decreasing the severity of one or more symptoms of a condition being treated.
  • the effect may be prophylactic in terms of completely or partially delaying the onset or recurrence of a disease, condition, or symptoms thereof, and/or may be therapeutic in terms of a partial or complete cure for a disease or condition and/or adverse effect attributable to the disease or condition.
  • Treatment covers any ireatmeni of a disease or condition of a mammal, particularly a human, and includes: (a) preventing the disease or condition from occurring in a subject which may be predisposed to the disease or condition but has not yet been diagnosed as having it; (b) inhibiting the disease or condition (e.g., arresting its development); or (c) relieving the disease or condition (e.g., causing regression of the disease or condition, providing improvement in one or more symptoms).
  • Treating a metaplasia (e.g. , Barrett's esophagus) and/or a neoplasia (e.g. , esophageal cancer) in a subject refers to improving (improving the subject's condition), alleviating, delaying or slowing progression or onset, decreasing the severity of one or more symptoms associated with a metaplasia (e.g., Barrett's esophagus) and/or a neoplasia (e.g., esophageal cancer).
  • treating a metaplasia or neoplasia includes any one or more of:
  • metaplastic/neoplastic cells reducing growth, proliferation and/or survival of metaplastic/neoplastic cells, killing metaplastic/neoplastic cells (e.g. , by necrosis, apoptosis or autophagy), decreasing
  • metaplasia/neoplasia size decreasing rate of metaplasia/neoplasia size increase, halting increase in metaplasia/neoplasia size, improving ability to swallow, decreasing internal bleeding, decreasing incidence of vomiting, reducing fatigue, decreasing the number of metastases, decreasing pain, increasing survival, and increasing progression free survival.
  • Example 1 Identification of esophageal, cancer mformatiye loci.
  • Methylated informative loci were initially identified using the technique of reduced representation bisulfite sequencing (RRBS) in a discovery set of 23 paired biopsies of normal squamous esophagus and matched esophageal adenocarcinomas, along with biopsies of 8 Barrett's esophagus tissue, and along with brushings of 8 Barrett's esophagus tissues (one BE brushing case also having a matched biopsy).
  • RRBS reduced representation bisulfite sequencing
  • Discovery data were initially analyzed for each individual CpG residue in the RRBS data set. Individuai CpGs were considered methylated in EAC if they showed methylation in less than 10% of DNA sequence reads in all of the informative squamous samples, where at least 4 squamous samples were informative, where an informative sample had equal to or greater than 20 reads covering the CpG, and if 8 or more of the informative EAC samples demonstrated percent methylation at a level that was at least 20 percentage points greater than the methylation le vel of the most methylated normal squamous sample.
  • CpGs were similarly defined as methylated in Barrett's esophagus if they showed methylation of less than 10% of DNA sequence reads of ail informative squamous samples, where an informative sample had equal to or greater than 20 reads covering the CpG, and if 3 or more of the informative BE samples demonstrated percent methylation at a level that was at least 20 percentage points greater than the methylation level of the most methylated normal squamous sample.
  • CpGs meeting criteria for methylation in both EACs and BE were defined as methylated in both EAC and BE, Such methylated CpGs were then aggregated into patches by grouping together methylated CpGs that were within 200bp of one another. Patches may consist of 1 CpG up to any number of CpGs that meet the above criteria.
  • the names assigned to 428 genomic patches defined as methylated in both EAC and BE by the criteria above were recorded, and the sequences of these loci correspond to SEQ ID NOs: 1 -856.
  • the genomic coordinates of the genomic patches defined as methylated by the above criteria were also recorded.
  • the genomic sequences of these patches on the respective genomic (+) and (-) strands were determined and recorded, (Upper and lower case designations were used according to those of the UCSC browser, where lower ease sequences are lower complexity DNA sequences).
  • C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethy fated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as meihylation at that base.
  • sequences represent the group of ail combinations of all sequences in which 0, 1 , or more than one Y is converted to a T.
  • the reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
  • the bisulfite converted sequences of the fully methylated form of the corresponding patches i.e.
  • Patches were expanded by 100 base pairs on either side so as to accommodate either the design of amplification pimers or to enclude additional presumptively methylated bases.
  • the sequences of these expanded patches correspond to SEQ ID NOs: 2569-3424 and their genomic coordinates were also recorded.
  • the genomic sequences of these expanded patches on the respective genomic (+) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences).
  • the bisulfite converted sequences of these corresponding expanded patches i.e.
  • C residues that may be methylated or unmemyiated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base.
  • sequences represent the group of all combinations of all sequences in which 0, 1, or more than one Y is con verted to a T.
  • the reverse complements of the bisulfite con verted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
  • the bisulfite converted sequences of the fully methylated form of the corresponding expanded patches i.e.
  • CpG islands overlapping patches that may contain additional CpGs that are methylated coordinately with patches were also defined.
  • the sequences of these CpG islands correspond to SEQ ID NOs: 5137-5926,
  • the genomic coordinates of the CpG islands were also recorded.
  • the genomic sequences of these expanded patches on the respective genomic ( +) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DN A sequences).
  • the bisulfite converted sequences of these corresponding CpG islands i.e.
  • Regions of Interest were defined that provided preferred regions for design of PGR araplicons that would encompass preferred patches.
  • the genomic coordinates of the ROI were also recorded.
  • the sequences of the (+) strands of the Regions of Interest correspond to SEQ ID NOs: 8209-8222, and the sequences of the (-) strands Regions of interest correspond to SEQ ID NOs: 8251-8261 , (Upper and lower case designations were used according to those of the UCSC browser, where lower ease sequences are lower complexity DNA sequences).
  • the bisulfite converted sequences of these corresponding Regions of interest i.e.
  • C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base.
  • these sequences represent the group of all combinations of all sequences in which 0, 1 , or more than one Y is converted to a T.
  • the reverse complements of the bisulfite converted sequences of the (+) and (-) s trands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
  • the bisulfite converted sequences of the fully methylated form of the Regions of Interest i.e.
  • the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 8307-8320 and 8410-8414 for the bisulfite converted sequences of the (+) strands and see sequences of SEQ ID NOs: 8349-8362 and 8425-8429 and for die bisulfite converted sequences of the (-) strands), C residues that may be methylated or immethylatecl, and hence may be bisulfite converted to T (if immethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base.
  • these sequences represent the group of all combinations of ail sequences in which 0, 1 , or more than one Y is converted to a T.
  • the reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
  • the bisulfite converted sequences of the fully methylated form of the Arnplicons herein.
  • biopsies from: 23 EACs, 8 HGD, 15 non-dysplastic BE from individuals without known higher grade lesions.
  • biopsies were obtained from 5 eases of BE adjacent to HGD, and from 1 1 cases of BE adjacent to an EAC. These were not included in analyses of non-dysplastic BE.
  • biopsies were obtained from 33 normal squamous mucosa samples.
  • Table. 1 describes the performance in the confirmatory sample set using bisulfite sequencing analysis of amplicons of select loci having strong performance characteristics and identified in the studies discussed above.
  • columns C - S disclose the performance of the select amplicons.
  • the number of CpGs that were methylated was counted, and the read was classified as methylated or unmethyled using cutoffs for a required number of methylated CpGs on the amplicon.
  • Table 1 , row 3 lists the number of CpGs between the amplification primers for each of the amplicons, Table 1 , row 4 lists the number of CpGs that need to be methylated on an individual read to count that read as methylated (e.g.
  • Table 1 , rows 6, 7, and 8 record the sensitivity for detecting esophageal adenocarcinomas (EACs) (row 6), high grade dysplasias (HGD) (row 7) and non-dysplastic Barrett's esophagus (non-dysplastic BE) (row 8), using criteria in which a sample was detected if it demonstrated methylation in greater than 10% (0.1) of all DNA reads.
  • EACs esophageal adenocarcinomas
  • HCD high grade dysplasias
  • non-dysplastic BE non-dysplastic Barrett's esophagus
  • Table 1 row 9 records the specificity of each amplicon for not detecting normal squamous mucosa again using criteria in which a sample was detected if it demonstrated methylation in greater than 10% (0.1) of all DNA reads, Table 1 , row 11 records the specificity of each amplicon for not detecting normal squamous mucosa now using criteria in which a sample was detected if it demonstrated methylation in greater than 1% (0,01) of all DNA reads.
  • Table 1 column B provides the same data for detecting methylation in the Vimentin (VIM) locus amplified using primers disclosed in Li et al. (Li M, et al. (2009) Sensitive digital uantification of DNA methylation in clinical samples. Nat Biotechnol 27(9):858-863).
  • hese primers correspond to SEQ ID NOs: 8445-8446.
  • the amplicons amplified using these rimers are as follows:
  • Vimentin amplicon (+) strand (SEQ ID NO: 16207):
  • Vimentin amplicon (-) strand (SEQ ID NO: 16208):
  • Amplicons (and patches) need not be used individually, but can be combined into anels for detection of esophageal neoplasia. Examples of such panels, and their associated erformance statistics, are provided in Table I , columns T through AG mat provide the markers in the panel and the sensitivity and specificity resulting from the marker combination when the combination is positive if any member of the combination is positive).
  • the sensitivity for detection of EAC (100%), HGD (88%), and BE (100%) is the same mong the combinations shown of: all amplicons, 17 amplicons, 15 amplicons, 4 amplicons, hree of four combinations of 3 amplicons (columns Y, Z, AA), and for one combination of 2 mplicons (column AF).
  • Specificity for not detecting normal squamous (97%), at a detection utoff of 10% of reads being methylated, is the same for all combinations shown of: 15 mplicons, 4 amplicons, 3 amplicons, or 2 amplicons.
  • Discovery data were also analyzed for each individual CpG residue in the RRBS data set to identify loci that could be used to distinguish EAC from BE.
  • Individual CpGs were considered methylated in EAC versus BE if they showed methylation of less than 10% of reads of all informative BE samples, where at least 3 BE samples were informative, and if they showed methylation of less than 10% of reads of all informative normal squamous samples, and where an informative sample had equal to or greater man 20 reads covering the CpG, and if 6 or more of the EAC samples demonstrated percent methylation at a level mat was at least 20 percentage points greater than the methylation level of the most methylated BE sample.
  • CpGs meeting criteria for methylation in EAC versus and BE are defined as methylated in EAC vs BE. Such methylated CpGs were then aggregated into patches in instances in which methylated CpGs were within 200bp one another.
  • genomic patches defined as methylated in EACs versus BE in the discovery set were identified (see SEQ ID NOs: 8447-8818).
  • the genomic coordinates of the genomic patches defined as methylated by the above criteria were also recorded.
  • the genomic sequences of these patches on the respective genomic (+) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences).
  • the bisulfite converted sequences of these corresponding patches i.e.
  • C residues mat may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at mat base.
  • sequences represent the group of all combinations of all sequences in which 0, 1, or more man one Y is converted to a T.
  • the reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
  • the bisulfite converted sequences of the fully methylated form of the corresponding patches i.e.
  • Patches were expanded by 100 base pairs on either side so as to accommodate either the design of amplification pimers or to enc!ude additional presumptively methylated bases.
  • the sequences of these expanded patches correspond to SEQ ID NOs: 9563-9934 and their genomic coordinates were also recorded.
  • the genomic sequences of these expanded patches on the respective genomic (+) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences).
  • the bisulfite converted sequences of these corresponding expanded patches i.e.
  • the bisulfite converted sequence of the (-(-) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 9935-10120 and 10307- 10492 for the bisulfite converted sequences of the respective ( f) and (-) strands), C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methy!ation at that base.
  • sequences represent the group of ail combinations of all sequences in which 0, 1, or more than one Y is converted to a T.
  • the reverse complements of the bisulfite converted sequences of the (-(-) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
  • the bisulfite converted sequences of the fully methylated form of the corresponding expanded patches i.e.
  • the sequences of these CpG islands correspond to SEQ ID NOs: 10679-10972.
  • the genomic coordinates of the CpG islands were also recorded.
  • the genomic sequences of these expanded patches on the respective genomic (+) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences).
  • the bisulfite converted sequences of these corresponding CpG islands i.e.
  • C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base.
  • sequences represent the group of all combinations of all sequences in which 0, 1, or more than one Y is converted to a T.
  • the reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
  • the bisulfite converted sequences of the fully methylated form of the corresponding CpG islands i.e.
  • Regions of Interest were defined that provided preferred regions for design of PCR amplicons that would encompass preferred patches.
  • the genomic coordinates of the ROI were also recorded.
  • the sequences of the (+) strands of the Regions of Interest correspond to SEQ ID NOs: 12563-12568, and the sequences of the (-) strands Regions of Interest correspond to SEQ ID NOs: 12581-12586. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences).
  • the bisulfite converted sequences of these corresponding Regions of Interest i.e.
  • C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at mat base.
  • these sequences represent the group of all combinations of all sequences in which 0, 1, or more man one Y is converted to a T.
  • the reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
  • the bisulfite converted sequences of the fully methylated form of the Regions of Interest i.e.
  • the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand were determined and recorded (see sequences of SEQ ID NOs: 12605-12610 and 12650-12652 for the bisulfite converted sequences of the (+) strands and see sequences of SEQ ID NOs: 12623-12628 and 12659-12661 and for the bisulfite converted sequences of the (-) strands).
  • the bisulfite converted sequences of the fully methylated form of the Amplicons i.e. in which all Y bases in every bisulfite converted sequence are retained as aC
  • corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ IDNOs: 12611-12616 and 12653-12655 for the bisulfite converted sequences of the fully methylated form of the (+) strands and see sequences of SEQ ID NOs: 12629-12634 and 12662-12664 for the bisulfite converted sequences of the fully methylated form of the (-) strands).
  • Table 2 describes the performance in the confirmatory sample set using bisulfite sequencing analysis of amplicons from select loci defined as methylated in EACs versus BE and having preferred marker characteristics.
  • Columns B - J disclose the performance of the amplicons in the confirmatory data set. hi this data set, methylation was calculated as the average level of methylation of all CpGs in between the primers for amplifying the amplicon. For each read across each ampHcon the number of CpGs that were methylated was counted and the read was classified as methylated or unmethyled using cutoffs for a required number of methylated CpGs on the amplicon.
  • rows 8 and 9 record the specificity of each amplicon for not detecting non-dysplastic BE (row 8) and for not detecting normal squamous mucosa (row 9) again using criteria in which a sample was detected if it demonstrated methylation in greater than 10% (0.1) of all DNA reads.
  • rows 11 and 12 record the specificity of each amplicon for not detecting non-dysplastic BE (row 11) and for not detecting normal squamous mucosa (row 12) using criteria in which a sample was detected if it demonstrated methylation in greater than 1% (0.01) of all DNA reads.
  • Ampllcons (and patches) need not be used individually, but can be combined into panels for detection of esophageal neoplasia. Performance statistics of selected panels of amplicons are provided in Table 2 columns K through V mat provides the sensitivity and specificity of the panels (when the combination is positive if any member of the combination is positive).
  • RRBS discovery data was analyzed to identify CpG residues that demonstrated: i) at least 3 informative BE, which in every informative BE demonstrated at least 90% methylation; and where ii) no more than 5% of informative normal squamous samples demonstrated methylation level below 90%,;and that iii) demonstrated at least 6 informative EAC, where in these informative EACs the level of methylation was at least 20 percentage points lower than the methylation level of the least methylated BE.
  • CpGs meeting these criteria are defined as unmethylated in EAC versus BE. Such unmethylated CpGs were then aggregated into patches by grouping together unmethylated CpGs that were within 200bp of one another. Unmethylated in EAC patches may consist of 1 CpG up to any number of CpGs that meet the above criteria.
  • Example 3 Identification of esophageal cancer informative locito detect progression of esophageal neoplasia
  • Biopsy samples (that overlapped with the confirmatory biopsy sample set) were further analyzed in tests of panels of markers for detecting the progression of Barrett's esophagus to Barrett's esophagus high grade dysplasia (HGD) or to esophageal adenocarcinoma (EAC).
  • Figure 1 shows the sensitivity (percentage of samples detected), the specificity (percentage of samples not detected), the total number of samples studied, and the total number of positive samples. Three panels of markers were selected for study. The first marker pane!
  • the second panel consisted of detecting at least one of the following four methylated markers: Upl5-1, Up35-1, Up27, and UplO (using bisulfite sequencing analysis of the corresponding amplicons and using the criteria for detection specified in table 2A).
  • the second panel consisted of testing for somatic non- synonymous mutations in TP53 in assays in which TP53 was amplified from genomic DNA using a set of PGR amplicons that spanned the TP53 coding region and in which Next Generation DNA Sequencing was then used to compare TPS 3 sequences from esophageal lesions versus matched normal esophagus tissue. Samples were classified as detected if a TP53 mutant allele frequency of greater than or equal to 10% was identified.
  • the third panel was a combination of detection of methylation in any of Upl 5-1, Up35-1 , Up27, and UplO or detection of mutation in TP53.
  • Table 3 shows the individual performance of biomarkers in detecting the different sample types of the 1st validation biopsies set using different cut-off criteria for methylation than the analysis of table 2. Shown is the performance for detection of different sample types of 5 different methylated DNA markers tested by bisulfite sequencing analysis of the corresponding amplicons (Up3, Up 10, Up27, Up35-1, Up35-2). Table 3 specifies the number of CpGs required to be methylated on a DNA sequence read to classify that read as methylated for this analysis. Results are presented when samples are considered methylated if greater than or equal to 1% of all DNA sequence reads are classified as methylated, or if greater than or equal to 10% of all DNA sequence reads are classified as methylated.
  • TP53 was amplified from genomic DNA using a set of PGR amplicons that spanned the TP53 coding region and in which Next Generation DNA Sequencing was then used to compare TP53 sequences from esophageal lesions versus matched normal esophagus tissue. Shown are rates of sample detection (expressed as sensitivity or specificity) when samples are classified as TP53 mutant if greater than or equal to 3% of TP53 reads are scored as mutant, or if greater than or equal to 10% of TP53 reads are scored as mutant.
  • Table 4 shows the performance of selected combinations of the methylated DNA markers (Up3, Up 10, Up27, Up35-1 , Up35-2) for detection of different sample types in the experiment presented in Table 3, Samples are scored as methylated if any member of the marker combination panel scores the sample as methylated. Results are presented when the individual markers are considered methylated if greater than or equal to 1% of all DNA sequence reads are classified as methylaied, or if greater than or equal to 10% of all DNA sequence reads are classified as methylated.
  • Table 5 shows performance of selected combinations of methylated DNA markers (Up3, Up 10, Up27, Up35-1 , Up35-2) plus testing for mutations in TP53 (p53) for detection of different sample types. Samples are scored as detected if any member of the marker combination panel scores the sample as methylated or if analysis for TP53 mutations scores the sample as TPS 3 mutant. Shown is the performance of the marker panel in which samples are scored as detected if any methylation marker is detected as methylated at greater than or equal to 1% of DNA reads, or if TPS 3 is detected as mutant at greater than or equal to 10% of the DNA sequence reads.
  • the sample set included brushings from 49 esophageal adenocarcinomas (EAC); 14 carcinomas of the gastroesophageal junction (JCA); 8 Barrett's esophagus with low grade dysplasia (LGD); 9 Barrett's esophagus with high-grade dysplasia (HGD); 33 cases of Barrett's esophagus without dysplasia from cases without HGD or EAC, otherwise termed non-dysplastic BE, that included 1.3 cases of short segment Barrett's esophagus (SSBE).
  • EAC esophageal adenocarcinomas
  • JCA gastroesophageal junction
  • JCA gastroesophageal junction
  • LGD low grade dysplasia
  • HSD high-grade dysplasia
  • SSBE non-dysplastic BE
  • Table 6 shows the individual performance of biomarkers in detecting the different sample types of the validation brushings set. Shown is the performance for detection of different sample types of 5 different methylated DNA markers analyzed by bisulfite sequencing of selected ampiicons (Up3, Up 10, Up27, Up35-1, Up35-2). The table specifies the number of CpGs required to be methylated on a DNA sequence read to classify that read as methylated. Results are presented when samples are considered methylated if greater than or equal to 1% of all DNA sequence reads are classified as methylated, or if greater than or equal to 10% of all DNA sequence reads are classified as methylated.
  • Table 7 shows the performance of selected combinations of the methylated DNA markers (Up3 5 Up 10, Up27, Up35-1, Up35-2) for detection of different, sample types in the esophageal brush ings samples presented in Table 6. Samples are scored as methylated if any member of the marker combination panel scores the sample as methylated. Results are presented when the individual markers are considered methylated if greater than or equal to 1% of all DNA sequence reads are classified as methylated, or if greater than or equal to 10% of all DNA sequence reads are classified as methylated.
  • Table 8 shows the performance of selected combinations of methylated DNA markers (Up3, Up1O, Up27, Up35 ⁇ l , Up35-2) plus testing for mutations in TP53 ( ⁇ 53) for detection of different sample types in the esophageal brushings samples presented in Tables 6 and 7. Samples are scored as detected if any member of the marker combination pane! scores the sample as methylated or if analysis for TPS 3 mutations scores the sample as TP53 mutant. Shown is the performance of the marker panel in which samples are scored as detected if any methylation marker is detected as methylated at greater than or equal to 1% of DNA reads, or if TPS 3 is detected as mutant at greater than or equal to 3% or at greater than or equal to 10% of the DNA sequence reads.
  • A!so shown is the performance of the marker pane! in which samples are scored as detected if any methylation marker is detected as methylated at greater thars or equal to 10% of DMA reads, or if TP53 is detected as mutant at greater than or equal to 3% or at greater than or equal to 10% of the DNA sequence reads. Marker combination with greater than 90% specificity in all BE without dysplasia are preferred yellow.
  • Marker combinations that additionally show superior sensitivity for EAC are further preferred, Particularly preferred marker combinations are: Up35-2 methylation plus TP53 mutation; Up35-2 methylation plus Up3 methylation plus TPS 3 mutation; Up 10 methylation plus Up3 methylation plus TP53 mutation; Up35-2 methylation plus Up1O methylation plus TP53 mutation; Up1O methylation plus Up27 methylation plus TP53 mutation: Up35-2 methylation plus Up3 methylation plus Up10 methylation plus TP53 mutation.
  • Table 9 summarizes the side by side comparison of 8 methylated DNA markers in
  • FFPE tissue samples of the stomach and esophagus that capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma. Intestinal metaplasia is abbreviated as IM.
  • Table 9 denotes for each marker the number of methylated cytosine bases required to be detected on a DNA sequence read to classify that read as methylated. Samples are detected as methylated if greater than or equal to 1% of DNA sequence reads are classified as methylated.
  • Table 10 summarizes the side by side comparison of 8 methylated DNA markers in FFPE tissue samples of the stomach and esophagus that capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma, intestinal metaplasia is abbreviated as iM.
  • Table 10 denotes for each marker the number of methylated cytosine bases required to be detected on a DNA sequence read to classify that read as methylated. Samples are detected as methylated if greater than or equal to 10% of DNA sequence reads are classified as methylated,
  • Table 1 1 summarizes performance of different panels comprised of combinations of methylated DNA markers in FFPE tissue samples of the stomach and esophagus that capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma. Intestinal metaplasia is abbreviated as IM.
  • Tables 9 and 10 denote for each marker the number of methylated cytosine bases required to be detected on a DMA sequence read to classify that read as methylated. Samples are detected as methylated if greater than or equal to 1 % of DNA sequence reads are classi fied as methylated for any member of the marker panel.
  • Table 12 summarizes performance of different panels comprised of combinations of methylated DNA markers in FFPE tissue samples of the stomach and esophagus thai capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma. Intestinal metaplasia is abbreviated as IM.
  • Tables 9 and 10 denote for each marker the number of methylated cytosine bases required to be detected on a DNA sequence read to classify that read as meihylaled. Samples are detected as methylated if greater than or equal to 10% of DN A sequence reads are classified as methylated for any member of the marker panel.
  • TP53 exons 2-11 were amplified using a multiplexed series of primer pairs covering all coding sequences and splice junctions.
  • the primers contained additional 5' end sequences that were then used for secondary amplification that introduced barcode sequences and Illumina 15 and 17 sequences into the final FCR products.
  • FCR products were mixed, purified and analyzed on an Illumina MiSeq instrument. Data analysis was performed using CLCBio software (Qiagen) and VarianiSiudio software (Illumina).

Landscapes

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

Abstract

The disclosure provides methods for identifying genomic loci that are differentially methylated in neoplastic cancers, e.g., esophageal cancers. Identification of methylated genomic loci, and optionally in combination with the identification of somatic mutations in TP53, has numerous uses, including for example, to characterize disease risk, to predict responsiveness to therapy, to non-invasively diagnose subjects and to treat subjects determined to have gastrointestinal neoplasias.

Description

METHODS AND COMPOSITIONS FOR DETECTING ESOPHAGEAL NEOPLASIAS AND/OR METAPLASIAS IN THE ESOPHAGUS elated Applications
This application claims the benefit of priority to United States provisional application rial number 62/099,021 , filed December 31, 2014. The disclosure of the foregoing plication is hereby incorporated by reference in its entirety. unding
Work described herein was supported by grant nos. UOI CAI 52756; U54CA163060;nd P50CA 150964. The United States Government has certain rights in the invention. ackground
Over 15,000 new cases of esophageal cancer were diagnosed in 2010, and there wereearly as many deaths from this cancer alone. As with other cancers, this rate can beecreased by improved methods for diagnosis. Although methods for detecting esophageal ncer exist, the methods are not ideal. Generally, a combination of endoscopy, isolation of lls (for example, via collection of cells/tissues from a fluid sample or from a tissue sample),nd/or imaging technologies are used to identify cancerous cells and tumors. While upperndoscopy, usually performed by a gastroenterologist, can detect neoplasias of the esophagus, well as of the stomach and duodenum, it is an uncomfortable and expensive procedure. ther detection procedures, such as barium esophogography are also available, but are sociated with false positives, false negatives, and cost and discomfort issues.
Because of the disadvantages of existing methods for detecting or treating esophagealeoplasias/cancers, new methods are needed for esophageal neoplasia cancer diagnosis and erapy. ummary Of The Disclosure
in certain aspects, the present disclosure is based in part on the discovery of particularuman genomic DNA regions (also referred to herein as informative loci or patches) in which the cytosines within CpG dinucleotides are differentially methylated in esophageal neoplasia compared to normal human tissues.
In one embodiment, the method comprises assaying for the presence of differentially methylated genomic loci in a tissue sample or a bodily fluid sample from a subject. The tissue sample may he obtained from biopsies of the gastrointestinal tract, including hut not limited to the esophagus and stomach, Tissue samples may be obtained as a biopsy, or as a swab or brushing of the gastrointestinal tract (e.g., esophagus or stomach), or other organs believed to contain cancerous cells or tissues. Exemplary bodily fluids include blood, serum, plasma, saliva, spit, esophageal washing, a blood-derived fraction, or urine, in one embodiment, the method involves methylation-sensitive restriction enzyme(s), In another embodiment, the method involves methylation-sensitive PGR. In another embodiment, the method involves restriction enzynie/metbylation-sensitive PCR. In yet another embodiment, the method comprises reacting DNA from the sample with a chemical compound that converts non-methylated cytosine bases (also called "con version-sensitive" cytosines), but not methylated cytosine bases, to a different nucleotide base. In an embodiment, the chemical compound is sodium bisulfite, which converts unmethylated cytosine bases to uracil. The compound-converted DNA is then amplified using a methylation-sensitive polymerase chain reaction (MSP) employing primers that amplify the compound-converted DNA template if cytosine bases within CpG dinucleotides of the DNA from the sample are methylated, Production of a PCR product indicates that the subject has cancer or precancerous adenomas, in some embodiments, compound-converted DNA is amplified by bisulfite specific methylation indifferent PCR primers and methylation of the parental DNA template is inferred by DNA sequence analysis of the bisulfite converted and amplified product. Other methods for assaying for the presence of methylated DNA are known in the art. In certain embodiments, the method optionally comprises detecting the presence or absence of a somatic mutation in TP53.
In another embodiment, the present invention provides a detection method of prognosis of a neoplasia (or metaplasia) (e.g.. upper gastrointestinal neoplasia, esophageal neoplasia) in a subject known to have or suspected of having neoplasia (or metaplasia), In some embodiments, the neoplasia is cancer. Such method comprises assaying for the presence of methylated informative loci in a tissue sample or bodily fluid from the subject. In certain cases, it is expected that detection of methylated informative loci in a tissue sample or bodily fluid from the subject is indicative of an advanced state of cancer (e.g., gastrointestinal cancer such as esophageal cancer). In other cases, detection of methylated informative loci in a tissue sample or sample from other bodily fluids may be indicative of a cancer that will respond to therapeutic agents that demethylate DNA or reactivate expression of genes located within methylated informative loci, in certain embodiments, the method optionally comprises detecting the presence or absence of somatic mutation(s) in TP53. In certain cases, detection of somatic mutaiion(s) in TP53 in a tissue sample or bodily fluid from the subject is indicative of an advanced state of cancer (e.g., gastrointestinal cancer such as esophageal cancer). In other cases, detection of somatic mutation(s) in TP53 in a tissue sample or sample from other bodily fluids may be indicative of a cancer that will respond to therapeutic agents that demethylate DNA or reactivate expression of genes located within methylated informative loci.
In another embodiment, the present invention pro vides a method of monitoring over time the status of neoplasia (e.g., gastrointestinal neoplasia such as esophageal neoplasia) in a subject. in some embodiments, the neoplasia is a cancer. In some embodiments, the neoplasia is a metaplasia such as Barrett's esophagus. In certain embodiments, the method optionally comprises detecting the presence or absence of somatic mutation(s) in TP53 in combination with detecting the methylation status of any of the informative loci disclosed herein. In certain cases, detection of the appearance of somatic mutation(s) in TPS 3 over time in a tissue .sample or bodily fluid from the subject is indicative of neoplastic progression, in other cases, detection of a disappearance of a somatic mutation(s) in TP53 over time in a tissue sample or sample from other bodily fluids may be indicative of neoplastic regression.
In another embodiment, the present invention provides a method of e valuating therapy in a subject having cancer or suspected of having neoplasia (e.g., gastrointestinal neoplasia such as esophageal neoplasia), In some embodiments, the neoplasia is a cancer. In some embodiments, the neoplasia is a metaplasia such as Barrett's esophagus. In certain embodiments, the method optionally further comprises detecting the presence or absence of somatic mutation(s) in TP53. The present invention also relates to oligonucleotide primer sequences for use in assays (e.g., methylation-sensitive PGR assays or Hpall assays) designed to detect the methyiation status of the informative methylated genomic loci.
The present invention also provides a method of inhibiting or reducing growth of neoplasia cells (e.g., gastrointestinal neoplasia such as esophageal neoplasia). In some embodiments, the neoplasia is a cancer. In some embodiments, the neoplasia is a metaplasia such as Barrett's esophagus.
in some embodiments, the disclosure provides for a method of delecting the presence of an esophageal neoplasia or metaplasia in the esophagus in a human subject, comprising: a) obtaining a human sample; and b) assaying the sample for the presence of methyiation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1
- 856, 2569 - 3424, 51.37 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047
- 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, or complements or fragments thereof; wherein methyiation of the nucleotide sequence is indicative of an esophageal neoplasia or metaplasia in the esophagus. In some embodiments, the disclosure provides for a method of detecting an esophageal neoplasia or metaplasia in the esophagus, comprising: a) obtaining a human sample; and b) assaying said sample for the presence of DNA methyiation by assay in a bisulfite converted DNA for retention of a eytosine base at any of the Y posi tions present in one or more of the nucleotide sequences having at least 90% identical to the sequence of any one or more of SEQ ID NOs: 857-1284, 1713-2140, 3425-3852, 4281-4708, 5927-6321, 6717-711 1, 7559-7584, 761 1 - 7636, 7715-7740, 7767-7792, 7867-7890, 7915-7938, 7991-8004, 8019-8032, 8075-8088, 8103-81 16, 8157-8169, 8183-8195, 8223-8236, 8265-8278, 8307-8320, 8349-8362, 8410- 8414 or 8425-8429: wherein methyiation of the nucleotide sequence is indicative of metaplasia in Ihe esophagus or esophageal neoplasia, in some embodiments, the sample is assayed for the presence of methyiation at any of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of SEQ ID NOs: 7991-8004, 8019-8032, 8075-8088, 8103-81 16, 8157-8169, 8183-8195, 8223-8236, 8265-8278, 8307-8320, 8349-8362, 8410-8414 or 8425-8429. In some embodiments, the disclosure provides for method of detecting an esophageal neoplasia, comprising: a) obtaining a human sample; and b) assaying the sample for the presence of methyl atlon within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447- 8818, 9563-9934, 10679-10972, 11561-11662, 1 1867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or complements or fragments thereof, in some embodiments, the disclosure provides for a method of detecting an esophageal neoplasia, comprising: a) obtaining a human sample; and b) assaying the sample for the presence of DNA methylation by assay in a bisulfite converted DNA for retention of a cytosine base at any of the Y positions present in one or more of the nucleotide sequences ha ving at least 90% identical to the sequence of any one or more of SEQ ID NOs: 8819-9004, 9191 -9376, 9935-10120, 10307-10492, 10973-1 1 1 19, 1 1267-11413, 11663-11713, 11765-1 1815, 11969-12019, 12071-12121, 12267-12313, 12361-12407, 12467-12472, 12479-12484, 12503-12508, 12515-12520, 12539-12544, 12551 -12556, 12569-12574, 12587-12592, 12605-12610,
12623-12628, 12650-12652, or 12659-12661 ; wherein methylation of the nucleotide sequence is indicative of metaplasia in the esophagus. In some embodiments, the sample is assayed for the presence of DNA methylation by assay in a bisulfite converted DNA for retention of a cytosine base at any of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of SEQ ID NOs: 1 1663-
1 1713, 1 1765-11815, 11969-12019, 12071-12121, 1226742313, 12361-12407, 12467-12472, 12479-12484, 12503-12508, 12515-12520, 12539-12544, 12551 -12556, 12569-12574, 12587-12592, 12605-12610, 12623-12628, 12650-12652, or 12659-12661. In some embodiments, the subject has been assayed for having a somatic mutation in TPS 3, in some embodiments, the esophageal neoplasia or metaplasia in the esophagus is selected from the group consisting of Barrett's esophagus (BE), and/or, Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia (HGD), and/or esophageal adenocarcinoma (EAC), In some embodiments, the sample is obtained from a subject suspected of having or is known to have an esophageal neoplasia or metaplasia in the esophagus, in some embodiments, the assay is methylation-specific PGR. in some embodiments, the methods disclosed herein further comprise: a) treating DNA from the sample with a compound that converts a non-methylated cytosine base in the DNA to a different base; b) amplifying a region of the compound converted nucleotide sequence with a forward primer and a reverse primer; and c) analyzing the meihylation patterns of the nucleotide sequences. In some embodiments, the methods disclosed herein further comprise: a) treating DNA from the sample with a compound that converts a non-methylated cytosine base in the DNA to a different base: b) amplifying a region of the compound converted nucleotide sequence with a forward primer and a reverse primer; and c) detecting the presence and/or amount of the amplified product. In some embodiments, the compound used to treat DNA is a bisulfite compound. In some embodiments, the assay comprises using a
methylation-sensitive restriction enzyme. In some embodiments, the methylation-sensitive restriction enzyme is selected from HpaII SmaI, Saell, Eagl, BstUI, and BssHII. In some embodiments, the sample is a bodily fluid selected from the group consisting of blood, saliva, spit, an esophageal washing, serum, plasma, a blood-derived fraction, stool, urine, and a colonic effluent. In some embodiments, the sample is derived from a tissue, In some embodiments, the sample is a biopsy. In some embodiments, the sample is a brushing.
In some embodiments, the disclosure provides for a method of monitoring over time an esophageal neoplasia or metaplasia in the esophagus comprising: a) detecting the presence of meihylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 -· 8424, or complements or fragments thereof:, or complements or fragments thereof; from a sample from a subject for a first time; and b) detecting the level of methylaiion of the nucleotide sequence in a sample from the same subject at a later time. In some embodiments, the disclosure provides for a method of monitoring over time an esophageal neoplasia or metaplasia in the esophagus comprising: a) detecting the meihylation status of DNA by assay in a bisulfite converted DNA for retention of a cytosine base at one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 857-1284, .1713-2140, 3425-3852, 4281-4708, 5927-6321 , 6717-71 1 1 , 7559-7584, 761 1-7636, 7715-7740, 7767- 7792, 7867-7890, 7915-7938, 7991 -8004, 8019-8032, 8075-8088, 8103-8116, 8157-8169, 8183-8195, 8223-8236, 8265-8278, 8307-8320, 8349-8362, 8410-8414 or 8425-8429 from a sample from a subject for a first time; and b) detecting the methylation status of the nucleotide sequence in a sample from the same subject at a later time; wherein absence of methylation in the nucleotide sequence taken at a later time and the presence of methylation in the nucleotide sequence taken at the first time is indicative of neoplasia or metaplasia regression; and wherein presence of methylation in the nucleotide sequence taken at a later time and the absence of methylation in the nucleotide sequence taken a t the first time is indicative of neoplasia or metaplasia progression. In some embodiments, the disclosure provides for a method of monitoring over time an esophageal neoplasia comprising: a) de tecting the presence of methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 1 1561 -11662, 1 1 867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 1258142586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or complements or fragments thereof; from a sample from a subject for a first time: and h) detecting the level of methylation of the nucleotide sequence in a sample from the same subject at a later time. In some embodiments, the disclosure provides for a method of monitoring over time an esophageal neoplasia comprising: a) detecting the methylation status of a DNA molecule by assay in a bisulfite converted DNA for retention of a cytosine base of one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 8819-9004, 9191-9376, 9935-10120, 10307- 10492, 10973-1 1 1 19, 1 1267-1 1413, 11663-1 1713, 1 1765-11815, 1 1969-12019, 12071-12121 , 12267-12313, 12361-12407, 12467-12472, 12479-12484, 12503-12508, 12515-12520, 12539-12544, 12551 -12556, 12569-12574, 12587-12592, 12605-12610, 12623-12628, 12650-12652, or 12659-12661 from a sample from a subject for a first time: and b) detecting the methylation status of the nucleotide sequence in a sample from the same subject at a later time; wherein absence of methylation in the nucleotide sequence taken at a later time and the presence of methylation in the nucleotide sequence taken at the first time is indicative of neoplasia regression; and wherein presence of methylation in the nucleotide sequence taken at a later time and the absence of methylation in the nucleotide sequence taken at the first time is indicative of neoplasia progression, in some embodiments, the sample is a bodily fluid selected from the group consisting of blood, saliva, spit or an esophageal washing. In some embodiments, the sample is derived from tissue.
In some embodiments, the disclosure provides for a method of treating a subject having a metaplasia in the esophagus or neoplasia, comprising the step of treating the subject with chemotherapy , radia tion therapy and/or with the resection of the neoplasia or metaplasia; and/or with ablation of the neoplasia or metaplasia; wherein the subject has been determined to have methylation in a sequence tha t is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 · · 8306, 8335 -· 8348, 8405 - 8409, or 8420 - 8424, or complements or fragments thereof. In some embodiments, the disclosure provides for a method of treating a subject having a metaplasia in the esophagus or neoplasia, comprising the step of treating the subject with chemotherapy, radiation therapy and/or with the resection of the neoplasia or metaplasia; and/or with ablation of the neoplasia or metplasia; wherein the subject has been determined to have DNA methylation by assay in a bisulfite converted DNA for retention of a cytosine base of one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 857-1284, 1713-2140, 3425-3852, 4281-4708, 5927-6321 , 6717-71 1 1 , 7559-7584, 761 1-7636, 7715-7740, 7767- 7792, 7867-7890, 7915-7938, 7991 -8004, 8019-8032, 8075-8088, 8103-8116, 8157-8169, 8183-8195, 8223-8236, 8265-8278, 8307-8320, 8349-8362, 8410-8414 or 8425-8429. In some embodiments, the disclosure provides for a method of treating a subject having an esophageal neoplasia, comprising the step of treating the subject with chemotherapy, radiation therapy, ablation, and/or with neoplasia resection; wherein the subject has been determined to have methylation methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 1 1561-11662, 1 1867- 1 1968, 12173-12266, 12455-12466, 12491 -12502, 12527- 12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or complements or fragments thereof. In some embodiments, the disclosure provides for a method of treating a subject having an esophageal neoplasia, comprising the step of treating the subject with
chemotherapy, radiation therapy, ablation, and/or with neoplasia resection; wherein the subject has been determined to ha ve rnethylation of a DNA molecule by assay in a bisulfite converted DNA for retention of a cytosine base of one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of; SEQ ID NOs: 8819-9004, 9191 -9376, 9935-10120, 10307-10492, 10973- 11119, 1 1267-1 1413, 11663-11713, 1 1765-11815, 11969-12019, 12071 -12121, 12267-12313, 12361-12407, 12467-12472, 12479-12484, 12503-12508, 12515-12520, 12539-12544, 12551-12556, 12569-12574, 12587-12592, 12605-12610, 12623-12628, 12650-12652, or 12659-12661. In some embodiments, the subject has been assayed for having a somatic mutation in TPS 3.
In some embodiments, the disclosure provides for a bisulfite converted sequence having at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136,
5927-7506, 7559 - 7662, 77.15 - 7818, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434, and/or fragments thereof, and/or the reverse complements thereof including all unique fragments of these sequences and their reverse complements, in some embodiments, the disclosure provides for a panel of bisulfite converted sequences selected from the sequences having at least 90% identity to any one or more of SEQ IX) NOs: 857 -2568, 3425-5136, 5927-7506, 7559 - 7662, 7715 - 7818, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434, and/or fragments thereof, and/or the reverse complements thereof including all unique fragments of these sequences and their reverse complements, in some embodiments, the panel corresponds to any combination of the sequences having any of SEQ I'D NOs: 8307-8334, 8349-8376, 8410- 8419 or 8425-8434. In some embodiments, the disclosure provides for an oligonucleotide primer that hybridizes to any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136, 5927-7506, 7559 - 7662, 7715 - 7S1 8, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434. In some embodiments, the primer is any of the primers having at least 90% identity to any one or more of SEQ ID NOs: 8377-8404 or 8435-8444, or fragments or complements thereof. In some embodiments, the primers are combined as forward and reverse primers for PGR amplification of any of the bisulfite converted sequences having at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136,
5927-7506, 7559 - 7662, 7715 - 7818, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434, and/or fragments thereof, and/or the reverse complements thereof including all unique fragments of these sequences and their reverse complements. In some embodiments, one of the primers in the primer pairs has a nucleotide sequence that is at least 90% identity to any one or more of SEQ ID NOs: 8377-8404 or 8435-8444, or fragments or complements thereof. In some embodiments, the disclosure provides for a panel of primer pairs selected from any of the primer pairs disclosed herein. In some embodiments, the panel corresponds to a combination of primer pairs for amplifying any of the combinations of SEQ ID NOs: 8307-8334, 8349- 8376, 8410-8419 or 8425-8434.
In some embodiments, the disclosure provides for a bisulfite converted sequence having at least 90% identity to any one or more of SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 11560; 1 1663 - 11866, 1 1969 - 12172, 12267 - 12454; 12467 - 12490, .12503 - 12526, 12539 - 12562, 12569 - 12580, 12587 - 12598, 12605 - 12616, 12623 - 12634, 12650 - 12655, and 12659 - 12664, and the reverse complements thereof, including all unique fragments of these sequences and their reverse complements. In some embodiments, the disclosure provides for a pane! of bisulfite converted sequences selected from any of these sequences. In some embodiments, the disclosure provides for an oligonucleotide primer that hybridizes to any of these sequences. In some embodiments, the panel corresponds to any combination of any of the sequences of SEQ ID NOs: 12605-12616, 12623-12634, 12650- 12655, or 12659-12664. In some embodiments, the disclosure provides for primers having at least 90% identity to any one or more of SEQ ID NOs: 12635-12646, and 12665-12670, or fragments or complements thereof. In some embodiments, the primers are combined as forward and reverse primers for PGR amplification of any of the bisulfite converted sequences having at least 90% identity to any one or more of SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 11560; 1 1663 - 11866, 1 1969 - 12172, 12267 - 12454; 12467 -· 12490, 12503 - 12526, 12539 - 12562, 12569 - 12580, 12587 - 12598, 12605 - 12616, 12623 - 12634, 12650 - 12655, and 12659 - 12664, and the reverse complements thereof, including all unique fragments of these sequences and their reverse complements, in some embodiments, one of the primers in the primer pairs has a nucleotide sequence that is at least 90% identity to any one or more of SEQ ID NOs: 12635-12646, and 12665-12670, or fragments or complements thereof. In some embodiments, the disclosure provides for a panel of primer pairs selected from any of the primer pairs disclosed herein. In some embodiments, the panel corresponds to the combination of primer pairs for amplifying any combination of any of the sequences of SEQ ID NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664.
in some embodiments, the disclosure provides for a method of selecting an individual to undergo a diagnostic procedure to determine the presence of Barrett's esophagus, of Barrett's esophagus with low grade dysplasia, of Barrett's esophagus with high grade dysplasia, or of esophageal adenocarcinoma, by obtaining a biological sample from an individual , and determining the presence in DNA from that sample of DNA methylation present in any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, and/or fragments thereof, and/or the reverse complements thereof. In some embodiments, the method further comprises determining the presence of a somatic mutation in TP53, In some embodiments, DNA methylation is detected by cutting one of the DNA sequences with a methylation sensitive restriction enzyme. In some embodiments, DNA methylation is detected by bisulfite converting DNA from the sample and detecting the presence of any of the bisulfite converted DNA sequences ha ving at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136, 5927-7506, 7559 - 7662, 7715 - 7818, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434, and/or fragments thereof and/or the reverse complements thereof including all unique fragments of these sequences and their reverse complements. In some embodiments, the bisulfite converted sequences are detected using any of: DNA sequencing, next generation sequencing, methylation specific PGR, methylation specific PGR combined with a fluorogenic hybridization probe, and real time methylation specific PGR. In some embodiments, the bisulfite converted sequences are detected using PGR amplification employing a PGR primer or primer pair comprising the nucleotide sequence of any of SEQ ID NOs: 8377-8404 or 8435-8444, in some embodiments, the method further comprises the step of determining the nucleotide sequence of the bisulfite converted sequences. In some embodiments, the percent of the target sequence that is methylated in any of the individual target sequences is greater than 1%, or greater than 5%, or greater than 10%. In some embodiments, the biological sample is a tissue sample, including a sample of the esophagus. In some embodiments, the tissue sample is a biopsy or a brushing, including a biopsy or a brushing of the esophagus. In some embodiments, the biological sample is a body fluid, in some embodiments, the body fluid is blood, saliva, spit or an esophageal washing,
In some embodiments, the disclosure provides for a method of selecting an individual to undergo a diagnostic procedure to determine presence of Barrett's esophagus with low grade dysplasia, of Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma; or to undergo a treatment for for Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia or for esophageal adenocarcinoma; or to undergo enhanced surveillance for the development of Barrett's esophagus with low grade dysplasia, of Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma, by obtaining a biological sample irorn an individual, and determining the presence in DNA from that sample of methylation present in any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 1 1561-1 1662, 1 18674 1968, 12173-12266, 12455-12466, 12491 -1.2502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658. In some embodiments, the disclosure provides for a method of treating a subject having Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia or esophageal adenocarcinoma, comprising the step of treating the subject with chemotherapy, radiation therapy, and/or with resection of an esophageal lesion or with ablation of an esophageal lesion, wherein the subject has been determined to have methylation at any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818, 9563- 9934, 10679-10826, 1 1561 -11662, 1 1867-1 1968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658. In some embodiments, the disclosure provides for a method of selecting an individual to undergo enhanced surveillance for the development of Barren's esophagus with high grade dysplasia or of esophageal adenocarcinoma, wherein the subject has been determined to have rnethylation at any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818, 9563-9934, 10679-10826, 1 1561-11662, 11867- 11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658. In some embodiments, the methods disiosed herein fuxther comprises determining the presence of a somatic mutation in TP53. In some embodiments, DNA rnethylation is detected by cutting one of the DNA sequences with a rnethylation sensitive restriction enzyme, in some embodiments, DNA rnethylation is detected by bisulfite converting DNA from the sample and detecting the presence of any of the bisulfite converted DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 1 1560: 1 1663 - 11866, 11969 - 12172, 12267 - 12454; 12467 - 12490, 12503 - 12526, 12539 - 12562, 12569 - 12580, 12587 - 12598, 12605 - 12616, 12623 - 12634, 12650 - 12655, and 12659 - 12664, and the reverse complements thereof, including all unique fragments of these sequences and their reverse complements or panels comprising any combination of those sequences, or detecting the presence of uncoverted cytosine residues at any of the Y bases in any of those sequences or panels. In some embodiments, the bisulfite converted sequences or the presence of unconverted cytosine residues at any of the Y bases of these sequence or panels are detected using any of: DNA sequencing, next generation sequencing, rnethylation specific PGR, rnethylation specific PGR combined with a iluorogemc hybridization probe, and real time rnethylation specific PGR. In some embodiments, the bisulfite converted sequences or the presence of unconverted cytosine residues in the bisulfite converted sequences are detected using PGR amplification employing any of the PGR primers or primer pairs disclosed herein. In some embodiments, the percent of the target sequence that is methylated in any of the individual target sequences is greater than 1%, or greater than 5%, or greater than 10%. In some embodiments, the biological sample is a tissue sample, including a sample of the esophagus. In some embodiments, the tissue sample is a biopsy or a brushing, including a biopsy or brushing of the esophagus, i n some embodiments, the biological sample is a body fluid, in some embodiments, the body fluid is blood, saliva, spit or an esophageal washing, in some embodiments, the individual is known to have Barrett's esophagus, In some embodiments, the individual is known to have Barrett's esophagus with low grade dysplasia,
in some embodiments, the disclosure provides for a method of determining the response of an individual with esophageal cancer to therapy by detection in a body fluid of methylation in any of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 11561-1 1662, 1 1867- 1 1968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658 and/or fragments thereof, and/or the reverse complements thereof; wherein increasing levels of methylation over time are indicati ve of disease progression and a need for a change in therapy, and wherein absence of increase in levels of methylation over time or decrease in levels of mediylation over time are indicative that a change in therapy is not required. In some embodiments, any of the methods disclosed herein further comprise detecting the presence of a somatic mutation in TP53. In some embodiments, the DNA methylation is detected by bisulfite converting DNA from a body fluid and detecting the presence of any of the bisulfite converted DNA sequences disclosed herein.
In some embodiments, the disclosure provides for a method of monitoring over time an esophageal neoplasia comprising: a) detecting in a sample from a subject for a first time (i) the methylation status of one or more of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 -- 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 1 1561-1 1662, 1 1867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581 -12586, 12599-12604, 1261 7-12622, 12647-12649 or 12656-12658 from thai sample; and (ii) the somatic mutation status of TP53 from that sample; and b) detecting in a sample from the same subject at a later time (i) the methylation status of one or more Of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 1 1561-11662, 11867-1 1968, 1217342266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658; and (ii) the somatic mutation status of TP53 from the same subject at a later time, in some embodiments, the neoplasia regression is indicated by: (i) the sample taken at the first time having methylation in any of Up15-L Up35-1, Up35~2, Up3, Up27, or Up1O, or having a somatic mutation in TP53; and (ii) the sample at a later time lacking methylation in Up1 5-I , Up35-1 , Up35-2, Up3, Up27, Up10, and lacking a somatic mutation in TPS 3. In some embodiments, the neoplasia progression is indicated by: (i) the sample taken at the first time lacking methylation in Up] 5-1, Up35-1, Up35-2, Up3, Up27, and Up10, and lacking a somatic mutation in TP53; and (ii) the sample taken at a later time having methylation in any of Up15-1 , Up35~I, Up35-2, Up3, Up27, Up10, or having a somatic mutation in TP53. in some embodiments, the biological sample is a tissue sample, including a sample of the esophagus. In some embodiments, the tissue sample is a biopsy or a brushing, including a biopsy or a brushing of the esophagus. In some embodiments, the biological sample is a body fluid, in some embodiments, the body fluid is blood, saliva, spit or an esophageal washing.
In some embodiments, the disclosure provides for a method of detecting the presence of an esophageal neoplasia in a human subject, comprising: a) obtaining a human sample; and b) detecting the methylation status of one or more of the DNA sequences ha ving at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679- 10826, 1 1561 -11662, 11867-1 1968, 12173-12266, 12455-12466, 12491 -12502, 12527-12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658 from that sample. In some embodiments, the method further comprises the step of detecting the somatic mutation status of TPS 3 from that sample. In some embodiments, the TPS 3 somatic mutation is detected at a later time than the detection of the .methylation status of any of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 1 1561-11662, 1 1867-1 1968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, In some embodiments, a) the presence of methylation at any of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293
- 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424; and b) the absence of methylation at any one of the combination of Up1 5-1 , Up35-1 , Up35-2, Up3, Up27, and Up 10; and c) the absence of a somatic mutation in TP53 is indicative of non-dysplastic Barret's esophagus. In some embodiments, a) the presence of methylation at any of SEQ ID NOs: 1 -- 856, 2569 - 3424, 5137 - 5926: 7507 - 7558, 7663 ~ 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 --- 8409, 8420 - 8424; and b) the absence of methylation in all members of an assay panel selected from any or all of Up15-1 , Up35-1, Up35-2, Up3, Up27, and Up10; and c) the absence of a somatic mutation in TP53 is indicative of non-dysplastic Barret's esophagus. In some embodiments, a) the presence of methylation at any of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251
- 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 -- 8424; and b) the absence of methylation at Up-3 and Up35-2; and c) the absence of a somatic mutation in TP53 is indicative of non-dysplastic Barret's esophagus. In some embodiments, the presence of methylation at any one of Up15-1 , Up.35~l , Up35-2, Up3, Up27, and Up 10; or the presence of a somatic mutation in TPS 3 is indicative of esophageal adenocarcinoma, of Barrett's esophagus with low grade dysplasia, or of Barrett's esophagus with high grade dysplasia. In some embodiments, the presence of methylation at any one of Up15-1 , Up35-I, Up35~2, Up3, Up27, or Up 10, is indicative of esophageal adenocarcinoma, of Barrett's esophagus with low grade dysplasia, or of Barrett's esophagus with, high grade dysplasia. In some embodiments, the biological sample is a tissue sample, including a sample of the esophagus, in some embodiments, the tissue sample is a biopsy or a brushing, including a. biopsy or a brushing of the esophagus, in some embodiments, the biological sample is a body fluid, in some embodiments, the body fluid is blood, saliva, spit or an esophageal washing.
In some embodiments, the disclosure provides for a bisulfite-converted nucleotide sequence comprising the bisulfite-converted nucleotide sequence of any one of the following: Up3, Up10, Up1 5-1, Up1 5-2, Up20-1 , Up20-2, Up20-2, Up27, Up35-1, Up35-2, SqBE2, SqBES, SqBE7, SqBE9, SqBE1 O, SqBE1 l-1, SqBE1 l-2, SqBE13, SqBE14-2, SqBE1S, SqBE16-l, SqBE16-2, SqBE17-l, SqBE1 S, 3qBE22~l, SqBE22-2 or SqBE23. in some embodiments, the bisulfite-converted nucleotide sequence of claim 98, wherein the sequence comprises the bisulfite-converted nucleotide sequence of any one of the follo wing: Up3, Up10, Up1 5-1} Up15~2, Up20-1, Up20-2, Up20-2, Up27, Up35-1, or Up35-2. In some embodiments, the bisulfite-converted nucleotide sequence of claim 98, wherein the sequence comprises the bisulfite-converted nucleotide sequence of any one of the following: SqBE2, SqBES, SqBE7, SqBE9, SqBE1O, SqBEI 1-1, SqBE1 1-2, SqBEB, SqBE14-2, SqBE1S, SqBE16-l, SqBE16-2, SqBE174, SqBE1 S, SqBE22-l, SqBE22-2 or SqBE23.
In some embodiments, the disclosure provides for a bisulfite-converted nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8307-8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8410, 841 1, 8412,
8414, 8415, 841.6, 8417, 8419, 8425, 8426, 8427, 8429, 8430, 8431, 8432, 8434, 12605- 12616, 12623-12634, 12650-12655, or 12659-12664. In some embodiments, the sequence comprises a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8307- 8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371 -8376, 8410, 841 1, 8412, 8414,
8415, 8416, 8417, 8419, 8425, 8426, 8427, 8429, 8430, 8431 , or 8432, 8434. In some embodiments, the sequence comprises a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664. In some embodiments, the sequence comprises a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: 12652, 12655, 12661 , and 12664.
Brief Description of the Figures
Figure 1 shows the sensitivity (percentage of samples detected), the specificity
(percentage of samples not detected), the total number of samples studied, and the total number of positive samples from the panel of markers studied in Example 3. Three panels of markers were selected for study. Detailed Description Of The Invention
I. Definitions
For convenience, certain terms employed in the specification, examples, and appended claims are collected here. Unless defined otherwise, ail technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. The materials, methods and examples are illustrative only, and are not intended to be limiting. All publications, patents and other documents mentioned herein are incorporated by reference in their entirety.
Each embodiment of the invention described herein may be taken alone or in combination with one or more other embodiments of the invention.
Throughout this specification, the word ''"comprise" or variations such as "comprises" or "comprising" will be understood to imply the inclusion of a stated integer or groups of integers but not the exclusion of any other integer or group of integers.
The articles "a" and "an" are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element. The terms "adenoma" is used herein to describe any precancerous neoplasia or benign tumor of epithelial tissue, for example, a precancerous neoplasia of the gastrointestinal tract, pancreas, and/or the bladder.
The term "blood-derived fraction" herein refers to a component or components of whole blood. Whole blood comprises a liquid portion (i.e., plasma) and a solid portion (i.e., blood cells). The liquid and solid portions of blood are each comprised of multiple components; e.g., different proteins in plasma or different cell types in the solid portion. One of these components or a mixture of any of these components is a blood-derived fraction as long as such fraction is missing one or more components found in whole blood.
The term "esphophagus" is intended to encompass the upper portion of the digestive system spanning from the back of the oral cavity, passing downwards through the rear part of the mediastinum, through the diaphragm and into the stomach.
The term "esophageal cancer" is used herein to refer to any cancerous neoplasia of the esophagus.
"Barrett's esophagus" as used herein refers to an abnormal change (metaplasia) in the cells of the lower portion of the esophagus. Barrett's is characterized the finding of intestinal metaplasia in the esophagus,
A "brushing" of the esophagus, as referred to herein, may be obtained using any of the means known in the art. In some embodiments, a brushing is obtained by contacting the esophagus with a brush, a sponge, a balloon, or with any other device or substance that contacts the esophagus and obtains an esophageal sample.
"Cells," "host cells" or "recombinant host cells" are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cel l. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
The terms "compound", "test compound," "agent", and "molecule" are used herein interchangeably and are meant to include, but are not limited to, peptides, nucleic acids, carbohydrates, small organic molecules, natural product extract libraries, and any other molecules (including, hut not limited to, chemicals, metals, and organometallic compounds),
The term "compound-converted DNA" herein refers to DNA that has been treated or reacted with a chemical compound that, converts unmethylated C bases in DNA to a different nucleotide base. For example, one such compound is sodium bisulfite, which converts unmethylated C to U. If DNA that contains conversion-sensitive cytosme is treated with sodium bisulfite, the compound-converted DNA will contain U in place of C, If the DNA which is treated with sodium bisulfite contains only methylcytosine, the compound-converted DNA will not contain uracil in place of the methylcytosine,
The term "de-methylating agent" as used herein refers agents that restore activity and/or gene expression of target genes silenced by niethylation upon treatment with the agent. Examples of such agents include without limitation 5-azacytidme and 5-aza-2'-deoxycytidine.
The term "detection" is used herein to refer to any process of observing a marker, or a change in a marker (such as for example the change in the niethylation state of the .marker), in a biological sample, whether or not the marker or the change in the marker is actually detected. In other words, the act of probing a sample for a marker or a change in the marker, is a "detection" even if the marker is determined to be not present or below the level of sensitivity. Detection may be a quantitative, semi-quantitative or non-quantitative
observation.
The term "differentially methylated nucleotide sequence" refers to a region of a genomic loci that is found to be methylated in a in cancer tissues or cell lines, but. not methylated in the normal tissues or cell lines,
The term "neoplasia" as used herein refers to an abnormal growth of tissue. As used herein, the term "neoplasia" may be used to refer to cancerous and non-cancerous tumors, as well as to Barrett's esophagus (which may also be referred to herein as a metaplasia) and Barrett's esophagus with dysplasia. In some embodiments, the Barrett's esophagus with dysplasia is Barrett's esophagus with high grade dysplasia, in some embodiments, the Barrett's esophagus with dysplasia is Barrett's esophagus with low grade dysplasia. In some embodiments, the neoplasia, is a cancer (e.g. , esophageal adenocarcinoma). "Gastrointestinal neoplasia" refers to neoplasia of the upper and lower gastrointestinalact. As commonly understood in the art, the upper gastrointestinal tract includes the ophagus, stomach, and duodenum; the lower gastrointestinal tract includes the remainder of e small intestine and all of the large intestine.
The terms "healthy", "normal." and "non-neoplastic" are used interchangeably herein refer to a subject or particular cell or tissue that is devoid (at least to the limit of detection)f a disease condition, such as a neoplasia.
"Homology" or "identity" or "similarity" refers to sequence similarity between twoeptides or between two nucleic acid molecules. Homology and identity can each beetermined by comparing a position in each sequence which may be aligned for purposes ofomparison. When an equivalent position in the compared sequences is occupied by the samease or amino acid, men the molecules are identical at that position; when the equivalent siteccupied by the same or a similar amino acid residue (e.g., similar in steric and/or electronicature), then the molecules can be referred to as homologous (similar) at that position.
xpression as a percentage of homology/similarity or identity refers to a function of theumber of identical or similar amino acids at positions shared by the compared sequences. A quence which is "unrelated or "non-homologous" shares, in some embodiments, less than0% identity, and in particular embodiments, less than 25% identity with a sequence of the esent invention. In comparing two sequences, the absence of residues (amino acids orucleic acids) or presence of extra residues also decreases the identity and
omology/ similarity.
The term "homology" describes a mathematically based comparison of sequencemilarities which is used to identify genes or proteins with similar functions or motifs. Theucleic acid and protein sequences of the present invention may be used as a "query quence" to perform a search against public databases to, for example, identify other family embers, related sequences or homologs. Such searches can be performed using the
BLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) JMol Biol.
5:403-10. BLAST nucleotide searches can be performed with the NBLAST program, ore=100, wordlength=12 to obtain nucleotide sequences homologous to nucleic acid olecules of the invention, BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res.
25(17):3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and BLAST) can be used. See www.ncbi.nim.nih.gov.
As used herein, "identity" means the percentage of identical nucleotide or amino acid residues at corresponding positions in two or more sequences when the sequences are aligned to maximize sequence matching, i.e., taking into account gaps and insertions. Identity can be readily calculated by known methods, including but not limited to those described in
(Computational Molecular Biology, Lesk, A, M, ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993: Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H, G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M, and
Devereux, J., eds.. M Stockton Press, New York, 1991 ; and Carillo, H,, and Lipraan, D,, SIAM J. Applied Math., 48: 1073, 1988). Methods to determine identity are designed to give the largest match between the sequences tested. Moreover, methods to determine identity are codified in publicly available computer programs. Computer program methods to determine identity between two sequences include, but are not limited to, the GCG program package (Devereux, J., et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, and FAST A (Altschul, S. F. et al., J. Moke. Biol. 215: 403-410 (1990) and Altschul et al. Nuc. Acids Res. 25: 3389-3402 (1997)). The BLAST X program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S., et al, NCBI NLM NIH Bethesda, Md.
20894; Altschul, S., et a!., J. Mol. Biol. 215: 403-410 (1990». The well known Smith Waterman algorithm may also be used to determine identity.
The term "including" is used herein to mean, and is used interchangeably with, the phrase "including but not limited to."
The term "isolated" as used herein with respect to nucleic acids, such as DNA or RNA, refers to molecules in a form which does not occur in nature. Moreover, an "isolated nucleic acid" is meant to include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state.
The term "methylation-specific PGR" ("MSP") herein refers to a polymerase chain reaction in which amplification of the compound-converted template sequence is performed. Two sets of primers are designed for use in MSP. Each set of primers comprises a forward primer and a reverse primer. One set of primers, called methylation-specific primers (see below), will amplify the compound-converted template sequence if C bases in CpG dinucleotides within the DNA are methylated. Another set of primers, called unmethylati on- specific primers or primers for unmethylated sequences and the like (see belowr), will amplify the compound-converted template sequences if C bases in CpG dinucleotides within the DNA are not methylated.
As used herein, the term "nucleic acid" refers to polynucleotides such as
deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA), The term should also be understood to include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single- stranded (such as sense or antisense) and double-stranded polynucleotides.
"Operably linked" when describing the relationship between two DNA regions simply means that they are functionally related to each other. For example, a promoter or other transcriptional regulatory sequence is operably linked to a coding sequence if it controls the transcription of the coding sequence.
The term "or" is used herein to mean, and is used interchangeably with, the terra "and/or", unless context clearly indicates otherwise.
The terms "proteins" and "polypeptides" are used interchangeably herein.
A "sample" includes any material that is obtained or prepared for detection of a molecular marker or a change in a molecular marker such as for example the methylation state, or any material that is contacted with a detection reagent or detection device for the purpose of detecting a molecular marker or a change in the molecular marker.
As used herein, "obtaining a sample" includes directly retrieving a sample from a subject to be assayed, or directly retrieving a sample from a subject to be stored and assayed at a later time. Alternatively, a sample may be obtained via a second party. That is, a sample may be obtained via, e.g., shipment, from another individual who has retrieved the sample, or otherwise obtained the sample.
A "subject" is any organism of interest, generally a mammalian subject, such as a mouse, and in particular embodiments, a human subject,
As used herein, the term "specifically hybridizes" refers to the ability of a nucleic acid probe/primer of the invention to hybridize to at least 12, 15, 20, 25, 30. 35, 40, 45, 50 or 100 consecutive nucleotides of a target sequence, or a sequence complementary thereto, or naturally occurring mutants thereof, such that it has, in some embodiments, less than 15%, less than 10%, or less than 5% background hybridization to a cellular nucleic acid (e.g., mRNA or genomic DNA) other than the target gene. A variety of hybridization conditions may be used to detect specific hybridization, and the stringency is determined primarily by the wash stage of the hybridization assay. Generally high temperatures and low salt
concentrations give high stringency, while low temperatures and high salt concentrations give low stringency. Low stringency hybridization is achieved by washing in, for example, about 2.0 x SSC at 50 °C, and high stringency is achieved with about 0,2 x SSC at 50 °C. Further descriptions of stringency are provided below.
As applied to polypeptides, the term "substantial sequence identity" means that two peptide sequences, when optimally aligned such as by the programs GAP or BESTFIT using default gap, share at least 90 percent sequence identity, in some embodiments, at least 95 percent sequence identity, or at least 99 percent sequence identity or more, in some embodiments, residue positions which are not identical differ by conservative amino acid substitutions. For example, the substitution of amino acids having similar chemical properties such as charge or polarity is not likely to affect the properties of a protein. Examples include glutamine for asparagine or glutamic acid for aspartic acid.
An "informative loci" as used herein, refers to any of the nucleic acid sequences disclosed herein that may have altered (e.g. , increased) methylation in a sample (e.g., an esophageal tissue sample) from a subject having Barrett's esophagus and/or an esophageal neoplasia as compared to the methylation patterns of the corresponding nucleic acid sequence in a sample from a healthy control subject. The term "Up3" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12563, 12581, 12599, 12617, or fragments or reverse complements thereof. In some embodiments, the Up3 sequence refers to a bisuliite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%>, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12569, 12587, 12605 or 12623, or fragments or reverse complements thereof, in some embodiments, the Up3 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12575, 12593, 1261 1 or 12629, or fragments or reverse complements thereof, hi some embodiments, the Up3 sequence may he amplified using primers comprising the sequence of SEQ ID NOs: 12635 and/or 12641, or fragments or reverse complements thereof.
The term "Up 10" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%*, 92%, 93%), 94%, 95%, 96%, 97%, 98%, 99% or
100% identity to the sequence of SEQ ID NO: 12564, 12582, 12600 or 1261 8, or fragments or reverse complements thereof. In some embodiments, the Up10 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12570, 12588, 12606 or 12624, or fragments or reverse complements thereof In some embodiments, the Up 10 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12576, 12594, 12612 or 12630, or fragments or reverse complements thereof. In some embodiments, the Up10 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12636 and/or 12642, or fragments or reverse complements thereof,
The term "Up1 5-1 " as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%,, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12565, 12583, 12601 or 12619, or fragments or reverse complements thereof. In some embodiments, the Up15-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12571, 12589, 12607 or 12625, or fragments or reverse complements thereof. In some embodiments, the Up15-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 32577, 12595, 12613 or 12631, or fragments or reverse complements thereof. In some embodiments, the Up 15-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12637 and/or 12643, or fragments or reverse complements thereof
The term "Up 15-2" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12565, 12583, 12647 or 12656, or fragments or reverse complements thereof, in some embodiments, the Up] 5-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12571, 12589, 12650 or 12659, or fragments or reverse complements thereof. In some embodiments, the Up] 5-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12577, 12595. 12653 or 12662, or fragments or reverse complements thereof. In some embodiments, the Up15-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12665 and/or 12668, or fragments or reverse complements thereof.
The term "Up20-1 " as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12566, 12584, 12602 or 12620, or fragments or reverse complements thereof. In some embodiments, the Up20-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12572, 12590. 12608 or 12626, or fragments or reverse complements thereof. In some embodiments, the Up20-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12578, 12596,
12614 or 12632, or fragments or reverse complements thereof. In some embodiments, the ϋρ20-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12638 and/or 12644, or fragments or reverse complements thereof.
The term "Up20-2" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12566, 12584, 12648 or 12657, or fragments or reverse complements thereof In some embodiments, the Up20-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12572, 12590, 12651 or 12660, or fragments or reverse complements thereof In some embodiments, the Up20-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12578, 12596, 12654 or 12663, or fragments or reverse complements thereof. In some embodiments, the Up20-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12666 and/or 12669, or fragments or reverse complements thereof
The term "Up27" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or
100% identity to the sequence of SEQ ID NO: 12567, 12585, 12603 or 12621, or fragments or reverse complements thereof, in some embodiments, the Up27 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12573, 12591, 12609 or 12627, or fragments or reverse complements thereof. In some embodiments, the Up27 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12579, 12597,
12615 or 12633, or fragments or reverse complements thereof, In some embodiments, the Up27 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12639 and/or 12645, or fragments or reverse complements thereof,
The term "Up35-1" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12568, 12586, 12604 or 12622, or fragments or reverse complements thereof. In some embodiments, the Up35-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%», 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12574, 12592, 12610 or 12628, or fragments or reverse complements thereof. In some embodiments, the Up35~l sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12580, 12598, 12616 or 126.34, or fragments or reverse complements thereof. In some embodiments, the Up35-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12640 and/or 12646, or fragments or reverse complements thereof.
The term "Up35-2" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12568, 12586, 12649 or 12658, or fragments or reverse complements thereof. In some embodiments, the Up35-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 12574, 12592, 12652 or 12661 , or fragments or reverse complements thereof. In some embodiments, the Up35-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ I D NO: 12580, 12598, 12655 or 12664, or fragments or reverse complements thereof In some embodiments, the Up35-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 12667 and/or 12670, or fragments or reverse complements thereof.
The term "SqBE 2" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8209, 8251 , 8293 or 8335, or fragments or reverse complements thereof. In some embodiments, the SqBE 2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ I D NO: 8223, 8265, 8307 or 8349, or fragments or reverse complements thereof. In some embodiments, the SqBE 2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%», 85%, 90%, 91%, 92%, 93%, 94%>, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8237, 8279,
8321 or 8363, or fragments or reverse complements thereof. In some embodiments, the SqBE 2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8377 and/or 8391, or fragments or reverse complements thereof.
The term "SqBE 5" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8210, 8252, 8294 or 8336, or fragments or reverse complements thereof. In some embodiments, the SqBE 5 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8224, 8266, 8308 or 8350, or fragments or reverse complements thereof. In some embodiments, the SqBE 5 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8238, 8280,
8322 or 8364, or fragments or reverse complements thereof. In some embodiments, the SqBE 5 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8378 and/or 8392, or fragments or reverse complements thereof.
The term "SqBE 7" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 821 1 , 8253, 8295 or 8337, or fragments or reverse complements thereof. In some embodiments, the SqBE 7 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%. 98%, 99% or 1 00% identity to the sequence of SEQ ID NO: 8225, 8267, 8309 or 8351 , or fragments or reverse complements thereof. In some embodiments, the SqBE 7 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8239, 8281, 8323 or 8365, or fragments or reverse complements thereof. In some embodiments, the SqBE 7 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8379 and/or 8393, or fragments or reverse complements thereof. sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%. 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8212, 8254, 8296 or 8338, or fragments or reverse complements thereof, hi some embodiments, the SqBE 9 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%. or 100% identity to the sequence of SEQ ID NO: 8226, 8268, 83 10 or 8352, or fragments or reverse complements thereof. In some embodiments, the SqBE 9 sequence refers to a hi sulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%), 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8240, 8282, 8324 or 8366, or fragments or reverse complements thereof. In some embodiments, the SqBE 9 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8380 and/or 8394, or fragments or reverse complements thereof.
'"(lie term "SqBE 10" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8213, 8255, 8297 or 8339, or fragments or reverse complements thereof. In some embodiments, the SqBE 10 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8227, 8269, 831 1 or 8353, or fragments or reverse complements thereof. In some embodiments, the SqBE S O sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%}, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8241 , 8283, 8325 or 8367, or fragments or reverse complemenis (.hereof. Irs some embodiments, the SqBE 10 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8381 and/or 8395, or fragments or reverse complements thereof
The term "SqBE 1 1-1" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8214, 8256, 8298 or 8340, or fragments or reverse complements thereof. In some embodiments, the SqBE 1 1 -1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%», 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8228, 8270, 8312 or 8354, or fragments OF reverse complements thereof. In some embodiments, the SqBE 1 1 -1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8242, 8284,
8326 or 8368, or fragments or reverse complements thereof. In some embodiments, the SqBE 1 1 -1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs:
8382 and/or 8396, or .fragments or reverse complements thereof.
The term "SqBE 1 1 -2" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8214, 8256, 8405 or 8420, or fragments or reverse complements thereof, in some embodiments, die SqBE- 3 1 -2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8228, 8270, 8410 or 8425, or fragments or reverse complements thereof. In some embodiments, the SqBE 1 1-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8242, 8284, 8415 or 8430, or fragments or reverse complements thereof. In some embodiments, the SqBE 1 1 -2 sequence may be amplified using primers comprising the sequence of SEQ I D NOs: 8435 and/or 8440, or fragmenis or reverse complements thereof. The term "SqBE 13" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8215, 8257, 8299 or 8341 , or fragments or reverse complements thereof. In some embodiments, the SqBE 13 sequence refers to a bisulfite, converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%. 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 1(50% identity to the sequence of SEQ ID NO: 8229, 8271, 8313 or 8355, or fragments or reverse complements thereof, In some embodiments, the SqBE 13 sequence refers to a. bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8243, 8285,
8327 or 8369, or fragments or reverse complements thereof. In some embodiments, the SqBE 13 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8383 and/or 8397, or fragments or reverse complements thereof,
The term "SqBE 14-2" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8216, 8258, 8406 or 8421 , or fragments or reverse complements thereof. In some embodiments, the SqBE 14-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8230, 8272, 841 1 or 8426, or fragments or reverse complements thereof. In some em bodiments, the SqBE 14-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8244, 8286, 8416 or 8431, or fragments or reverse complements thereof. In some embodiments, the SqBE 14-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8436 and/or 8441 , or fragments or reverse complements thereof
The term "SqBE 15" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8217, 8259, 8301 or 8343, or fragments or reverse complements thereof In some embodiments, the SqBE 15 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8231 , 8273, 8315 or 8357, or fragments or reverse complements thereof. In some embodiments, the SqBE 15 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8245, 8287, 8329 or 8371 , or fragments or reverse complements thereof. In some embodiments, the SqBE 15 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8385 and/or 8399, or fragments or reverse complements thereof.
The term "SqBE 16-1" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ I D NO: 8218, 8260, 8302 or 8344, or fragments or reverse complements thereof. In some embodiments, the SqBE 16-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8232, 8274, 8316 or 8358, or fragments or reverse complements thereof. In some embodiments, die SqBE 16-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8246, 8288, 8330 or 8372, or fragments or reverse complements thereof. In some embodiments, the SqBE 16-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8386 and/or 8400, or fragments or reverse complements thereof.
The term "SqBE 16-2" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 821 8, 8260, 8407 or 8422, or fragments or reverse complements thereof. In some embodiments, the SqBE 16-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8232, 8274, 8412 or 8427, or fragments or reverse complements thereof. In some embodiments, the SqBE 16-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8246, 8288, 8417 or 8432, or fragments or reverse complements thereof. In some embodiments, the SqBE
16- 2 sequence may be amplified using primers comprising the sequence of S EQ ID NOs: 8437 and/or 8442, or fragments or reverse complements thereof.
The term "SqBE 17-1" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8219, 8261 , 8303 or 8345, or fragments or reverse complements thereof. In some embodiments, the SqBE 17-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8233, 8275, 8317 or 8359, or fragments or reverse complements thereof. In some embodiments, the SqBE 17-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8247, 8289,
8331 or 8373, or fragments or reverse complements thereof, in some embodiments, the SqBE
17- 1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8387 and/or 8401, or fragments or reverse complements thereof,
The term "SqBE) 18" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8220, 8262, 8304 or 8346, or fragments or reverse complements thereof. In some embodiments, the SqBE 18 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8234, 8276, 8318 or 8360, or fragments or reverse complements thereof. In some embodiments, the SqBE 18 sequence refers to a bisulfite converted product, of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 9(5%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8248, 8290,
8332 or 8374, or fragments or reverse complements thereof. In some embodiments, the SqBE 18 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8388 and/or 8402, or fragments or reverse complements thereof.
The term "SqBE 22-1" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%. 95%, 96%, 97%, 98%,, 99% or 100% identity to the sequence of SEQ ID NO: 8221 , 8263, 8305 or 8347, or fragments or reverse complements thereof. In some embodiments, the SqBE 22-1 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at. least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%· or 100% identity to the sequence of SEQ ID NO: 8235, 8277, 8319 or 8361, or fragments or reverse complements thereof, in some embodiments, the SqBE 22-1 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ I'D NO: 8249, 8291 , 8333 or 8375, or fragments or reverse complements thereof, in some embodiments, the SqBE 22-1 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8389 and/or 8403, or fragments or reverse complements thereof
The term "SqBE 22-2" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8221 , 8263, 8409 or 8424, or fragments or reverse complements thereof. In some embodiments, the SqBE 22-2 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8235, 8277, 8414 or 8429, or fragments or reverse complements thereof. In some embodiments, the SqBE 22-2 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8249, 829 i ,
8419 or 8434, or fragments or reverse complements thereof. In some embodiments, the SqBE 22-2 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8439 and/or 8444, or fragments or reverse complements thereof.
The term "SqBE 23" as used herein refers to a nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%», 95%, 96%, 97%, 98%? 99% or 100% identity to the sequence of SEQ ID NO: 8222, 8264, 8306 or 8348, or fragments or reverse complements thereof. In some embodiments, the SqBE 23 sequence refers to a bisulfite converted nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%>, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8236, 8278, 8320 or 8362, or fragments or reverse complements thereof. In some embodiments, the SqBE 23 sequence refers to a bisulfite converted product of a methylated nucleotide sequence comprising a sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to the sequence of SEQ ID NO: 8250, 8292, 8334 or 8376, or fragments or reverse complements thereof. In some embodiments, the SqBE 23 sequence may be amplified using primers comprising the sequence of SEQ ID NOs: 8390 and/or 8404, or fragments or reverse complements thereof.
In some instances, any of the nucleotide sequences disclosed herein contain one or more "Y" positions. Cytosine residues that may be methylated or umiiethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a€ (if methylated), are designated with a 'Ύ," In some embodiments, a parental nucleotide sequence is fully unmethylated if the sequence comprises a T at every Y position following bisulfite conversion. In some embodiments, a parental nucleotide sequence is fully methylated if the sequence comprises a C at every Y position following bisulfite conversion, in some embodiments, a parental nucleotide sequence is partially methylated if the sequence comprises at least one C at a Y position and at least one T at a Y position of the sequence following bisulfite conversion. In some embodiments, the bisulfite converted sequences disclosed herein comprise at least one C at a Y position and at least one T at a Y position, i.e., the parental sequence is partially methylated. II. Overview
This disclosure, is based at least in part on the recognition that differential methylation of particular genomic loci may be indicative of neoplasia of the upper gastrointestinal tract, e.g., esophagus. The present findings demonstrate that methylation at these genomic loci may be a useful biomarker of neoplasia in the upper gastrointestinal tract. The present findings further demonstrate that the status of methylation at these genomic loci used in combination with the status of somatic mutation(s) in TP53 may be a highly sensitive and specific biomarker of neoplasia in the upper gastrointestinal tract.
In general neoplasias may develop through one of at least three different pathways, termed chromosomal instability, microsatellite instability, and the CpG island methyiator phenotype (CIMP). Although there is some overlap, these pathways tend to present somewhat different biological behavior, By understanding the pathway of tumor
development, the target genes involved, and the mechanisms underlying the genetic instability, it is possible to implement strategies to detect and treat the different types of neoplasias.
This disclosure is based, at least in part on the recognition that certain target genes may be silenced or inactivated by the differential raethylation of CpG islands in the 5 s flanking or promoter regions of the target gene, CpG islands are clusters of cytosine- guanosine residues in a DNA sequence, which are prominently represented in the 5-flanking region or promoter region of about half the genes in our genome. In particular, this application is based at least in part on the recognition that differential methylation of particular genomic loci may be indicative of neoplasia of the upper gastrointestinal tract including, but not limited to, esophageal neoplasia,
Additionally, this disclosure is based, at least in part, on the recognition that somatic mutations in TP53 (e.g. , any of the somatic TP53 mutations disclosed herein), in combination with methylation of certain informative loci as disclosed herein, may serve as useful indicators of neoplasia, including esophageal neoplasia (e.g., esophageal adenocarcinoma). In certain embodiments, the TPS 3 somatic mutation is any of the TP53 mutations disclosed herein, in certain embodiments, the TP53 somatic mutation is any nonsynonymous somatic mutation known in the art, in certain embodiments, the TP53 somatic mutation is any one or more mutation at any one or more amino acid residue corresponding to amino acid residue 72, 105, 108, 1 10, 1 13, 124, 127, 132, 144, 152, 163, 175, 183, 194, 213, 214, 218, 232, 234, 248, 265, 273, 278, 306, 337, 347, or 639 of SEQ ID NO: 16205. In certain embodiments, the TP53 somatic mutation is any one or more mutation selected from the group consisting of: Leu194Arg, GlylOSAsp, Arg273His, Tyrl 63His, Ile232Thr, Arg213Ter, Arg273His,
Arg248Gln, Argl 75His, Argl 1 OdeiinsGlnSer, Ser 183Ter, Arg248G!n, Arg337Leu, Lysl 32Arg, Leu265ThrfsTer7, Arg306Ter, Cysl24TrpfsTer25s Pro72Args Val218Glu, His2!4Leu, Ginl44Ter, Phe1 13Ser, Tyr234His, Seii27Phe, Pro278Ala, Ala347Thr, and Prol52Leu of SBQ ID NO: 16205. In certain embodiments, the TP53 mutation is any one or more mutation at any one or more nucleotide position corresponding to nucleotide position 108, 215, 314, 338, 380, 395, 430, 455, 487, 524, 548, 581 , 637, 639, 641 , 653, 695, 700, 743, 818, 832, 916, 1010, or 1039 of SEQ ID NO: 16206.
The sequence of SEQ ID NO: 16205 (corresponding to GenBank Accession No.
N?_00Q537.3) is as follows:
MEEPQSDPSVEPPLSQETFSDLWKLLPENNVLSPLPSQAMDDLMLSPDDIEQW FTEDPGPDEAPPJviPEAAPPVAPAPAAPTPAAPAPAPSWPLSSSVPSQKTYQGSYGFRL GFLHSGTAKSVTCTYSPALNKMFCQLAKTCPVQLWVDSTPPPGTRVRAMAIYKQSQ HMTEVVRR.CPHHERCSDSDGLAPPQHLiRVEGNI.RVEY i.J3DRNTFRHSVVVPYEPPE VGSDCTTIHYNYMCNSSCMGGMNRRPILTIITLEDSSGNLLGRNSFEVRVCACPGRDR RTEEEENLRKKGEPHHELPPGSTKRALPNNTSSSPQPKKKPLDGEYFTLQIRGRERFEM FRELNEALELKDAQAGKEPGGSRAHSSHLKSKKGQSTSRHKKLMFKTEGPDSD
The sequence of SEQ ID NO: 16206 (corresponding to GenBank Accession No.
NM 000546.5) is as follows:
Figure imgf000039_0001
i
Figure imgf000040_0001
Figure imgf000041_0001
Esophageal adenocarcinoma (BAG) has steadily increased in incidence over recent decades. With an 85% mortality rate this cancer is the most, rapidly increasing cause of cancer mortality from solid tumors in the American population. There has thus been substantial interest in development of screening approaches for early detection of EAC and its precursor lesions of Barrett's esophagus (BE), However, the majority of BACs develop in patients without prior symptoms, and current approaches of endoscopic screening of individuals with persistent symptoms of gastroesophageal reflux disease, combined with longitudinal screening of those found to ha ve BE, have accordingly not had significant impact on reducing deaths from EACs.
As noted above, early detection of gastrointestinal neoplasia (e.g., neoplasia of the upper gastrointestinal tract) coupled with appropriate intervention, is important for increasing patient survival rates. Present systems for screening for esophageal neoplasia are deficient for a variety of reasons, including a lack of specificity and/or sensitivity (e.g., barium swallow) or a high cost and intensive use of medical resources (e.g., upper endoscopy or CT scan).
Alternative systems for detection of esophageal neoplasia would be useful in a wide range of other clinical circumstances as well. For example, detecting esophageal neoplasia may select the patient to undergo therapies that include, but are not limited, to resection of the neoplasia (via endoscopic resection or surgical resection), ablation of the neoplasia, chemotherapy, or radiation therapy. As a further example, patients who have received surgical and/or pharmaceutical therapy for esophageal cancer may experience a relapse. It would be advantageous to have an alternative system for determining whether such patients have a recurrent or relapsed neoplasia of the upper gastrointestinal tract. As a further example, an alternative diagnostic system would facilitate monitoring an increase, decrease or persistence of neoplasia of the upper gastrointestinal tract in a patient known to have such a neoplasia. A patient undergoing chemotherapy may be monitored to assess the effectiveness of the therapy. III, Methvlation of fefomiative Loci as .Disease Bioroarkers.
The present disclosure relates at least in part to the identification of genomic loci whose altered DNA methylation is indicative of the presence of esophageal neoplasias and/or metaplasias that include Barrett's esophagus (BE) and/or esophageal adenocarcinoma (EAC), In some embodiments, the Barrett's esophagus is associated with dysplasia. In some embodiments, the dysplasia is high-grade dysplasia, in some embodiments, the dysplasia is low-grade dysplasia, In some embodiments, the methylation patterns of the informative loci as disclosed herein are determined in a sample taken from a subject as described herein and may be used to distinguish between subjects having Barrett's esophagus and subjects having high grade dysplasia and/or low grade dysplasia and/or esophageal adenocarcinoma.
Examples of the informative loci are provided herein.
In some embodiments, any of the nucleotide sequences disclosed herein, or fragments or reverse complements thereof, may contain one or more "Y" residues, Cytosine residues that may be methylated or unmethyiaied, and hence may be bisulfite converted to T (if unmethyiaied) or remain as a C (if methylated), are designated with a 'Ύ." In some embodiments, one or more of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) designates a methylated C. In some embodiments, one or more of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) designates an unmethyiaied C. In some embodiments, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, I I , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) correspond to methylated C residues. In some embodiments, at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues in any of t he sequences disclosed herein (or fragments or reverse complements thereof) correspond to unmethyiaied C residues. In some embodiments, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) correspond to methylated C residues. In some embodiments, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues in any of the sequences disclosed herein (or fragments or reverse complements thereof) are correspond to unmethyiaied C residues, in some embodiments, any of the sequences disclosed herein (or fragments or reverse complements thereof) is bisulfite-converied. In some embodiments, at least 1 , 2, 3, 4. 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, .17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues in any of the bisulfite-converted sequences disclosed herein (or fragments or reverse complements thereof) correspond to C, In some embodiments, at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues in any of the bisulfite-converted sequences disclosed herein (or fragments or reverse complements thereof) correspond to T. In some embodiments, at least 10%, 20%, 30%, 40%. 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues in any of the bisulfite-converted sequences disclosed herein (or fragments or reverse complements thereof) correspond to C residues. In some embodiments, at least. 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues in any of the bisulfite-converted sequences disclosed herein (or fragments or reverse complements thereof) correspond to T residues.
In some embodiments, an informative loci in a subject is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g. , Barrett's esophagus) or neoplasia {e.g..
Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma) if the loci is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% methylated, in some embodiments, a DNA sample from a subject is treated with bisulfite, and the resulting bisulfite sequence
corresponds to any of the nucleotide sequences disclosed herein comprising a "Y" nucleotide. In some embodiments, if at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 1 8, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues of the bisulfite-converted sequence have a C, the sequence is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g. , esophageal cancer such as esophageal adenocarcinoma). In some embodiments, a DNA sample from a subject is treated with bisulfite, and the resulting bisulfite sequence corresponds to any of the nucleotide sequences disclosed herein comprising a 'Ύ" nucleotide, in some embodiments, if at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues of the bisulfite-converted sequence have a C, the sequence is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g. , Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma). The disclosure provides for informative loci that may be used to assess whether a subject (e.g. a human) has or is prone to developing a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal
adenocarcinoma), in some embodiments, one or more informative loci, as defined herein, may be used for determining whether a subject has or is likely to develop, a metaplasia (e.g., Barrett's esophagus), In some embodiments, one or more informative loci, as defined herein, may be used for determining whether a subject has or is likely to develop, a neoplasia (e.g., Barrett's esophagus with high grade dysplasia, or an esophageal cancer such as esophageal adenocarcinoma). In some embodiments, one or more informative loci, as defined herein, may be used to distinguish between whether a subject has a metaplasia in the esophagus (e.g., Barrett's esophagus) or an esophageal neoplasia (e.g., Barrett's esophagus with high grade dysplasia, or an esophageal cancer such as esophageal adenocarcinoma).
In some embodiments, the informative loci include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1-428, 2569-2996, 5137-5531 , 7507-7532, 7663-7668, 7819-7842, 7963-7976, 8047-8060, 8131- 8143, 8209-8222, 8293-8306, 8405-8409, 8447-8632, 9563-9748, 10679-10825, 11561- 1161 1 , 1 1867-1 1917, 12173-12219, 12455-12460, 12491-12496, 12527-12532, 12563-12568, 12599-12604, 12647-12649, 12671 -12907, 14093-14329, 15515-15537, 15653-15692, 15893-15932, 16133-16137, 16163-16165, 16181 -16183, or 16199, or fragments or complements thereof, in particular embodiments, the informative loci include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 7963-7976, 8047-8060, 8131-8143, 12455- 12460, 12491-12496, 12527-12532, 16163-16165, 16181-16183. or 16199, or fragments or complements thereof. In some embodiments, the informative loci are associated with increased methyl ation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject, in some embodiments, the informative loci that are associated with increased methylation in both Barrett's esophagus and esophageal
adenocarcinoma samples include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%», 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1 -428, 2569-2996, 5137- 5531, 7507-7532, 7663-7668, 7819-7842, 7963-7976, 8047-8060, 8131-8143, 8209-8222, 8293-8306, or 8405-8409, or fragments or complements thereof. In particular embodiments, the informative loci thai are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 7963-7976, 8047- 8060, or 8131-8143, or fragments or complements thereof, In some embodiments, the informative loci are associated with increased methylation in an esophageal adenocarcinoma sample and/or a Barrett's with low grade or high grade dysplasia as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia. In some embodiments, the informative loci that, are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 8447- 8632, 9563-9748, 10679-10825, 1 1561 -1 161 1 , 1 1867-1 1917, 12173-12219, 12455-12460, 12491-12496, or 12527-12532, or fragments or complements thereof, in particular embodiments, the informative loci that are associated with increased methylation in Barrett's with low grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 12455-12460, 12491-12496, or 12527-12532, or fragments or complements thereof. In particular embodiments, the informative loci that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 12455-12460, 12491-12496, or 12527-12532, or fragments or complements thereof, in some embodiments, the informative loci are associated with reduced methylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus. In some embodiments, the informative loci thai are associated with reduced methylation in an esophageal adenocarcinoma sample include any¬ one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 12671-12908, 14093- 14329, 15515-15537, 15653-15692, 15893-15932, 16133-16137, 16163-16165, 16181 -16183, or 16199, or fragments or complements thereof. In particular embodiments, the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 16163- 16165, 16181-16183, or 16199, or fragments or complements thereof.
In some embodiments, the informative loci or amplicon of the informative loci are treated with an agent, such as bisulfite. In some embodiments, the informative loci include sequences that have been treated with bisulfite. In some embodiments, the disclosure provides for bisulfite control sequences of any of the plus DNA strands disclosed herein. In some embodiments, the disclosure provides for bisulfite-treated unme thy Sated sequences of any of the plus DNA strands disclosed herein. In some embodiments, the bisulfite-converted plus-strand control DNA sequences include any one or more having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 857-1284, 3425-3852, 5927-6321 , 7559-7584, 7715-7740, 7867-7890, 7991-8004, 8075-8088, 8157-8169, 8223-8236, 8307-8320, 8410-8414, 8819-9004, 9935-10120, 10973- 1 1 1 19, 1 1663-1 1713, 1 1969-12019, 12267-12313, 12467-12472, 12503-12508, or 12539- 12544, 12569-12574, 12605-12610, 12650-12652, or fragments or complements thereof. In particular embodiments, the bisulfite-converted plus-strand control DNA sequences include any one or more having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 7991 -8004, 8075-8088, 8157-8169, 8223-8236, 8307-8320, 8410-8414, 12467-12472, 12503-12508, or 12539-12544, 12569- 12574, 12605-12610. 12650-12652, or fragments or complements thereof, in some embodiments, the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject, in some embodiments, the disclosure provides for bisuifiie-treated mtmefhylated sequences of any of the plus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject. In some embodiments, the bisulfite converted sequences of any of the plus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples are selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 857-1284, 3425-3852, 5927-6321, 7559-7584, 7715-7740, 7867- 7890, 7991-8004, 8075-8088, 8157-8169, 8223-8236, 8307-8320, or 8410-8414, or fragments or complements thereof. In particular embodiments, the bisulfite converted sequences of any of the plus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisulfite- converted methylated plus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of: SEQ ID NOs: 7991-8004, 8075-8088, 8157-8169, 8223-8236, 8307-8320, or 8410-8414, or fragments or complements thereof, in some embodiments, the disclosure provides for bisulfite-treatecl unmethylated sequences of any of the plus DNA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia. In some embodiments, the bisulfite converted sequences of any of the plus DNA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more bisulfite-converted methylated plus-strand DNA sequences selected from the group consisting having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 8819-9004, 9935-10120, 10973-1 1 1 19, 1 1663-1 1713, 11969-12019, 12267-12313, 12467-12472, 12503-12508,12539-12544, 12569-12574, 12605- 12610, or 12650-12652, or fragments or complements thereof, in particular embodiments, the bisulfite converted sequences of any of the plus DMA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or hi gh grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any¬ of SEQ ID NOs: 12467-12472, 12503-12508, 12539-12544, 12569-12574, 12605-12610, or 12650-12652, or fragments or complements thereof. In some embodiments, the informative loci are associated with reduced meihylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus. In some embodiments, the disclosure provides for methylated control sequences of the plus DNA strand that are associated with reduced meihylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus. In some embodiments, the methylated control sequences of any of the plus DNA strands that are associated with reduced meihylation in an esophageal adenocarcinoma sample include any one or more bisulfite-con verted methylated plus-strand DN A sequences selected from the group consisting having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 13145-13381, 14567-14803, 15561-15583, 15733-15772, 15973-16012, 16143-16147, 16169-16171, 16187-16189 or 16201 , or fragments or complements thereof. In particular embodiments, the methylated control sequences of any of the plus DNA strands that are associated with reduced
meihylation in an esophageal adenocarcinoma sample include any one or more bisulfite- converted methylated plus-strand DNA sequences selected from the group consisting having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 16169-16171 , 16187-16189 or 16201 , or fragments or complements thereof,
In some embodiments, the informative loci or amplicon of the informative loci are treated with an agent, such as bisulfite, In some embodiments, the informative loci include sequences that have been treated with bisulfite, In some embodiments, the informative loci include methylated nucleic acid sequences that have been treated with bisulfite, in some embodiments, the bisulfite-converted methylated plus-strand DNA sequences have at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1285-1712, 3853-4280, 6322-6716, 7585-7610, 7741-7766, 7891- 7914, 8005-8018, 8089-8102, 8170-8182, 8237-8250, 8321-8334, 8415-841 9, 9005-9190, 10121-10306, 1 1 120-1 1266, 11714-1 1764, 12020-12070, 12314-12360, 12473-12478, 12509-12514 or 12545-12550, 12575-12580, 12611-12616, 12653-12655, or fragments or complements thereof, in particular embodiments, the bisulfite-converted methylated plus- strand DNA sequences have at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 8005-8018, 8089-8102, 8170-8182, 12473-12478, 12509-12514 or 12545-12550, or fragments or complements thereof. In some embodiments, the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject. In some embodiments, the informative loci that are associated with increased methy!ation in both Barrett's esophagus and esophageal
adenocai'cinoma samples include any one or more bisulfite-converted methylated plus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1285-1712, 3853-4280, 6322-6716, 7585-7610, 7741-7766, 7891 -7914, 8005- 801 8, 8089-8102, 8170-8182, 8237-8250, 8321 -8334, or 8415-8419, or fragments or complements thereof. In particular embodiments, the informative loci that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisulfite-converted methylated plus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to: SEQ ID NOs: 8005-8018, 8089- 8102, 8170-81 82, 8237-8250, 8321-8334, or 8415-8419, or fragments or complements thereof. In some embodiments, the informative loci are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia, in some embodiments, the informative loci that are associated with increased methvlation in an esophageal adenoearcmoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more bisulfite-converted methylated plus-strand DMA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%., 98%, 99% or 100% identity to SEQ ID NOs: 9005-9190, 10121-10306, 11 120-11266, 1 1714-1 1764, 12020-12070, 12314-12360, 12473-12478, 12509-12514, 12545-12550, 12575-12580, 12611-12616, or 12653-12655, or fragments or complements thereof, in particular embodiments, the informative loci that are associated with increased methvlation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 12473- 12478, 12509-12514, 12545-12550, 12575-12580, 12611-12616, or 12653-12655, or fragments or complements thereof, In some embodiments, the informative loci are associated with reduced methylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having BaiTett's esophagus, In some embodiments, the informative loci that are associated with reduced methylation in an esophageal
adenocarcinoma sample include any one or more bisulfite-converted methylated plus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 13382-13618, 14804-15040, 15584-15606, 15773-15812, 16013-16052, 16148-16152, 16172-16174, 16190-16192 or 16202. in particular embodiments, the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 16172-16174, 16190-16192 or 16202, or fragments or complements thereof.
In some embodiments, the informative loci include sequences associated with any of the minus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 429-856, 2997- 3424, 5532-5926, 7533-7558, 7689-7714, 7843-7866, 7977-7990, 8061 -8074, 8144-8156, 8251-8264, 8335-8348, 8420-8424, 8633-8818, 9749-9934, 10826-10972, 1 1612-1 1662, 11918-1 1968, 12220-12266, 12461-12466, 12497-12502, 12533-12538, 12581-12586, 12617-12622, 12656-12658, 12909-13144, 14330-14566, 15538-15560, 15693-15732, 15933-15972, 16138-16142, 16166-16168, 16184-16186 or 16200, or fragments or complements thereof. In particular embodiments, the informative loci include sequences associated with any of the minus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%», 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 7977-7990, 8061-8074, 8144-8156, 8251 -8264, 8335-8348, 8420-8424, 12461-12466, 12497-12502, 12533-12538, 12581-12586, 12617-12622, 12656-12658, 16166-16168, 16184-16186 or 16200, or fragments or complements thereof. In some embodiments, the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject. In some embodiments, the informative loci that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include sequences associated with any one or more of the minus strand DN A sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 429-856, 2997-3424, 5532-5926, 7533-7558, 7689- 7714, 7843-7866, 7977-7990, 8061-8074, 8144-8156, 8251-8264, 8335-8348, 8420-8424, or fragments or complements thereof. In particular embodiments, the informative loci that are associated with increased methylation in both Barrett' s esophagus and esophageal adenocarcinoma samples include sequences associated with any one or more of the plus strand DNA sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%., 96(½., 97%, 98%), 99% or 100% identity to any of SEQ ID NOs: 8251-8264, 8335-8348, 8420- 8424, or fragments or complements thereof. In some embodiments, the informative loci are associated with, increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia, In some embodiments, the informative ioci that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%., 97%, 98%», 99% or 100% identity to any of SEQ I D NOs: 8633-8818, 9749- 9934, 10826-10972, 1 1612-1 1662, 1 1918-1 1968, 12220-12266, 12461-12466, 12497-12502, 12533-12538, 12581-12586, 1261 7-12622, or 12656-12658, or fragments or complements thereof, in particular embodiments, the informative loci that are associated with increased methylaiion in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NQs: 12461-12466, 12497-12502, 12533-12538, 12581-12586, 12617-12622, or 12656-12658, or fragments or complements thereof. In some embodiments, the informative loci are associated with reduced methylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus. In some embodiments, the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 12909-13144, 14330-14566, 15538-15560, 15693-15732, 15933- 15972, 16138-16142, 16166-16168, 16184-16186 or 16200, or fragments or complements thereof, in particular embodiments, the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%. or 100% identity to any of SEQ ID NOs: 16166-16168, 16184-16186 or 16200, or fragments or complements thereof.
In some embodiments, the informative loci or amplicon of the informative Soei are treated with an agent, such as bisulfite. In some embodiments, the informative loci include sequences that have been treated with bisulfite. In some embodiments, the disclosure provides for bisulfite control sequences of any of the minus DNA strands disclosed herein, in some embodiments, the disclosure provides for hisulfite-treated sequences of any of the minus DNA strands disclosed herein, in some embodiments, the bisulfite-eonverted minus-strand control DNA sequences include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs; 1713-2140, 4281 -4708, 6717-71 1 1 , 761 1 -7636, 7767-7792, 7915-7938, 8019- 8032, 8103-81 16, 8183-8195, 8265-8278, 8349-8362, 8425-8429, 9191 -9376, 10307-10492, 1 1267-11413, 1 176541815, 12071-12121 , 12361-12407, 12479-12484, 12515-12520, 12551 -12556, 12587-12592, 12623-12628, or 12659-12661 , or fragments or complements thereof, in particular embodiments, the bisulfite-eonverted minus-strand control DNA sequences include any one or more the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%», 99% or 100% identity to any of SEQ I D NOs: 8019- 8032, 8103-81 16, 8183-8195, 12479-12484, 12515-12520, or 12551 -12556, or fragments or complements thereof. In some embodiments, the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject, in some embodiments, the disclosure provides for bisulfite-treated sequences of any of the minus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject. In some embodiments, the sequences of any of the minus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples are selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of SEQ ID NOs: 1713-2140, 4281-4708, 6717-71 1 1 , 761 1-7636, 7767- 7792, 7915-7938, 8019-8032, 8103-81 16, 8183-8195, 8265-8278, 8349-8362, or 8425-8429, or fragments or complements thereof, in particular embodiments, the sequences of any of the minus DNA strands that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisulfite- converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%*, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to: SEQ ID NOs: 8019-8032, 8103-81 16, 8183-8195, 8265-8278, 8349-8362, or 8425-8429, or fragments or complements thereof, in some embodiments, the disclosure provides for bisulfite-treated sequences of any of the minus DNA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia, in some embodiments, the sequences of any of the minus DNA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more bisulfite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 9191-9376, 10307-10492, 1 1267-1 1413, 1 1765-1 1815, 12071 -12121 , 12361-12407, 12479- 12484, 12515-12520, or 12551 -12556, 12587-12592, 12623-12628, or 12659-12661 , or fragments or complements thereof. In particular embodiments, the unmethylated sequences of any of the minus DNA strands that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%s, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 12479- 12484, 12515-12520, 12551-12556, 12587-12592, 12623-12628, or 12659-12661, or fragments or complements thereof, in some embodiments, the informative loci are associated with reduced methylalion in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus. In some embodiments, the disclosure provides for methylated control sequences of the minus DNA strand that are associated with reduced methylalion in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus, in some embodiments, the methylated control sequences of any of the minus DNA strands that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more bisulfite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%s, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 13619-13855, 15041 -15277, 15607-15629, 15813-15852, 16053-16092, 16153-161 57, 16175-1 6177, 16192-16195 or 16203, or fragments or complements thereof in particular embodiments, the methylated control sequences of any of the minus DNA strands that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more bisulfite- converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%), 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 16175-16177, 16192-16195 or 1 6203, or fragments or complements thereof.
In some embodiments, the informative loci or amplicon of the informative loci are treated with an agent, such as bisulfite. In some embodiments, the informative loci include sequences that have been treated with bisulfite. In some embodiments, the informative loci include methylated nucleic acid sequences that have been treated with bisulfite. In some embodiments, the bisuifite-converted methylated minus-strand DNA sequences include any one or more of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%. or 100% identity to SEQ ID NOs: 2141-2568, 4709-5136, 7112-7506, 7637-7662, 7793-7818, 7939-7962, 8033-8046, 81 17-8130, 8196-8208, 8279-8292, 8363- 8376, 8430-8434, 9377-9562, 10493-10678, 1 1414-1 1560, 1 1816-1 1866, 12122-12172, 12408-12454, 12485-12490, 12521 -12526, 12557-12562, 12593-12598, 12269-12634, or 12662-12664, or fragments or complements thereof. In particular embodiments, the bisuifite- converted methylated minus-strand DNA sequences include any one or more of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%>, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 8033-8046, 81 17-81 30, 8196-8208, 8279-8292, 8363-8376, 8430-8434, 12485-12490, 12521-12526, 12557-12562, 12593-12598, 12269-12634, or 12662-12664, or fragments or complements thereof. In some embodiments, the informative loci are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples, as compared to the same sample types taken from a healthy control subject. In some embodiments, the informative loci that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisuifite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 2141 -2568, 4709-5136, 71 Ϊ 2- 7506, 7637-7662, 7793-7818, 7939-7962, 8033-8046, 81 17-8130, 8196-8208, 8279-8292, 8363-8376, or 8430-8434, or fragments or complements thereof. In particular embodiments, the i.nfonnaiive loci that are associated with increased methylation in both Barrett's esophagus and esophageal adenocarcinoma samples include any one or more bisuifite-converted methylated minus-strand DN A sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to: SEQ ID NOs: 8033-8046, 81 17-8130, 8196-8208, 8279-8292, 8363-8376, or 8430~8434,or fragments or complements thereof. In some embodiments, the informative loci are associated with increased meihylation in an esophageal adenocarcinoma sample or a Barrett's with high grade dysplasia sample as compared to a sample of the same type taken from a subject having Barrett's esophagus without dysplasia. In some embodiments, the informative loci that are associated with increased methylation in an esophageal
adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more bisuifite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%*, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 9377-9562, 10493- 10678, 11414-1 1560, 11816-1 1866, 12122-12172, 12408-12454, 12485-12490, 12521 -12526, 12557-12562, 12593-12598, 12269-12634, or 12662-12664, or fragments or complements thereof. In particular embodiments, the informative loci that are associated with increased methylation in an esophageal adenocarcinoma sample or a Barrett's with low grade or high grade dysplasia sample include any one or more of the sequences of sequences having at least 80%, 85%, 87%, 90%, 91 %, 92%», 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 12485-12490, 12521 -12526, 12557-12562, 12593-12598, 12269-12634, or 12662-12664, or fragments or complements thereof, in some embodiments, the informative loci are associated with reduced methylation in an esophageal adenocarcinoma sample as compared to a sample of the same type taken from a subject having Barrett's esophagus. In some embodiments, the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more bisuifite-converted methylated minus-strand DNA sequences selected from the group consisting of sequences having at least 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 13856-14092, 15278-15514, 15630-15652, 15853-15892, 16093-16132, 16158-16162, 16178-16180. .16196-16198, or 16204, or fragments or complements thereof. In particular embodiments, the informative loci that are associated with reduced methylation in an esophageal adenocarcinoma sample include any one or more of the sequences having at least 80%, 85%, 87%, 90%, 91%, 92%. 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NOs: 16178-16180, 161 96-16198, or 16204, or fragments or complements thereof.
In some embodiments, the disclosure provides for a bisuifite-converted nucleotide sequence comprising the bisulfite-conveited nucleotide sequence of any one of the following: Up3, Up10, Up15-1, Up1 5-2, Up20-L Up20-2, Up20-2, Up27, Up35~L Up35-2, SqBE2, SqBE5, SqBE7, SqBE9, SqBE1O, SqBE1 l-I , SqBE1 I-2, SqBE13, SqBE14-2, SqBE1S, SqBE16-l, SqBE16-2, SqBE17-l, SqBE18, SqBE22-l , SqBE22-2 or SqBE23. In particular embodiments, the sequence comprises the bisuSiile-converted nucleotide sequence of any one of the following: Up3, Up 10, Up15-1. Up15-2, Up20-1 , Up2G-2, Up20-2, Up27, Up35-1, or Up35~2. In particular embodiments, the sequence comprises the hisuliite-converted nucleotide sequence of any one of the following: SqBE2, SqBE5, SqBE7, SqBE9, SqBE1O, SqBE1 l-1, SqBE1 l~2, SqBE13, SqBE14-2, SqBE1 S, SqBE16-l, SqBE16-2, SqBE17~l , SqBE1 S, SqBE22-l , SqBE22-2 or SqBE23.
In some embodiments, the disclosure provides for a panel of any of the sequences disclosed herein, in some embodiments, the panel comprises any of the following
combinations of sequences: a) Up3, Up10, Up1 5~L Up15-2, Up20-1 , Up20-2, Up27, Up35-1, and Up35-2; b) Up3, Up1 5-1 , Up15-2, Up20-1 , Up27, and Up35-1 ; c) Up10, Up3, Up15-1 , Up15~2, Up20-L Up27, and Up35-1 ; d) Up35-2, Up3, Up15~L Up15-2, Up20-1 , Up27 and Up35-1 ; e) Up15-1 and Up354 ; i) Up15-1, Up35-1, and Up10; g) Up15-1 , Up354 and Up20-1 ; h) Up15-1 , Up35-1 , Up10, and Up ! 5 -2; i) Up15-1, Up35-1, Up10, and Up27; j) Up15-1 , Up35-1 , Up15-2, and Up20-1 ; k) Up1 5-1} Up35~L Up15-2 and Up27; l)Up15-l, Up35-1 , Up20-1 , and Up27: m) Up3 and Up35-1 ; n) Up3 and Cp35-2; o)Up3 and Up10; p) Up3 and Up27; q) Up35-1 and Up35-2; r) Up35-1 and Up27; s)Up35-2 and Up10; t) Up10 and Up27; u) Up3, Up35-1 and Up35~2; v) Up3, Up35-1 and Up10; w) Up3, Up35-1, and Up27; x) Up3, Up35-2 and Up10; y) Up3, Up35-2, and Up27; z) Up3, Up10, and Up27; aa)Up35-l , Up10, and Up27; ab) Up35-2, Up10, and Up27; ac) Up3, Up35-1 , Up35-2 and Up1 O; ad) Up3, Up35-1, Up35-2 and Up27; ae) Up35-1, Up35-2, Up10 and Up27; af) Up3, Up35-2, Up10 and Up27; ag) Up3, Up35-1, Up10 and Up27: ah) Up3, Up10, Up27, Up35-1, and Up35-2; ai) Up35-1 and Up1O, aj) Up35-1 and Up27; ak) Up35-2 and Up10; al) Up35-2 and Up27; am) Up3, Up35-1 and Up35-2; an) Up3, Up35-1, and Up1O; ao) Up3, Up35-1, and Up27; ap) Up3, Up35-2 and Up 10; aq) Up3, Up35-2 and Up27; ar) Up3, Up1O and Up27; at) Up35-1, Up10, and Up27; an) Up3, Up35-1, Up35-2, and Up10; av) Up3, Up35-1, Up35-2 and Up27; aw) IJp35-l , Up35-2, Up10 and Up27; ax) Up3, Up35-2, Up1 O and Up27; ay) Up3, Up35-1 , Up1 O and Up27; az) Up3, Up1 O, Up27, Up35-1 , and Up35-2; ba) SqBE5 and SqBE7; bb) SqBE5 and SqBE16; be) SqBE5 and SqBE17; bd) SqBE5 and SqBE18; be)
SqBE7 and SqBE16; bf) SqBE 7 and SqBE17; SqBE7 and SqBE17; bg) SqBE7 and SqBE18; bh) SqBE 16 and SqBE 17 and bi) SqBE16 and SqBE18. In some embodiments, the disclosure provides for a method of detecting the methylation status of the sequences in any of the panels disclosed herein. In some embodiments, the disclosure provides for a method of detecting the methylation status of the sequences in any of the panels disclosed herein, and further comprises detecting the mutation status of p53. In particular embodimenis, the disclosure provides for a method of a) detecting the methylation status of a panel comprising the sequences of Up-3 and Up35-2, and b) further detecting the mutation status of TP53,
In some embodiments, the disclosure provides for a method of detecting the methylation status of any of the loci disclosed herein, and further comprises detecting the methylation status of vimentin. In some embodiments, the vimentin methylation is detected in a manner consistent with that described in Li et al, (Li M, et al, (2009) Sensitive digital quantification of D'NA methylation in clinical samples. Nat Biotechnol 27(91:858-863). In some embodiments, the vimentin methylation patterns are determined in a nucleotide sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NO: 16207 or 16208. In some embodiments, the methylation patterns are determined in any of the following nucleic acid sequence combinations: a) vimentin and SQBE5; b) vimentin and SQBE7, c) vimentin and SQBE 16, d) vimentin and SQBE 17 or e) vimentin and SQBE18.
In particular embodiments, the disclosure provides for a nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:8209-8222, 8251-8264, 8293-8306, 8335-8348, 8405-8409, 8420-8424, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or fragments and/or reverse complements thereof. In particular embodiments, the disclosure provides for a nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: 12563-12568, 12581- 12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or fragments and/or reverse complements thereof. In particular- embodiments, the disclosure provides for a nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:8209- 8222, 8251-8264, 8293-8306, 8335-8348, 8405-8409, or 8420-8424, or fragments and/or reverse complements thereof.
in some embodiments, the disclosure provides for a bisuiilte-con verted nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences; SEQ ID NOs: 8307-8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8411, 8412, 8414, 8416, 8417, 8419, 8426, 8427, 8429, 8431 , 8432, 8434, 1260542616, 12623-12634, 12650- 12655, or 12659-12664, or fragments and/or reverse complements thereof. In some embodiments, the sequence comprises a sequence having at least 80%, 85%», 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 1G0% identity to any of the following sequences: SEQ ID NOs; 8307-8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8411 , 8412, 8414, 8416, 8417, 8419, 8426, 8427, 8429, 8431, or 8432, 8434. In some
embodiments, the sequence comprises a sequence having at. least 80%, 85%, 90%, 91 %. 92%, 93%, 94%, 95%, 96%», 97%, 98%, 99%. or 100% identity to any of the following sequences: SEQ ID NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664,
In some embodiments, the disclosure provides for a bisuifite-eonverted nucleotide sequence comprising a sequence having at least 80%, 85%, 90%., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8223-8250, 8265-8292, 12569-12580, or 12587-12598, or fragments and/or reverse complements thereof. In some embodiments, the sequence comprises a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences; SEQ ID NOs: 8223-8250 or 8265-8292. In some
embodiments, the sequence comprises a sequence having at least 80%. 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 12569-12580 or 12587-12598,
in some embodiments, the sample for use in any of the methods disclosed herein is a tissue sample taken from the subject. In some embodiments, the sample is tissue sample from the esophagus, in some embodiments, the sample is a biopsy or a brushing. In some embodiments, the sample is a biopsy or brushing of the esophagus. In some embodiments, the sample is a body fluid. In some embodiments, the body fluid is blood, serum, saliva, spit, stool, urine or an esophageal washing.
The present disclosure contemplates methods of selecting an individual to undergo a diagnostic procedure to determine the presence of Barrett's esophagus, Barrett's esophagus with dysplasia (e.g., Barrett's esophagus with low-grade or high-grade dysplasia), or of esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA meihylatiors in at least one of any of the sequences disclosed herein. In some embodiments, the disclosure provides for a method of selecting a subj ect for monitoring of esophageal neoplasia, wherein the presence of DNA methylation in at least one of any of the sequences disclosed herein is detected in a sample from the subject, in some embodiments, detection is achieved by any one or more of DNA sequencing, next generation sequencing, methylation specific PGR, methylation specific PGR combined with a fluorogenic hybridization probe, real time methylation specific PGR. or hybridization to an array. In some embodiments, the detection in the sample is indicative that the subject is at high risk of progression to esophageal neoplasia (e.g., esophageal cancer). In some embodiments, the subject is monitored by endoscopy. In some embodiments, a sample from a subject in which DNA methylation of at least one of any of the sequences disclosed herein is detected, is indicative that the subject should be administered a particular treatment. In some embodiments, the treatment is selected from the group consisting of endoscopic removal or ablation of an esophageal neoplasia, and/or surgery, radiation, or chemotherapy treatment of esophageal adenocarcinoma. In some embodiments, the sequence is any one or more sequence selected from the group consisting of a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 1- 856, 2569-3424, 5137-5926; 7507-7558, 7663-7714, 7819-7866, 7963-7990, 8047-8074, 8131-8156, 8209-8222, 8251-8264, 8293-8306, 8335- 8348, 8405-8409, and 8420-8424, or fragments or complements thereof.
The present disclosure also contemplates methods of selecting an individual to undergo a diagnostic procedure to determine presence of Barrett's esophagus with low-grade dysplasia, Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA meihylation in at least one of any of the sequences disclosed herein, in some embodiments, the sequence is any one or more sequence selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: : SEQ ID NOs: 8447 - 8818, 9563 - 9934, 10679 - 10972, 11561 - 11662, 1 1867 - 1 1968, 12173 - 12266, 12455 - 12466, 12491 - 12502, 12527 - 12538, 12563 - 12568, 12581 - 12586, 12599 - 12604, 12617 -- 12622, 12647 - 12649, or 12656 ~ 12658, or fragments or complements thereof The present disclosure, further contemplates that the mediod may further comprise determining the status of somatic mutation(s) in TP53 in the sample, For example, a method comprising
determining the status of methylation of chromosomal loci e.g., Up15-L Up35-1 , Up35-2, Up3, Up27, and Up 10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated.
The present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample. For example, a method comprising determining the status of methylation of chromosomal loci e.g. , Up15-1 , Up35-1 , Up35-2, Up3, Up27, and Up 10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated, in certain embodiments, the method optionally further comprises detecting the presence or absence of a somatic mutation in TP53. in certain embodiments, the TP53 somatic mutation, as described herein, is any one or more mutation at any one or more amino acid residue corresponding to amino acid residue 72, 105, 108, 1 10, 113, 124, 127, 132, 144, 152, 163, 175, 183, 194, 213, 214, 218, 232, 234, 248, 265, 273, 278, 306, 337, 347, or 639 of SEQ ID NO: 16205. In certain embodiments, the TP53 somatic mutation is any non-synonomous somatic mutation known in the art. In certain embodiments, the TP53 somatic mutation is any one or more mutation selected from the group consisting of: Leul94Arg, GlylOSAsp, Arg273His, Tyrl63His, i.!e232Thr, Arg213Ter, Arg273His, Arg248Gln, Argl75His, Argl 1 OdelinsGlnSer, Serl 83Ter, Arg24SGln,
Arg337Leu, Lys132Arg, Leu265ThrfsTer75 Arg306Ter, Cysl24TrpfsTer25, Pro72Arg, Val218Glu, His214Leu, Glnl44Ter, Phel DSer, Tyr234His, Serl 27Phe, Pro278Ala,
Ala347Thr, and Pro 152Leu of SEQ ID NO; 16205 in certain embodiments, the TP53 mutation is any one or more non-synonymous somatic mutation at any one or .more nucleotide position corresponding to nucleotide position 108, 215, 314, 338, 380, 395, 430, 455. 487, 524, 548, 581, 637, 639, 641 , 653, 695, 700, 743, 818, 832, 916, 1010, or 1039 of SEQ ID NO: 16206.
The present disclosure also contemplates methods of selecting an individual to undergo a treatment for Barrett's esophagus, Barrett's esophagus with low grade dysplasia, Barrett' s esophagus with high grade dysplasia or for esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA raethylation in at least one of any of the sequences disclosed herein.' In some embodiments, the sequence is any one or more sequence selected from the group consisting of sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926: 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, or fragments or complements thereof. In some embodiments, the present disclosure further contemplates that the method may further comprise determining the status of somatic rnutation(s) in TP53 in the sample. For example, a method comprising determining the status of methylation of chromosomal loci e.g., Upi 5-1 , Up35-1 , Up35~2, Up3, Up27, and Up1 O in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated.
The present disclosure also contemplates methods of selecting an individual to undergo a treatment for Barrett's esophagus, Barrett's esophagus with low-grade dysplasia, Barrett's esophagus with high grade dysplasia or for esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA methylation in at least one of any of the sequences disclosed herein. In some embodiments, the sequence is any one or more sequence selected from the group consisting of sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8447 - 8818, 9563 - 9934, 10679 - 10972, 11561 - 1 1662, 1 1867 - 1 1968, 12173 - 12266, 12455 - 12466, 12491 - 12502, 12527 - 12538, 12563 - 12568, 12581 - 12586, 12599 - 12604, 12617 - 12622, 12647 - 12649, or 12656 - 12658, or fragments or complements thereof. The present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP 53 in the sample. For example, a method comprising
determining the status of methylation of chromosomal loci e.g., Up15-1, Up35~l, Up35~2, Up3, Up27, and Up 10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated. The present disclosure also contemplates methods of selecting an individual to undergo enhanced surveillance for the development of Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining in the sample the presence of DNA methylation in at least one of any of the sequences disclosed herein, in some embodiments, the sequence is any one or more sequence selected from the group consisting of a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 8447 - 8818, 9563 -· 9934, 10679 - 10972, 1 1561 - 1 1662, 1 1867 - 1 1968, 12173 - 12266, 12455 - 12466, 12491 - 12502, 12527 - 12538, 12563 - 12568, 12581 - 12586, 12599 - 12604, 12617 - 12622, 12647 -· 12649, or 12656 - 12658, or fragments or complements thereof. The present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample. For example, a method comprising determining the status of methylation of chromosomal loci e.g., Up15-1, Up35-1, Up35-2, Up3, Up27, and Up10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated. The present disclosure also contemplates methods of determining the response of an individual with esophageal cancer to therapy by obtaining a biological sample from an individual with esophageal cancer, and determining the presence of methylation in at least one of any of the sequences disclosed herein. In some embodiments, the sequence is any one or more sequence selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%o identity to any of the following sequences:: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926: 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 81 56, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, 8447 - 8818, 9563 - 9934, 10679 ~ 10972; SEQ ID NOs: 1 1561 - 1 1662, 1 1867 - 1 1968, 12173 - 12266; SEQ ID NOs: 12455 - 12466, 12491 - 12502, 12527 - 12538, 12563 - 12568, 12581 - 12586, 12599™ 12604, 12617 - 12622, 12647 - 12649, and 12656 - 12658, or fragments or complements thereof. In some implementations, an increase in levels of methyiation over time is indicative of disease progression and a need for a change in therapy (such as modifying the dosing regime of an exiting therapy, or administering a new therapeutic(s) either alone or in combination with the existing therapy), and an absence of increase in levels of methyiation over time or decrease in levels of inethylation over time is indicative that a change in therapy is not required. The present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample. For example, a method comprising determining the status of methyiation of chromosomal loci e.g., Up1 5-1 , Up35-1 , Up35-2, Up3, Up27, and Up 10 in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated.
The present disclosure also contemplates method of distinguishing EAC and/or low/high grade dysplasia from BE by obtaining a biological sample from an individual, and determining in the sample the presence of DNA methyiation in at least one of any of the sequences disclosed herein. In some embodiments, the sequence is any one or more sequence selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96% , 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8447 - 8818, 9563 - 9934, 10679 - 10972; SEQ ID NOs: 1 1561 - 11662, 1 1867 - 1 1968, 12173 - 12266; SEQ ID NOs: 12455 - 12466, 12491 - 12502, 12527 - 12.538, 12563 - 12568, 12581 - 12586, 12599 - 12604, 12617 - 12622, .12647 - 12649, and 12656 - 12658, or fragments or complements thereof. The present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample. For example, a method comprising determining the status of methylation of chromosomal loci e.g., Up15-1 , Up35~l, Up35-2, Up3, Up27, and Up1 O in a sample; and determining the presence or absence of somatic mutation(s) in TP53 in the sample is contemplated, in certain embodiments, the absence of methylation at Up] 5-1 , Up35-1, Up35-2, Up3, Up27„ and Up10; and the absence of a somatic mutation in TP53 may be indicative of non-dysplastic Barrel's esophagus. In certain embodiments, the presence of methylation at any one of Up] 5-1 , Up35-1 , Up35-2. Up3. Up27, and Up10; or the presence of a somatic mutation in TPS 3 may be indicative of esophageal adenocarcinoma or of Barrett's with high grade dysplasia,
The present disclosure also contemplates method of distinguishing EAC and/or low/high grade dysplasia from BE by obtaining a biological sample from an individual, and determining in the sample the presence of DNA methylation in at least one of any of the sequences disclosed herein, in some embodiments, the sequence is any one or more sequence selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%>, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8447 - 8818, 9563 - 9934, 10679 - 10972; SEQ ID NOs: 11561 - 11662, 1 1867 - 1 1968, 12173 - 12266; SEQ ID NOs: 12455 - 12466, 12491 - 12502, 12527 - 12538, 12563 - 12568, 12581 - 12586, 12599 - 12604, 12617 - 12622, 12647 - 12649, and 12656 - 12658, or fragments or complements thereof. The present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP53 in the sample. For example, a method comprising determining the status of meihylation of chromosomal loci e.g., Up1 5-1 , Up35-1, Up35-2, Up3, Up27, and Up10 in a sample; and determining the presence or absence of somatic mutation(s) in TPS 3 in the sample is contemplated. In certain embodiments, the absence of methylation at Up1 S-1 , Up35-1, Up35-2, Up3, Up27, and Up] 0; and the absence of a somatic mutation in TP53 may be indicative of non-dysplastic Barret's esophagus, in certain embodiments, the presence of methylation at any one of Up15-1, Up35-1, Up35-2, Up3, Up27, and Up1O; or the presence of a somatic mutation in TPS 3 may be indicative of esophageal adenocarcinoma.
The present disclosure also contemplates a method of monitoring the progression (or regression) of esophageal neoplasias over time, The method involves detecting the methylation status of one or more nucleotide sequences selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, and 8420 - 8424, 8447 - 8818, 9563 - 9934, 10679 - 10972; SEQ ID NOs: 1 1561 - 1 1662, 1 1867 - 1 1968 12173 - 12266; SEQ ID NOs: 12455 - 12466, 12491 -- 12502, 12527 - 12538, 12563 - 12568, 12581 - 12586, 12599 - 12604, 12617 - 12622, 12647 - 12649, and 12656 - 12658, or fragments or complements thereof in samples from a subject at a first time and at a later time. In certain embodiments, neoplasia regression may be indicated by the absence of methylation in the nucleotide sequence taken at a later time and the presence of methyl ati on in the nucleotide sequence taken at the first time, In certain embodiments, neoplasia progression may be indicated by the presence of methylation in the nucleotide sequence taken at a later time and the absence of methylation in the nucleotide sequence taken at the first time. The present disclosure further contemplates that the method may further comprise determining the status of somatic mutation(s) in TP53 in the samples. In some embodiments, neoplastic regression may be indicated by the presence of methylated chromosomal loci e.g..
methylation of Up15-1 , Up35-1 , Up35-2, Up3, Up27, and/or Up10 or the presence of a somatic mutation in TP53 in a first sample; and the absence of methylated chromosomal loci e.g., unmethylated Up15-1 , Up35-1, Up35-2, Up3, Up27, and Up10, and the absence of somatic mutation(s) in TPS 3 in a later sample. In some embodiments, neoplastic progression may be indicated by the presence of unmethylated chromosomal loci e.g., unmethylated Up15-1 , Up35-1 , Up35-2, Up3, Up27, and Up1 O; and the absence of somatic mutation(s) in TPS 3 in a first sample; and the presence of methylated chromosomal loci e.g., methylated Up15-i, Up35-1, Up35-2, Up3, Up27, and/or Up1 O, or the presence of a somatic mutation in TPS 3 in a later sample.
The present disclosure also provides sequences that will hybridize under highly stringent conditions to the nucleotide sequences of any one or more of SEQ ID NOs: 1 -8444 and 8447-16204, or fragments or complements thereof. As discussed above, one of ordinary skill in the art will understand readily that appropriate stringency conditions which promote DNA hybridization can be varied. One of ordinary skill in the art will understand readily that appropriate stringency conditions which promote DNA hybridization can be varied. For example, one could perform the hybridization at 6.0 x sodium chloride/sodium citrate (SSC) at about 45 °C, followed by a wash of 2.0 x SSC at SO °C. For example, the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50 °C to a high stringency of about. 0,2 x SSC at 50 °C. In addition, the temperature in die wash step can be increased from low stringency conditions at room temperature, about 22 °C, to high stringency conditions at about 65 °C, Both temperature and salt may be varied, or
temperature or salt concentration may be held constant while the other variable is changed. In one embodiment, the disclosure provides nucleic acids which hybridize under low stringency conditions of 6 x SSC at room temperature followed by a wash at 2 x SSC at room
temperature.
In other embodiments, the disclosure also provides the methylated forms of the nucleotide sequences of any one or more of SEQ ID NOs: 1 -8444 and 8447-16204, or fragments thereof, wherein the cytosine bases of the CpG islands present in the sequences are methylated. In other words, the nucleotide sequences listed of any one or more of SEQ ID NOs: 1-8444 or 8447-16204 or fragments or complements thereof may be either in the methylated status (e.g.. as seen in neoplasias) or in the unmethylated status (e.g., as seen in normal cells). In further embodiments, the nucleotide sequences of the disclosure can be isolated, recombinant, and/or fused with a heterologous nucleotide sequence, or in a DNA library.
In certain embodiments, the present disclosure provides bisulfite-converted nucleotide sequences, for example, bisulfite-converted sequences selected from any of the sequences disclosed herein. In some embodiments, the sequence is selected from the group consisting of sequences having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 857 -2568, 3425-
5136, 5927-7506, 7559 - 7662, 7715 - 7818, 7867 - 7962, 7991 - 8046, 8075 - 8130, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434, and/or fragments thereof, and/or the reverse complements thereof, in yet other embodiments, the disclosure provides bisulfite-converted sequences selected from the group consisting of sequences having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 1 1560; 11663 - 1 1866, 1 1969 - 12172, 12267 - 12454; 12467 - 12490, 12503 - 12526, 12539 - 12562, 12569 - 12580, 12587 - 12598, 12605 - 12616, 12623 - 12634, 12650 - 12655, and 12659 - 12664, and/or fragments thereof, and/or the reverse complements thereof. In yet other embodiments, the disclosure provides bisulfiie-con verted sequences selected from the group consisting of sequences having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences:: SEQ ID NOs: 13145 - 14092, 14567 -15514, 15561 - 15652; 15733 - 15892, 15973 -- 16132, 16143 - 16162; 16169 - 16180, 16187 - 16198, and 16201 - 16204, and/or fragments thereof, and/or the reverse complements thereof.
A fragment of any of the nucleotide sequences disclosed herein may be of any length, so long as the meihylation status of that nucleotide sequence may be determined. In some embodiments, the nucleotide sequence is at least 10, 15, 25, 30, 40, 50, 60, 70, 80, 90, 100, 1 10, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 1200, 1400, 1500, 1700, or 2000 nucleotides in length. In some embodiments, the nucleotide sequence is at least 10-2000, 10-1000, 10-500, 10-200, 10-150, 10-100, 50-2000, 50-1000, 50-500, 50-200, 50-150, 50-100, 80-2000, 80-1000, 80-500, 80-150, 80-100, 100- 2000, 100-1000, 100-500, 100-200, or 100-150 nucleotides in length.
Such bisulfite-converted nucleotide sequences can be used for detecting the meihylation status, for example, by an MSP reaction or by direct sequencing (e.g., next generation sequencing). These bisulfite-converted sequences are also of use for designing primers for MSP reactions that specifically detect methylated or unmethylated nucleotide sequences following bisulfite conversion, In yet other embodiments, the bisulfite-converted nucleotide sequences of the disclosure also include nucleo tide sequences that will hybridize under highly stringent conditions to any nucleotide sequence of any one or more of SEQ ID NOs: 1-8444 and 8447-16204, or fragments or complements thereof.
In further aspects, the application provides methods for producing such bisulfite- converted nucleotide sequences, for example, the application provides methods for treating a nucleotide sequence with a bisulfite agent such that the unmethylated eytosine bases are converted to a different nucleotide base such as a uracil. In yet other aspects, the application provides oligonucleotide primers for amplifying a region within the nucleic acid sequence of any one or more of SEQ ID NOs: 1-8444 and 8447-16204, In certain aspects, a pair of the oligonucleotide primers can be used in a detection assay, such as the Hpall assay. In certain aspects, primers used in an MSP reaction can specifically distinguish between methylated and non-methylated DNA.
The primers of the disclosure have sufficient length and appropriate sequence so as to provide specific initiation of amplification nucleic acids. Primers of the disclosure are- designed to be "substantially" complementary to each strand of the nucleic acid sequence to be amplified. In some embodiments, the primer is selected from the group consisting of sequences having 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%», 97%, 98%, 99%, or 100% identity to any of SEQ ID NOs: 8377-8404, 8435-8446, 12635-12646, and 12665- 12670, In some embodiments, the primer comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 .1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, or 35 consecutive nucleotides of any of the primer sequences of SEQ ID NOs: 8377-8404, 8435- 8446, 12635-12646, and 12665-12670. While exemplary primers include the sequences of any sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%», or 100% identity to any of SEQ ID NOs: 8377-8404, 8435-8446, 12635-12646, and 12665-12670, or fragments thereof it is understood that any primers that hybridize with the bisulfite-eonverted sequence of any one or more of SEQ ID NOs: 1-8444 and 8447-16204 are included within the scope of this disclosure and is useful in the method of the disclosure for detecting methylated nucleic acid, as described. Similarly, it is understood that any primers that would serve to amplify a methyl atiori sensitive restriction site or sites within the differentially methylated region of the informative loci of any of the sequences of SEQ ID NOs: 1-8444 or 8447-16204, or fragments or complements thereof are included within the scope of this disclosure and is useful in the method of the disclosure for detecting nucleic methylated nucleic acid, as described.
The oligonucleotide primers of the disclosure may be prepared by using any suitable method, such as conventional phosphotriester and phosphodiester methods or automated embodiments thereof. In one such automated embodiment, dietbylphosphoramidites are used as starting materials and may be synthesized as described by Beaucage, et al. {Tetrahedron Letters, 22: 1859-1862, 1981). One method of synthesizing oligonucleotides on a modified solid support is described in U.S. Patent No. 4,458,066,
In certain aspects, the application provides assays and methods using any of the informative loci, or bisulfite converted methylated or unmethyiated sequences thereof! disclosed herein. In some embodiments, the informative loci comprise a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any of the sequences of SEQ ID NOs: 1-428, 2569-2996, 5137-5531 , 7507-7532, 7663- 7668, 7819-7842, 7963-7976, 8047-8060, 8131-8143, 8209-8222, 8293-8306, 8405-8409, 8447-8632, 9563-9748, 1067940825, 1 1561-1161 1 , 1 1867-1 191 7, 12173-12219, 12455-
12460, 12491-12496, 12527-12532, 12563-12568, 12599-12604, 12647-12649, 12671-12908, 14093-14329, 15515-15537, 1565345692, 15893-15932, 16133-16137, 16163-16165, 16181 -16183, 16199, 429-856, 2997-3424, 5532-5926, 7533-7558, 7689-7714, 7843-7866, 7977-7990, 8061-8074, 8144-8156, 8251 -8264, 8335-8348, 8420-8424, 8633-8818, 9749- 9934, 10826-10972, 1 1612-1 1662, 119184 1968, 12220-12266, 12461-12466, 12497-12502, 12533-12538, 12581-12586, 12617-12622, 12656-12658, 12909-13144, 14330-14566, 15538-15560, 15693-15732, 159334 5972, 16138-16142, 16166-16168, 16184-16186 or 16200 or any fragments or complements thereof, In some embodiments, the informative loci are used as molecular markers to distinguish between healthy ceils and metaplastic ceils (e.g., Barrett's esophageal cells). In some embodimenis, the informative loci are used as molecular markers to distinguish between healthy cells and neoplastic cells (e.g., cancer cells). In particular embodiments, the informative loci are used as molecular markers to distinguish between healthy cells and esophageal adenocarcinoma cells, In some embodiments, the informative loci are used as molecular markers to distinguish between Barrett's esophagus cells and cancer cells. In some embodiments, the informative loci are used as molecular markers to distinguish between Barrett's esophagus cells and esophageal adenocarcinoma eel is. For example, in one embodiment, the application provides methods and assays using any of the informative loci comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any one or more of SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 1 1561-1 1662, 1 1867-1 1968, 12173-12266, 12455- 12466, 12491-12502, 12527-12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649, OR 12656-12658, or any fragments or complements thereof as markers that distinguish between healthy cells and neoplasia cells. In other embodiments, the application provides methods and assays using the informative loci comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any one or more of SEQ ID NOs: 1 -856, 2569-3424, 5137-5926, 7507-7558, 7663-7714, 7819- 7866, 7963-7990, 8047-8047, 8131-8156, 8209-8222, 8251-8264, 8293-8306, 8335-8348, 8405-8409, 8420-8424, 8447-8818, 9563-9934, 10679-10972, 11561-1 1662, 1 1867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649, 12656-12658, 12671-13144, 14093-14566,
15515-15560, 15653-15732, 15893-15972, 16135-16142, 16163-16168, 16181-16186 and/or 16199-16200 or any fragments or complements thereof as markers that distinguish between healthy cells and cells derived from neoplasias of the upper gastrointestinal tract. In one aspect, a molecular marker of the invention is a differentially methylated sequence of an informative locus. In certain aspects, the application provides assays and methods using the informative loci comprising a sequence having at least. 80%, 85%, 90%, 9.1 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any one or more of SEQ ID NOs: 1- 856, 2569-3424, 5137-5926, 7507-7558, 7663-7714, 7819-7866, 7963-7990, 8047-8047, 8131-8156, 8209-8222, 8251-8264, 8293-8306, 8335-8348, 8405-8409, 8420-8424, 8447- 8818, 9563-9934, 10679-10972, 1 1561-11662, 1 1867-1 1968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 1261 7-12622, 12647-12649, 12656-12658, 12671-13144, 14093-14566, 15515-15560, 15653-15732, 15893-15972, 16135-16142, 16163-16168, 16181 -16186 and/or 16199-16200 or any fragments or complements thereof in combination with the status of somatic mutation(s) in TP53 as molecular markers that distinguish between healthy cells and cancer cells, For example, in one embodiment, the application provides methods and assays using the informative loci comprising a sequence having at least 80%o, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any one or more of SEQ ID NOs: 1 - 856, 2569-3424, 5137-5926, 7507-7558, 7663-7714, 7819-7866, 7963-7990, 8047-8047, 8131-8156, 8209-8222, 8251-8264, 8293-8306, 8335-8348, 8405-8409, 8420-8424, 8447- 8818, 9563-9934, 10679- 10972, 1 1561-1 1662, 1 1867-1 1968, 121 73-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 1259942604, 12617-12622, 12647-12649, 12656-12658, 1267143144, 14093-14566, 15515-15560, 15653-15732, 15893-15972, 16135-16142, 16163-16168, 16181 -16186 and/or 16199-16200 or any fragments or complements thereof and the status of somatic niutation(s) in TP53 as markers that distinguish between healthy cells and neoplasia cells (e.g. eaiicer/esophageal
adenocarcinoma cells). In other embodiments, the application provides methods and assays using the informative loci disclosed herein (e.g., chromosomal loci Up1 5-1, Up35-1, Up35-2, Up3, Up27, and Up 10) and the status of somatic mutation(s) in TP53 as markers that distinguish between healthy cells and cells derived from neoplasias of the upper
gastrointestinal tract, in one aspect, a molecular marker of the invention is a differentially methylated sequence of an informative locus.
In certain embodiments, the application provides assays for detecting differentially methylated nucleotide sequences. Thus, a differentially methylated nucleotide sequence, in its methylated state, can serve as a target for detection using various methods described herein and the methods that are well within the purview of the skilled artisan in view of the teachings of this application.
In certain aspects, such methods for detecting methylated nucleotide sequences are based on treatment of genomic DNA with a chemical compound which converts non- methylated C, but not methylated C (i.e., 5mC), to a different nucleotide base. One such compound is sodium bisulfite (also referred to simply as "bisulfite" herein), which converts C, but not 5mC, to U. Methods for bisulfite treatment of DNA are known in tire art (Herman, et ah, 1996, Proc Natl Acad Sei USA, 93:9821-6; Herman and Baylin, 1998, Current Protocols in Human Genetics, N, B, A. Dracopoli, ed., John Wiley & Sons, 2:10.6.1-10.6.10; U.S.
Patent No. 5,786,146). To illustrate, when a DNA molecule that contains unmethylated C nucleotides is treated with sodium bisulfite to become a compound-converted DNA, the sequence of that DNA is changed (C→ U). Detection of the U in the convened nucleotide sequence is indicative of an unmethyiated C.
The different nucleotide base (e.g., U) present in compound-converted nucleotide sequences can subsequently be detected in a variety of ways. In a particular embodiment, the present invention provides a method of detecting U in compound-converted DNA sequences by using "methylation sensitive PCR" (MSP) (see, e.g., Herman, et a!,, 1996, Proc. Natl. Acad Sci. USA, 93:9821-9826; U.S. Patent No. 6,265,171 ; U.S. Patent No, 6,017,704; U.S. Patent No. 6,200,756). In MSP, one set of primers (i.e., comprising a forward and a reverse primer) amplifies the compound-converted template sequence if C bases in CpG dinucieotides within the DNA are methylated. This set of primers is called "methylation-specific primers." Another set of primers amplifies the compound-converted template sequence if C bases in CpG dinucieotides within the 5' flanking sequence are not methylated. This set of primers is called "unmeihylation-specific primers."
In MSP, the reactions use the compound-converted DNA from a sample in a subject. In assays for methylated DNA, methylation- specific primers are used, in the case where C within CpG dinucieotides of the target sequence of the DNA are methylated, the methylaiion- specific primers will amplify the compound-converted template sequence in the presence of a polymerase and an MS P product will be produced. If C within CpG dinucieotides of the target sequence of the DNA is not methylated, the methylaiion-specific primers will not amplify the compound-converted template sequence in the presence of a polymerase and an MSP product will not be produced. In some embodiments, any of the bisulfite converted methylated sequences disclosed herein is used as a marker for a particular indication.
it is often also useful to run a control reaction for the detection of unmethylated DNA. The reaction uses the compound-converted DNA from a sample in a subject and
unmethylation-speeific primers are used. In the case where C within CpG dinucieotides of the target sequence of the DNA are unmethylated, the unmethylation specific primers will amplify the compound-converted template sequence in the presence of a polymerase and an MSP product will be produced. If C within CpG dinucieotides of the target sequence of the DNA is methylated, the unmethylation-specific primers will not amplify the compound- converted template sequence in the presence of a polymerase and an MSP product will not be produced. Note that a biologic sample will often contain a mixture of both neoplastic cells that give rise to a signal with methylation specific primers, and normal cellular elements that give rise to a signal with unmethylation-speeific primers. The unmethylation specific signal is often of use as a control reaction, but does not in this instance imply the absence of neoplasia as indicated by the positive signal derived from reactions using the methylation specific primers. In some embodiments, any of the bisulfite converted unmethylated sequences disclosed herein are used as controls. In some embodiments, the unmethylated control sequences are any of the bisulfite converted sequences of SEQ ID NOs: 857-1284, 3425-3852, 5927-6321, 7559-7584, 7715-7740, 7867-7890, 7991-8004, 8075-8088, 8157- 8169, 8223-8236, 8307-8320, 8410-8414, 8819-9004, 9935-10120, 10973-1 1119, 1 1663- 1 1713, 1 1969-12019, 12267-12313, 12467-12472, 12503-12508, or 12539-12544, 12569- 12574, 12605-12610, 12650-12652,1713-2140, 4281-4708, 6717-71 1 1, 761 1-7636, 7767- 7792, 7915-7938, 8019-8032, 8103-81 16, 8183-8195, 8265-8278, 8349-8362, 8425-8429, 9191-9376, 10307-10492, 1 1267-1 1413, 1 1765-11815, 12071-12121 , 12361 -12407, 12479- 12484, 12515-12520, 12551-12556, 12587-12592, 12623-12628, or 12659-12661 in which every "Y" position is a "T."
Primers for a MSP reaction are derived from the compound-converted template sequence. Herein, "derived from" means that the sequences of the primers are chosen such that the primers amplify the compound-converted template sequence in a MSP reaction. Each primer comprises a single-stranded DNA fragment which is at least 8 nucleotides in length. In some embodiments, the primers are less than 50 nucleotides in length, or in some embodiments, from 15 to 35 nucleotides in length. Because the compound -converted template sequence can be either the Watson strand or the Crick strand of the double-stranded DNA that is treated with sodium bisulfite, the sequences of the primers is dependent upon whether the Watson or Crick compound-converted template sequence is chosen to be amplified in the MSP. Either the Watson or Crick strand can be chosen to he amplified.
The compound-converted template sequence, and therefore the product of the MSP reaction, is, in some embodiments, between 20 to 3000 nucleotides in length, in other embodiments, the product of the MSP reaction is between 50 to 1000 nucleotides in length, in other embodiments, the product of the MSP reaction is between 50 to 500 nucleotides in length. In other embodiments, the product of the MSP reaction is between 80-150 nucleotides in length, in some embodiments, the product of the MSP reaction is at least 20, 30, 40, 50, 60, 70. 80, 90, 100, 1 10, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240 or 250 nucleotides in length. In some embodiments, the meihylation-specific primers result in an MSP product of a different length than the MSP product produced by the unmethylation- specific primers.
A variety of methods can be used to determine if an MSP product has been produced in a reaction assay. One way to determine if an MSP product has been produced in the reaction is to analyze a portion of the reaction by agarose gel electrophoresis. For example, a horizontal agarose gel of from 0.6 to 2.0% agarose is made and a portion of the MSP reaction mixture is electrophoresed through the agarose gel. After electrophoresis, the agarose gel is stained with ethidium bromide. MSP products are visible when the gel is viewed during illumination with ultraviolet light. By comparison to standardized size markers, it is determined if the MSP product is of the correct expected size.
Other methods can be used to determine whether a product is made in an MSP reaction. One such method is called "real-time PGR." Real-time PGR utilizes a thermal cycler (i.e., an instrument thai provides the temperature changes necessary for the PGR reaction to occur) that incorporates a fiuorimeter (i.e. an instrument that measures
fluorescence). The real-time PGR reaction mixture also contains a reagent whose
incorporation into a product can be quantified and whose quantification is indicative of copy number of that sequence in the template. One such reagent is a fluorescent dye, called SYBR Green I (Molecular Probes, Inc.; Eugene, Oregon) that preferentially binds double-siranded DNA and whose fluorescence is greatly enhanced by binding of double-stranded DNA.
When a PGR reaction is performed in the presence of SYBR Green I, resulting DNA products bind SYBR Green I and fluorescence. The fluorescence is detected and quantified by the fiuorimeter. Such technique is particularly useful for quantification of the amount of the product in the PGR reaction. Additionally, the product from the PGR reaction may be quantitated in "real-time PGR" by the use of a variety of probes thai hybridize to the product including TaqMan probes and molecular beacons. Quantitation may be on an absolute basis, or may be relative to a constitutively methylated DNA standard, or may be relative to an unmethylated DNA standard. In one instance the ratio of methylated derived product to unmethylated derived product may be constructed.
Methods for detecting methylation of the DNA according to the present disclosure are not limited to MSP, and may cover any assay for detecting DNA methylation. Another example method of detecting methylation of the DNA is by using "methylation-sensitive" restriction endonucleases. Such methods comprise treating the genomic DNA isolated from a subject with a methylation-sensitive restriction endonuclease and then using the restriction endonucSease-treated DNA as a template in a PGR reaction. Herein, methylation-sensitive restriction endonucleases recognize and cleave a specific sequence within the DNA if C bases within the recognition sequence are not methylated, If C bases within the recognition sequence of the restriction endonuclease are methylated, the DNA will not be cleaved, Examples of such methylation-sensitive restriction endonucleases include, but are not limited to Hpai l, SmaL SacII, Eagl, BstUi, and BssHIL In this technique, a recognition sequence for a methylation-sensitive restriction endonuclease is located within the template DNA, at a position between the forward and reverse primers used for the PGR reaction. In the case that a C base within the methylation-sensitive restriction endonuclease recognition sequence is not methylated, the endonuclease will cleave the DNA template and a PGR product will not be formed when the DNA is used as a template in the PGR reaction, in the case that a C base within the methylation-sensitive restriction endonuclease recognition sequence is methylated, the endonuclease wilt not cleave the DNA template and a PGR product will be formed when the DNA is used as a template in the PGR reaction. Therefore, methylation of C bases can be determined by the absence or presence of a PGR product (Kane, et a!,, 1997, Cancer Res, 57:808-1 1). No sodium bisulfite is used in this technique.
Yet another exemplary method of detecting methylation of the DNA is called the modified MSP, which method utilizes primers that are designed and chosen such that products of the MSP reaction are susceptible to digestion by restriction endonucleases, depending upon whether the compound-converted template sequence contains CpG dinueleotides or UpG ciinucleotides.
Yet other methods for detecting methylation of the DNA include the MS-SnuPE methods. This method uses compound-converted DNA as a template in a primer extension reaction wherein the primers used produce a product, dependent upon whether the compound- converted template contains CpG dinueleotides or UpG dinueleotides (see e.g., Gonzalgo, et al„ 1997, Nucleic Acids Res,, 25:2529-31). Another exemplary method of detecting rnethylation of the DNA is called COBRA (i.e., combined bisulfite restriction analysis). This method has been routinely used for DNA rnethylation detection and is well known in the art (see, e.g., Xiong, et al., 1997, Nucleic Acids Res, 25:2532-4), In this technique, methylation-sensitive restriction endonucleases recognize and cleave a specific sequence within the DNA if C bases within the recognition sequence are methylated. If C bases within the recognition sequence of the restriction endonuclease are not methylated, the DNA will not be cleaved. In some embodiments, the method utilizes methylation-sensitive restriction endonucleases.
Another exemplary method of detecting rnethylation of DNA requires hybridization of a compound converted DNA to an-ays that include probes that hybridize to sequences derived from a methylated template.
Another exemplary method of detecting rnethylation of DNA includes precipitation of methylated DNA with antibodies that bind meihylated DNA or with other proteins that hind methylated DNA, and then detection of DNA sequences in the precipitate. The detection of DNA could be done by PGR based methods, by hybridization to arrays, or by other methods known to those skilled in the art.
Another exemplary method of detecting methylated DNA is bisulfite sequencing that involves amplification of a target region of bisulfite converted DNA using rnethylation indifferent PGR primers that amplify converted DNAs derived from both methylated and unmethyiated templates. The rnethylation indifferent primers are often designed to be both rnethylation indifferent and bisulfite specific, i.e. to amplify only bisulfite converted target DNAs and not to amplify non-converted target sequences, in some embodiments, the amplified DN As then may be characterized by Next Generation Sequencing methods that allow each cytosine base in the original template to be assessed within each DNA sequence read for the presence of rnethylation (retention of cytosine) or the absence of rnethylation (conversion to thymidine). The percent of rnethylation at each cytosine base in the original template can then be calculated by the percent of DNA reads in which the cytosine is preserved as cytosine versus is converted to thymidine. Similarly, the percent of rnethylation across a region of interest can be assessed by determining a rule for assessing the region as methylated or unmethyiated in an individual DNA read (i.e. determining a cutoff for methylation in the region that will categorize the region as "methylated"), and then determining the percent of DNA reads in which the region qualifies as methylated.
In certain embodiments, the disclosure provides methods that involve directly sequencing the product resulting from an MSP reaction to determine if the compound- converted template sequence contains CpG dinucleotides or UpG dinucleotides. Molecular biology techniques such as directly sequencing a PGR product are well known in the art.
In some embodiments, methylation of DNA may be measured as a percentage of total DNA. High levels of methylation may be 1-100% methylation, for example, 1%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% methylation. Low levels of methylation may be 0%-0.99% methylation, for example, 0%, 0.1 %, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%. At least some normal tissues, for example, normal esophagus samples, may not have any detectable methylation.
The skilled artisan will appreciate that the present disclosure is based in part, on the recognition that any one of the informative loci disclosed herein may include nucleotide sequences that encode polypeptides that, for example, may function as a tumor suppressor gene. Accordingly, the application further provides methods for detecting such polypeptides in cell samples, in some embodiments, the disclosure provides detection methods by assaying such polypeptides so as to determine whether a patient has or does not have a disease condition. Further, such a disease condiiion may be characterized by decreased levels of such polypeptides. In certain embodiments, the disclosure provides methods for determining whether a patient is or is not likely to have cancer by detecting such polypeptides. In further embodiments, the disclosure provides methods for determining whether the patient is having a relapse or determining whether a patient's cancer is responding to treatment.
Optionally, such methods involve obtaining a quantitative measure of the protein in the sample. In view of this specification, one of skill in the art will recognize a wide range of techniques that may be employed to detect and optionally quantitate the presence of a protein. In some embodiments, a protein is detected with an antibody. In many embodiments, an antibody-based detection assay involves bringing the sample and the antibody into contact so that the antibody has an opportunity to bind to proteins having the corresponding epitope. In many embodiments, an antibody-based, detection assay also typically involves a system for detecting the presence of antibody-epitope complexes, thereby achieving a detection of the presence of the proteins having the corresponding epitope. Antibodies may be used in a variety of detection techniques, including enzyme-linked immunosorbent assays (ELISAs). immunoprecipitations, Western blots. Antibody-independent techniques for identifying a protein may also be employed. For example, mass spectroscopy, particularly coupled with liquid chromatography, permits detection and quantification of large numbers of proteins in a sample. Two-dimensional gel electrophoresis may also be used to identify proteins, and may be coupled with mass spectroscopy or other detection techniques, such as N-terminal protein sequencing, RNA aptamers with specific binding for the protein of interest may also be generated and used as a detection reagent. Samples should generally be prepared in a. manner that is consistent with the detection system to be employed. For example, a sample to be used in a protein detection system should generally be prepared in the absence of proteases.
Likewise, a sample to be used in a nucleic acid detection system should generally be prepared in the absence of nucleases. In many instances, a sample for use in an antibody-based detection system will not be subjected to substantial preparatory steps. For example, urine may be used directly, as may saliva and blood, although blood will, in certain embodiments, be separated into fractions such as plasma and serum.
in certain embodiments, a method of the disclosure comprises detecting the presence of an informative loci-expressed nucleic acid, such as an mRNA, in a sample, Optionally, the method involves obtaining a quantitative measure of the informative loci-expressed nucleic acid in the sample, in view of this specification., one of skill in the art will recognize a wide range of techniques that may be employed to detect and optionally qiiantitate the presence of a nucleic acid. Nucleic acid detection systems generally involve preparing a purified nucleic acid fraction of a sample, and subjecting the sample to a direct detection assay or an amplification process followed by a detection assay. Amplification may be achieved, for example, by polymerase chain reaction (PGR), reverse transcriptase (RT) and coupled RT- PCR. Detection of a nucleic acid is generally accomplished by probing the purified nucleic acid fraction with a probe that hybridizes to the nucleic acid of interest, and in many instances, detection involves an amplification as well, Northern blots, dot blots, microarrays, quantitative PGR, and quantitative RT-PCR are all well known methods for detecting a nucleic acid in a sample.
In certain embodiments, the disclosure provides nucleic acid probes that bind specifically to an informative loci nucleic acid. Such probes may be labeled with, for example, a fluorescent moiety, a radionuclide, an enzyme or an affinity tag such as a biotin moiety. For example, the TaqMan® system employs nucleic acid probes that are labeled in such a way that the fluorescent signal is quenched when the probe is free in solution and bright when the probe is incorporated into a larger nucleic acid.
immunoscintigraphy using monoclonal antibodies directed at the informative Iocs may be used to detect and/or diagnose a cancer. For example, monoclonal antibodies against the informative loci labeled with "Technetium, 11 'indium, 325Iodine-may be effectively used for such imaging. As will be evident to the skilled artisan, the amount of radioisotope to be administered is dependent upon the radioisotope. Those having ordinary skill in the art can readily formulate the amount of the imaging agent to be administered based upon the specific activity and energy of a gi ven radionuclide used as the active moiety. Typically 0.1 -100 millicuries per dose of imaging agent. 1-10 millicuries, or often 2-5 millicuries are administered. Thus, compositions according to the present invention useful as imaging agents comprising a targeting moiety conjugated to a radioactive moiety comprise 0.1-100 millicuries, in some embodiments 1 -10 millicuries, in some embodiments 2-5 millicuries, in some embodiments 1 -5 millicuries.
A variety of methods can be used to determine if TP 53 contains a somatic mutation, as will be evident to the skilled artisan. In some embodiments, the TP53 gene or protein sequence is determined and any change in the determined sequence relative to the wildtype sequence is detected. In some embodiments, the TP53 gene sequence is determined by PGR, RT-PCR, Northern Blot, Southern Blot, and/or in situ hybridization, Another way to determine if TP53 contains a somatic mutation may involve the use of an antibody- based detection assay (e.g. ELISA, immunohistocheniistry, and/or Western Blot). In some embodiments, the antibody-based detection assay utilizes an antibody that binds to a mutant TP53 protein with a tighter affinity than it binds to a wildtype TP53 protein. The skilled artisan will also readily appreciate methods of determining somatic mutations in TP53 based on the disclosures of US5843654, US5620848, EP0390323 and US5527676, ail of which are herein incorporated by reference in their entirely.
In some embodiments, the disclosure provides for a device useful for detecting the methylaiion status of any of the informative loci, or fragments or complements thereof, disclosed herein, in some embodiments, the disclosure provides for a kit comprising components useful for detecting the methylaiion status of the informative loci, or fragments, or complements thereof, disclosed herein. In some embodiments, the kit comprises a swallowable balloon for collecting an esophageal sample from the subject. In some embodiments, the kit comprises any of the swallowable balloon devices disclosed in WO 2015/089422, which is incorporated herein in its entirety.
In certain embodiments, the present disclosure provides drug screening assays for identifying test compounds which potentiate the tumor suppressor function of polypeptides encoded by sequences located in the informative loci disclosed herein. In one aspect, the assays detect test compounds which potentiate the expression level of polypeptides encoded by sequences located in the informative loci disclosed herein. In another aspect, the assays detect test compounds which inhibit the methylaiion of DNA, In certain embodiments, drag screening assays can be generated which detect test compounds on the basis of their ability to interfere with stability or function of polypeptides encoded by sequences located in the informative loci disclosed herein.
A variety of assay formats may be used and, in light of the present disclosure, those not expressly described herein will nevertheless be considered to be within the purview of ordinary skill in the art. Assay fomiats can approximate such conditions as protein expression level, methylaiion status of nucleotide sequences, tumor suppressing activity, and may be generated in many different forms. In many embodiments, the disclosure provides assays including both cell-free systems and cell-based assays which utilize intact cells.
Compounds to be tested can be produced, for example, by bacteria, yeast or other organisms (e.g., natural products), produced chemically (e.g., small molecules, including peptidomimetics), or produced recombinantly. The efficacy of the compound can be assessed by generating dose response curves from data obtained using various concentrations of the test compound. Moreover, a control assay can also be performed to provide a baseline for comparison. In the control assay, the formation of complexes is quantiiated in the absence of the test compound.
in many drug screening programs which test libraries of compounds and natural extracts, high throughput assays are desirable in order to maximize the number of compounds surveyed in a given period of time. Assays of the present invention which are performed in cell-free systems, such as may he developed with purified or semi-purified proteins or with lysates, are often preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound. Moreover, the effects of cellular toxicity and/or bioavailability of the test compound can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an alteration of binding affinity with other proteins or changes in enzymatic properties of the molecular target,
In certain embodiments, test compounds identified from these assays may be used in a therapeutic method of treating cancer.
Still another aspect of the application provides transgenic non-human animals which express a gene located within any one of the informative loci disclosed herein, or which have had one or more of such genomic gene(s) disrupted in at. least one of the tissue or cell-types of the animal.
In another aspect, the application provides an animal model for cancer, which has a mis-expressed allele of a gene located within any one of the informative loci disclosed herein. Such a mouse model can then be used to study disorders arising from mis-expression of genes located within any one of the informative loci disclosed herein.
Genetic techniques which allow for the expression of transgenes can be regulated via site-specific genetic manipulation in vivo are known to those skilled in the art. For instance, genetic systems are available which allow for the regulated expression of a recombinase that catalyzes the genetic recombination a target sequence. As used herein, the phrase "target sequence" refers to a nucleotide sequence that is genetically recombined by a recombinase, The target sequence is flanked by recombinase recognition sequences arid is generally either excised or inverted in cel ls expressing recombinase activity. Recombinase catalyzed recombination events can be designed such that recombination of the target sequence results in either the activation or repression of expression of the polypeptides. For example, excision of a target sequence which interferes with the expression of a recombinant gene can he designed to acti vate expression of that gene. This interference with expression of the protein can result from a variety of mechanisms, such as spatial separation of the gene from the promoter element or an internal stop codon, Moreover, the transgene can be made wherein the coding sequence of the gene is flanked recombinase recognition sequences and is initially transfeeted into cells in a 3' to 5' orientation with respect to the promoter element, in such an instance, inversion of the target sequence wall reorient the subject gene by placing the 5' end of the coding sequence in an orientation with respect to the promoter element, which aHow? for promoter driven transcriptional activation.
in an illustrative embodiment, either the crelloxP recombinase system of
bacteriophage PI (Lakso et al, (1992) Proc, Natl. Acad. Sci. USA 89:6232-6236; Orban et al, (1992) Proc. Natl. Acad. Sci. USA 89:6861-6865) or the FLP recombinase system of
Saccharomyces cerevislae (O'Gorraan et aL, (1991 ) Science 251 : 1351-1355; PCX publication WO 92/15694) can be used to generate in vivo site-specific genetic recombination systems. Cre recombinase catalyzes the site-specific recombination of an intervening target sequence located between loxP sequences. loxP sequences are 34 base pair nucleotide repeat sequences to which the Cre recombinase binds and are required for Cre recombinase mediated genetic recombination. The orientation of loxP sequences determines whether the intervening target sequence is excised or inverted when Cre recombinase is present (Abremski et al, (1984) J Biol. Chem. 259:1509-1514); catalyzing the excision of the target sequence when the loxP sequences are oriented as direct repeats and catalyzes inversion of the target sequence when loxP sequences are oriented as inverted repeats.
V. Subjects and Samples
In certain aspects, the invention relates to a subject suspected of having or 1ms: a cancer, a metaplasia, or a neoplasia of the upper gastrointestinal tract (e.g., esophageal cancer). Alternatively, a subject may be undergoing routine screening and may not necessarily be suspected of having such metaplasia or neoplasia. In some embodiments, the subject is a human subject, and the neoplasia is a neoplasia of the upper gastrointestinal tract, such as the esophagus, in some embodiments, the subject is a human subject, and the metaplasia is Barrett's esophagus.
Assaying for biomarkers discussed above in a sample from subjects not known to have, e.g., a metaplasia or neoplasia of the upper or lower gastrointestinal tract can aid in diagnosis of such a metaplasia or neoplasia in the subject. To illustrate, detecting the methylation status of the nucleotide sequences by MSP can be used by itself, or in
combination with detecting the somatic mutation status of TPS 3 or other various assays, to improve the sensitivity and/or specificity for detecting, e.g., a neoplasia of the upper or lower gastrointestinal tract, in some embodiments, such detection is made at an early stage in the development of cancer, so that treatment is more likely to be effective.
In some embodiments, an informative loci in a subject is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma) if the loci is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% methylated. In some embodiments, a DNA sample from a subject is treated with bisulfite, and the resulting bisulfite sequence
corresponds to any of the nucleotide sequences di sclosed herein comprising a "Y" nucleotide. In some embodiments, if at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, I I , 12, 13, 14, 15, 16. 17, 1 8, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues of the bisu!fite-converted sequence have a C, the sequence is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma). In some embodiments, a DNA sample from a subject is treated with bisulfite, and die resulting bisulfite sequence corresponds to any of the nucleotide sequences disclosed herein comprising a "Y" nucleotide. In some embodiments, if at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues of the bisulfite-converted sequence have a C, the sequence is considered "methylated" for the purposes of determining whether or not the subject is prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g. , Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma), In some embodiments, a subject is determined to be prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) (e.g., esophageal cancer such as esophageal adenocarcinoma) if a certain number of "Y"
nucleotides in a bisulfite converted sequence are cytosines. In some embodiments, the certain number is at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 of the Y residues of the bisuifite-converted sequence. In some embodiments, the certain number is least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the Y residues of the bisuifite-converted sequence. In certain embodiments, a subject is determined to be prone to developing and/or has developed a metaplasia in the esophagus (e.g., Barrett's esophagus) or neoplasia (e.g., esophageal cancer such as esophageal adenocarcinoma) (e.g., Barrett's esophagus with dysplasia such as high-grade or low-grade dysplasia) if a certain percentage of DNA molecules from a sample from a subject are determined to be "methylated," as defined herein. In some embodiments, the certain percentage of DNA molecules is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the DNA molecules from the sample are determined to be "methylated." In some embodiments, the the percentage of methylated DNA molecules is determined using next- generation sequencing. Exemplary cut-offs of DNA methylation and DN A molecule percentages may be found in the Examples section provided herein.
In addition to diagnosis, assaying of a marker in a sample from a subject not known to have, e.g., a metaplasia or neoplasia of the upper gastrointestinal tract, can be prognostic for the subject (i.e., indicating the probable course of the disease). To illustrate, subjects having a predisposition to develop a metaplasia or neoplasia of the upper gastrointestinal tract may possess methylated nucleotide sequences. Assaying of methylated informative loci in a sample from subjects either by itself, or in combination with assaying for somatic mutation(s) in TP53, can also be used to select a particular therapy or therapies which are particularly effective against, e.g., a neoplasia of the upper gastrointestinal tract in the subject, or to exclude therapies that are not likely to be effective. Assaying of methylated informative loci in samples from subjects that are known to have, or io have had, a cancer is also useful. For example, the present methods can be used to identify whether therapy is effective or not for certain subjects. One or more samples are taken from the same subject prior to and following therapy, and assayed for any one or more of the informative loci markers either by itself or themselves, or in combination with assaying for somatic mutation(s) in TP53. A finding that an informative locus is methylated in the sample taken prior to therapy and absent (or at a lower level) after therapy may indicate that the therapy is effective and need not be altered. In those eases where the informative locus is methylated in the sample taken before therapy and in the sample taken after therapy, it may be desirable to alter the therapy to increase the likelihood that the cancer will be reduced in the subject. Thus, the present method may obviate the need to perform more invasive procedures which are used to determine a patient's response to therapy.
Cancers frequently recur following therapy in patients with advanced cancers. In this and other instances, the assays of the invention are useful for monitoring over time the status of a cancer associated with silencing of genes located in the informative loci disclosed herein. For subjects in whom a cancer is progressing, there can be no DNA methyiation in some or all samples when the first sample is taken and then appear in one or more samples when the second sample is taken, For subjects in which cancer is regressing, DNA methyiation may be present in one or a number of samples when the first sample is taken and then be absent in some or all of these samples when the second sample is taken.
Samples for use with the methods described herein may be essentially any biological material of interest. For example, a sample may be a bodily fluid sample from a subject, a tissue sample from a subject, a solid or semi-solid sample from a subject, a primary cell culture or tissue culture of materials derived from a subject, cells from a cell line, or medium or other extracellular material from a cell or tissue culture, or a xenograft (meaning a sample of a cancer from a first subject, e.g., a human, that has been cultured in a second subject, e.g., an immuno-compromised mouse). The term "sample" as used herein is intended to encompass both a biological material obtained directly from a subject (which may be described as the primary sample) as well as any manipulated forms or portions of a primary sample. A sample may also be obtained by contacting a biological material with an exogenous liquid, resulting in the production of a lavage liquid containing some portion of the contacted biological material. Furthermore, the term "sample" is intended to encompass the primary sample after it has been mixed with one or more additive, such as preservatives, chelators, anti-clotting factors, etc.
In certain embodiments, a bodily fluid sample is a blood sample. In this case, the term
"sample" is intended to encompass not only the blood as obtained directly from the patient but also fractions of the blood, such as plasma, serum, cell fractions (e.g., platelets, erythrocytes, and lymphocytes), protein preparations, nucleic acid preparations, etc. In some embodiments, the bodily .fluid may be derived from the stomach, for example, gastric secretions, acid reflux, or vomit, in other embodiments, the bodily fluid may be a fluid secreted by the pancreas or bladder. In other embodiments, the body fluid may be saliva, spit, or an esophageal washing. Irs certain embodiments, a tissue sample is a biopsy taken from the mucosa of the
gastrointestinal tract. In other embodiments, a tissue sample is the brushings from, e.g., the esophagus of a subject.
A subject is in some embodiments a human subject, bat it is expected that the molecular markers disclosed herein, and particularly their homologs from other animals, are of similar utility in other animals. In certain embodiments, it may be possible to detect a biomarker described herein (e.g., DNA raethylation or protein expression level) directly in an organism without obtaining a separate portion of biological material. In such instances, the term "sample" is intended to encompass that portion of biological material that is contacted with a reagent or device involved in the detection process.
In certain embodiments, DNA which is used as the template in an MSP reaction is obtained from a bodily fluid sample. Examples of bodily fluids are blood, saliva, spit or an esophageal washing. Other body fluids can also be used. Because they can be easily obtained from a subject and can be used to screen for multiple diseases, blood or blood-derived fractions are especially useful. Blood-derived fractions can comprise blood, serum, plasma, or other fractions. For example, a cellular fraction can be prepared as a "buffy coat" (i.e., leukocyte-enriched blood portion) by centrifuging 5 ml of whole blood for 10 min at 800 times gravity at room temperature. Red blood cells sedknent most rapidly and are present as the bottom-most fraction in the centrifuge tube. The buffy coat is present as a thin creamy white colored layer on top of the red blood ceils. The plasma portion of the blood forms a layer above the buffy coat. Fractions from blood can also be isolated in a variety of other ways. One method is by taking a fraction or fractions from a gradient used in centrifugation to enrich for a specific size or density of cells.
DNA is then isolated from samples from the bodily fluids. Procedures for isolation of DNA from such samples are well known to those skilled in the art. Commonly, such DNA isolation procedures comprise lysis of any cells present in the samples using detergents, for example. After cell lysis, proteins are commonly removed from the DNA using various proteases, RNA is removed using RNase. The DNA is then commonly extracted with phenol, precipitated in alcohol and dissolved in an aqueous solution.
VI. Therapeutic methods.
In some embodiments, the disclosure provides for a method of determining whether a subject has any one or more of the methylated informative loci disclosed herein that are indicative of the presence of a metaplasia in the esophagus (e.g., Barrett's esophagus), wherein if the subject is determined to have a metaplasia in the esophagus (e.g., Barrett's esophagus), the subject is treated with an agent that treats the metaplasia in the esophagus (e.g. , Barrett's esophagus). In some embodiments, the disclosure provides for a method of treating a subject determined to have a metaplasia in the esophagus (e.g., Barrett's esophagus), in some embodiments, the treatment of a metaplasia in the esophagus (e.g., Barrett's esophagus) encompasses administration of any one or more of the following compounds: proton pump inhibitors (PPIs) such as omeprazole (Prilosec, Zegerid), lansoprazole (Prevacid), pantoprazole (Protonix), rabeprazoie (AcipHex), esomeprazole (Nexium), dexlansoprazole (Dexllant). Histamine H2 receptor blocking agents such as eimetidine (Tagamet), ranitidine (Zantac), famotidine (Pepcid) and nizatidine (Axid). Turns, Rolaids, or other quick-acting reflux medications. Prokinetic agents, or drugs thai help move food through the gastrointestinal tract more quickly, offer an attractive alternative either alone or in combination with acid inhibition, In some embodiments, the treatment of a metaplasia in the esophagus (e.g., Barrett's esophagus) is endoscopic mucosal resection (EMR);
photodynaraic therapy (PDT); radiofrequency ablation (UFA); argon plasma coagulation (APC): cryotherapy, and/or surgery (e.g. esophagectomy, anti-reilux surgery). In some embodiments, the disclosure provides for a method of determining whether a subject has any one or more of the methylated informative loci disclosed herein that are indicative of esophageal neoplasia (e.g., esophageal cancer), wherein if the subject is determined to have an esophageal neoplasia (e.g. , esophageal cancer), the subject is treated with an agent that treats the esophageal neoplasia (e.g., esophageal cancer). In some embodiments, the disclosure provides for a method of determining whether a subject has any one or more of the methylated informative loci disclosed herein in combination with any of the TP53 somatic mutations disclosed herein that are indicative of esophageal neoplasia (e.g., esophageal cancer), wherein if the subject is determined to have an esophageal neoplasia (e.g., esophageal cancer), the subject is treated with an agent that treats the esophageal neoplasia (e,g.> esophageal cancer). In some embodiments, the disclosure provides for a method of treating a subject determined to have esophageal neoplasia (e.g., esophageal cancer), in some embodiments, the esophageal neoplasia is Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia (HGD) and/or esophageal adenocarcinoma (EAC). in some embodiments, the treatment of esophageal neoplasia (e.g., esophageal cancer) encompasses surgery (e.g. esophagectomy), radiation therapy, ehemoradiation therapy and/or chemotherapy. In some embodiments, the treatment of esophageal neoplasia (e.g., esophageal cancer) encompasses administering one or more chemotherapeutic agent, such as any one or more therapeutic agent selected from the group consisting of: carboplatm and paelitaxel (Taxol®) (which may be combined with radiation): cispiatin and 5-fluorouracil (5~ FU) (often combined with radiation); ECF: epirubicine (Ellence®), cispiatin, and 5-FU (especially for gastroesophageal junction tumors); DCF: docetaxe! (Taxotere®), cispiatin, and 5-FU; Cispiatin with capecitabine (Xeloda®); oxaliplatin and either 5-FU or capecitabine; doxorubicin (Adriamycin®), bleomycin, mitomycin, methotrexate, vinorelbine (Navelbine®), topotecan, and irinotecan (Camptosar®). in some embodiments, for some esophagus cancers that overexpress the HER2 protein on the surface of their cells, chemotherapy may be used, along with the targeted drug trastuzumab. Ramucirumab may be used to treat cancers that start at the gastroesophageal (GE) junction when they are advanced, in some embodiments, the treatment encompasses endoscopic treatments, such as endoscopic mucosal resection (EMR) followed by treatment with proton pump inhibitors, photodynarme therapy (PL)T); radiofrequency ablation (RFA); laser ablation; argon plasma coagulation (APC); electrocoagulation (electrofulguration); or esophageal stent.
The terms "treatment", "treating", "alleviation" and the like are used herein to generally mean obtaining a desired pharmacologic and/or physiologic effect, and may also be used to refer to improving, alleviating, and/or decreasing the severity of one or more symptoms of a condition being treated. The effect may be prophylactic in terms of completely or partially delaying the onset or recurrence of a disease, condition, or symptoms thereof, and/or may be therapeutic in terms of a partial or complete cure for a disease or condition and/or adverse effect attributable to the disease or condition. "Treatment" as used herein covers any ireatmeni of a disease or condition of a mammal, particularly a human, and includes: (a) preventing the disease or condition from occurring in a subject which may be predisposed to the disease or condition but has not yet been diagnosed as having it; (b) inhibiting the disease or condition (e.g., arresting its development); or (c) relieving the disease or condition (e.g., causing regression of the disease or condition, providing improvement in one or more symptoms).
Treating a metaplasia (e.g. , Barrett's esophagus) and/or a neoplasia (e.g. , esophageal cancer) in a subject refers to improving (improving the subject's condition), alleviating, delaying or slowing progression or onset, decreasing the severity of one or more symptoms associated with a metaplasia (e.g., Barrett's esophagus) and/or a neoplasia (e.g., esophageal cancer). For example, treating a metaplasia or neoplasia includes any one or more of:
reducing growth, proliferation and/or survival of metaplastic/neoplastic cells, killing metaplastic/neoplastic cells (e.g. , by necrosis, apoptosis or autophagy), decreasing
metaplasia/neoplasia size, decreasing rate of metaplasia/neoplasia size increase, halting increase in metaplasia/neoplasia size, improving ability to swallow, decreasing internal bleeding, decreasing incidence of vomiting, reducing fatigue, decreasing the number of metastases, decreasing pain, increasing survival, and increasing progression free survival. Exemplification
The invention now being generally described, it will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.
Example 1 : Identification of esophageal, cancer mformatiye loci.
Methylated informative loci were initially identified using the technique of reduced representation bisulfite sequencing (RRBS) in a discovery set of 23 paired biopsies of normal squamous esophagus and matched esophageal adenocarcinomas, along with biopsies of 8 Barrett's esophagus tissue, and along with brushings of 8 Barrett's esophagus tissues (one BE brushing case also having a matched biopsy).
Discovery data were initially analyzed for each individual CpG residue in the RRBS data set. Individuai CpGs were considered methylated in EAC if they showed methylation in less than 10% of DNA sequence reads in all of the informative squamous samples, where at least 4 squamous samples were informative, where an informative sample had equal to or greater than 20 reads covering the CpG, and if 8 or more of the informative EAC samples demonstrated percent methylation at a level that was at least 20 percentage points greater than the methylation le vel of the most methylated normal squamous sample. CpGs were similarly defined as methylated in Barrett's esophagus if they showed methylation of less than 10% of DNA sequence reads of ail informative squamous samples, where an informative sample had equal to or greater than 20 reads covering the CpG, and if 3 or more of the informative BE samples demonstrated percent methylation at a level that was at least 20 percentage points greater than the methylation level of the most methylated normal squamous sample. CpGs meeting criteria for methylation in both EACs and BE were defined as methylated in both EAC and BE, Such methylated CpGs were then aggregated into patches by grouping together methylated CpGs that were within 200bp of one another. Patches may consist of 1 CpG up to any number of CpGs that meet the above criteria.
The names assigned to 428 genomic patches defined as methylated in both EAC and BE by the criteria above were recorded, and the sequences of these loci correspond to SEQ ID NOs: 1 -856. The genomic coordinates of the genomic patches defined as methylated by the above criteria were also recorded. The genomic sequences of these patches on the respective genomic (+) and (-) strands were determined and recorded, (Upper and lower case designations were used according to those of the UCSC browser, where lower ease sequences are lower complexity DNA sequences). The bisulfite converted sequences of these corresponding patches(i,e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 857-1281 and 1713-2140 for the bisulfite converted sequences of the respective (+) and (-) strands). C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethy fated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as meihylation at that base. Thus, these sequences represent the group of ail combinations of all sequences in which 0, 1 , or more than one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the corresponding patches (i.e. in which all Y bases in every bisulfite converted sequence are retained as a C), corresponding to the (÷} strand the (-) strand were determined and recorded (see sequences of SEQ ID NOs: 1285-1712 and 2141 -2568 for the bisulfite converted sequences of the fully methy lated form of the (+) and (~) strands respectively of the corresponding patches). The reverse complements of the bisulfite converted methylated (+) stand and (-) stand sequences will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
Patches were expanded by 100 base pairs on either side so as to accommodate either the design of amplification pimers or to enclude additional presumptively methylated bases. The sequences of these expanded patches correspond to SEQ ID NOs: 2569-3424 and their genomic coordinates were also recorded. The genomic sequences of these expanded patches on the respective genomic (+) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences). The bisulfite converted sequences of these corresponding expanded patches (i.e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 3425-3852 and 4281 -4708 for the bisulfite converted sequences of the respective (+) and (-) strands). C residues that may be methylated or unmemyiated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base. Thus, these sequences represent the group of all combinations of all sequences in which 0, 1, or more than one Y is con verted to a T. The reverse complements of the bisulfite con verted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the corresponding expanded patches (i.e. in which all Y bases in every bisulfite converted sequence are retained as a C), corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ ID NOs: 3853-4280 and 4789-5136 for the bisulfite converted sequences of the fully methylated form of the {+) and (-) strands respectively of the corresponding expanded patches). The reverse complements of the bisulfite converted methylated (+) stand and (-) stand sequences will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
CpG islands overlapping patches that may contain additional CpGs that are methylated coordinately with patches were also defined. The sequences of these CpG islands correspond to SEQ ID NOs: 5137-5926, The genomic coordinates of the CpG islands were also recorded. The genomic sequences of these expanded patches on the respective genomic ( +) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DN A sequences). The bisulfite converted sequences of these corresponding CpG islands (i.e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 5927-6321 and 6717-711 1 for the bisulfite converted sequences of the respective (+) and (-) strands). C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base. Thus, these sequences represent the group of all combinations of all sequences in which 0. 1 , or more than one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the corresponding CpG islands (i.e. in which all Y bases in every bisulfite converted sequence are retained as a C). corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ) IE) NOs: 6322-6716 and 71 12-7506 for the bisulfite converted sequences of the fully methylated form of the (+) and (-) strands respectively of the corresponding CpG islands). The reverse complements of the bisulfite converted methylated (+) stand and (-) stand sequences will, be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
Regions of Interest (ROI) were defined that provided preferred regions for design of PGR araplicons that would encompass preferred patches. The genomic coordinates of the ROI were also recorded. The sequences of the (+) strands of the Regions of Interest correspond to SEQ ID NOs: 8209-8222, and the sequences of the (-) strands Regions of interest correspond to SEQ ID NOs: 8251-8261 , (Upper and lower case designations were used according to those of the UCSC browser, where lower ease sequences are lower complexity DNA sequences). The bisulfite converted sequences of these corresponding Regions of interest (i.e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 8223-8236 and 8265-8278 for the bisulfite converted sequences of the respective (+) and (-) strands). C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base. Thus, these sequences represent the group of all combinations of all sequences in which 0, 1 , or more than one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (+) and (-) s trands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the Regions of Interest (i.e. in which all Y bases in every bisulfite converted sequence are retained as a C), corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ ID NOs; 8237-8250 and 8279-8292 for the bisulfite converted sequences of the fully methylated form of the (÷) and (-) strands respectively of the corresponding Regions of Interest). The reverse complements of the bisulfite converted methylated (+) stand and (~) stand sequences will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
Specific PGR Amplicons were defined within the Regions of Interest (RGI). The genomic coordinates of the Arnplicons were recorded. The sequences of the (+) strands of the Amplicons correspond to SEQ ID NOs: 8293-8306 and 8405-8409, and the sequences of the (-) strands of the Arnplicons correspond to SEQ ID NOs: 8335-8348 and 8420-8424. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences). The bisulfite converted sequences of these Arnplicons (i.e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 8307-8320 and 8410-8414 for the bisulfite converted sequences of the (+) strands and see sequences of SEQ ID NOs: 8349-8362 and 8425-8429 and for die bisulfite converted sequences of the (-) strands), C residues that may be methylated or immethylatecl, and hence may be bisulfite converted to T (if immethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base. Thus, these sequences represent the group of all combinations of ail sequences in which 0, 1 , or more than one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the Arnplicons (he. in which all Y bases in every bisulfite converted sequence are retained as a C), corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ ID NOs: 8321-8334 and 8415-8419 for the bisulfite converted sequences of the fully methylated form of the (+) strands and see sequences of SEQ I D NOs: 8363-8376 and 8430-8434 for the bisulfite converted sequences of the fully methylated form of the (~) strands). The reverse complements of the bisulfite converted methylated (+) stand and (-) stand sequences will be obvious to one of ordinary skill in the art arjd are also included by implication in this disclosure. Sequences of PGR primers used in amplification of the Amplicons are provided as SEQ ID NOs: 8377-8404 and 8435-8444.
Confirmatory analysis of candidate loci was then done using bisulfite sequencing of candidate loci that were amplified using bisulfite specific but methylation independent amplification primers. This employed a new sample set. of biopsies from: 23 EACs, 8 HGD, 15 non-dysplastic BE from individuals without known higher grade lesions. In addition, biopsies were obtained from 5 eases of BE adjacent to HGD, and from 1 1 cases of BE adjacent to an EAC. These were not included in analyses of non-dysplastic BE. In addition biopsies were obtained from 33 normal squamous mucosa samples.
Table. 1 describes the performance in the confirmatory sample set using bisulfite sequencing analysis of amplicons of select loci having strong performance characteristics and identified in the studies discussed above. In Table 1, columns C - S disclose the performance of the select amplicons. For each DNA sequence read across each amplicon, the number of CpGs that were methylated was counted, and the read was classified as methylated or unmethyled using cutoffs for a required number of methylated CpGs on the amplicon. Table 1 , row 3 lists the number of CpGs between the amplification primers for each of the amplicons, Table 1 , row 4 lists the number of CpGs that need to be methylated on an individual read to count that read as methylated (e.g. for SqBE 2 there are 1 6 CpG residues between the primers, and 14+ (meaning > = 14) CpGs must be methylated on a read to score it as methylated. Table 1 , rows 6, 7, and 8 record the sensitivity for detecting esophageal adenocarcinomas (EACs) (row 6), high grade dysplasias (HGD) (row 7) and non-dysplastic Barrett's esophagus (non-dysplastic BE) (row 8), using criteria in which a sample was detected if it demonstrated methylation in greater than 10% (0.1) of all DNA reads. Table 1 , row 9 records the specificity of each amplicon for not detecting normal squamous mucosa again using criteria in which a sample was detected if it demonstrated methylation in greater than 10% (0.1) of all DNA reads, Table 1 , row 11 records the specificity of each amplicon for not detecting normal squamous mucosa now using criteria in which a sample was detected if it demonstrated methylation in greater than 1% (0,01) of all DNA reads. As a comparator, Table 1 , column B provides the same data for detecting methylation in the Vimentin (VIM) locus amplified using primers disclosed in Li et al. (Li M, et al. (2009) Sensitive digital uantification of DNA methylation in clinical samples. Nat Biotechnol 27(9):858-863).
hese primers correspond to SEQ ID NOs: 8445-8446. The amplicons amplified using these rimers are as follows:
Vimentin amplicon (+) strand (SEQ ID NO: 16207):
tTYOTttTttTAtYGtAGGATGTTYGGYGGttYGGGtAtYGYGAGtYGGtYGAGtTttA tYGGAGtTAYGTGAtTAYGTttAttYGtAttTAtAGttTGGGtAGt
Vimentin amplicon (-) strand (SEQ ID NO: 16208):
GtTGtttAGGtTGTAGGTGYGGGTGGAYGTAGTtAYGTAGtTtYGGtTGGAGtTYG tYGGtTYGYGGTGttYGGGtYGtYGAAtATttTGYGGTAGGAGGAYGAG
Amplicons (and patches) need not be used individually, but can be combined into anels for detection of esophageal neoplasia. Examples of such panels, and their associated erformance statistics, are provided in Table I , columns T through AG mat provide the markers in the panel and the sensitivity and specificity resulting from the marker combination when the combination is positive if any member of the combination is positive).
The sensitivity for detection of EAC (100%), HGD (88%), and BE (100%) is the same mong the combinations shown of: all amplicons, 17 amplicons, 15 amplicons, 4 amplicons, hree of four combinations of 3 amplicons (columns Y, Z, AA), and for one combination of 2 mplicons (column AF). Specificity for not detecting normal squamous (97%), at a detection utoff of 10% of reads being methylated, is the same for all combinations shown of: 15 mplicons, 4 amplicons, 3 amplicons, or 2 amplicons. When specificity is determined using a utoff of 1% of reads being methylated, men among amplicons with the highest sensitivity,he highest specificity is 94%, demonstrated by the combination of 3 amplicons of Table 1, olumn Z, followed by 91% specificity demonstrated by combinations shown of: 4 amplicons,wo combinations of three amplicons, and one combination of 2 amplicons.
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Discovery data were also analyzed for each individual CpG residue in the RRBS data set to identify loci that could be used to distinguish EAC from BE. Individual CpGs were considered methylated in EAC versus BE if they showed methylation of less than 10% of reads of all informative BE samples, where at least 3 BE samples were informative, and if they showed methylation of less than 10% of reads of all informative normal squamous samples, and where an informative sample had equal to or greater man 20 reads covering the CpG, and if 6 or more of the EAC samples demonstrated percent methylation at a level mat was at least 20 percentage points greater than the methylation level of the most methylated BE sample. CpGs meeting criteria for methylation in EAC versus and BE are defined as methylated in EAC vs BE. Such methylated CpGs were then aggregated into patches in instances in which methylated CpGs were within 200bp one another.
186 genomic patches defined as methylated in EACs versus BE in the discovery set were identified (see SEQ ID NOs: 8447-8818). The genomic coordinates of the genomic patches defined as methylated by the above criteria were also recorded. The genomic sequences of these patches on the respective genomic (+) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences). The bisulfite converted sequences of these corresponding patches(i.e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (- ) strand) were determined and recorded (see sequences of SEQ ID NOs: 8819-9004 and 9191-9376 for the bisulfite converted sequences of the respective (+) and (-) strands). C residues mat may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at mat base. Thus, these sequences represent the group of all combinations of all sequences in which 0, 1, or more man one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the corresponding patches (i.e. in which all Y bases in every bisulfite converted sequence are retained as a C), corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ ID NOs: 9005-9190 and 9377-9562 for the bisulfite converted sequences of the fully methylated form of the (+) and (-) strands respectively of the corresponding patches). The reverse complements of the bisulfite converted methylated (÷) stand and (-) stand sequences will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
Patches were expanded by 100 base pairs on either side so as to accommodate either the design of amplification pimers or to enc!ude additional presumptively methylated bases. The sequences of these expanded patches correspond to SEQ ID NOs: 9563-9934 and their genomic coordinates were also recorded. The genomic sequences of these expanded patches on the respective genomic (+) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences). The bisulfite converted sequences of these corresponding expanded patches (i.e. the bisulfite converted sequence of the (-(-) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 9935-10120 and 10307- 10492 for the bisulfite converted sequences of the respective ( f) and (-) strands), C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methy!ation at that base. Thus, these sequences represent the group of ail combinations of all sequences in which 0, 1, or more than one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (-(-) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the corresponding expanded patches (i.e. in which ail Y bases in every bisulfite converted sequence are retained as a C), corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ ID NOs: 10121-10306 and 10493-10678 for the bisulfite converted sequences of the fully methylated form of the (+-) and (-) strands respectively of the corresponding expanded patches). The reverse complements of the bisulfite converted methylated (+) stand and (-) stand sequences will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. CpG islands overlapping patches that may contain additional CpGs that are methylated coordinately with patches were also defined. The sequences of these CpG islands correspond to SEQ ID NOs: 10679-10972. The genomic coordinates of the CpG islands were also recorded. The genomic sequences of these expanded patches on the respective genomic (+) and (-) strands were determined and recorded. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences). The bisulfite converted sequences of these corresponding CpG islands (i.e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 10973-11119 and 11267-11413 for the bisulfite converted sequences of the respective (+) and (-) strands). C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base. Thus, these sequences represent the group of all combinations of all sequences in which 0, 1, or more than one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the corresponding CpG islands (i.e. in which all Y bases in every bisulfite converted sequence are retained as a C), corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ ID NOs: 11120-11266 and 11414-11266 for the bisulfite converted sequences of the fully methylated form of the (+) and (-) strands respectively of the corresponding CpG islands). The reverse complements of the bisulfite converted methylated (+) stand and (-) stand sequences will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
Regions of Interest (ROI) were defined that provided preferred regions for design of PCR amplicons that would encompass preferred patches. The genomic coordinates of the ROI were also recorded. The sequences of the (+) strands of the Regions of Interest correspond to SEQ ID NOs: 12563-12568, and the sequences of the (-) strands Regions of Interest correspond to SEQ ID NOs: 12581-12586. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences). The bisulfite converted sequences of these corresponding Regions of Interest (i.e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences ofSEQ ID NOs: 12569-12574 and 12587-12592 for the bisulfite converted sequences of the respective (+) and (-) strands). C residues that may be methylated or unmethylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at mat base. Thus, these sequences represent the group of all combinations of all sequences in which 0, 1, or more man one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the Regions of Interest (i.e. in which all Y bases in every bisulfite converted sequence are retained as a C), corresponding to the (+) strand the (-) strand were determined and recorded (see sequences ofSEQ ID NOs: 12575-12580 and 12593-12598 for the bisulfite converted sequences of me fully methylated form of the (+) and (-) strands respectively of the corresponding Regions of Interest). The reverse complements of the bisulfite converted methylated (+) stand and (-) stand sequences will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure.
Specific PCR Amplicons were defined within the Regions of Interest (ROl). The genomic coordinates of the Amplicons were recorded. The sequences of the (+) strands of the Amplicons correspond to SEQ ID NOs: 12599-12604 and 12647-12649, and the sequences of the (-) strands of the Amplicons correspond to SEQ ID NOs: 12617-12622 and 12656-12658. (Upper and lower case designations were used according to those of the UCSC browser, where lower case sequences are lower complexity DNA sequences). The bisulfite converted sequences of these Amplicons (i.e. the bisulfite converted sequence of the (+) strand and the bisulfite converted sequence of the (-) strand) were determined and recorded (see sequences of SEQ ID NOs: 12605-12610 and 12650-12652 for the bisulfite converted sequences of the (+) strands and see sequences of SEQ ID NOs: 12623-12628 and 12659-12661 and for the bisulfite converted sequences of the (-) strands). C residues that may be methylated or unmemylated, and hence may be bisulfite converted to T (if unmethylated) or remain as a C (if methylated), were designated with a Y (where Y denotes C or T), and where, after bisulfite conversion, actual maintenance of a Y designated base as a C was scored as methylation at that base. Thus, these sequences represent the group of all combinations of all sequences in which 0, 1, or more than one Y is converted to a T. The reverse complements of the bisulfite converted sequences of the (+) and (-) strands will be obvious to one of ordinary skill in the art and are also included by implication in this disclosure. The bisulfite converted sequences of the fully methylated form of the Amplicons (i.e. in which all Y bases in every bisulfite converted sequence are retained as aC), corresponding to the (+) strand the (-) strand were determined and recorded (see sequences of SEQ IDNOs: 12611-12616 and 12653-12655 for the bisulfite converted sequences of the fully methylated form of the (+) strands and see sequences of SEQ ID NOs: 12629-12634 and 12662-12664 for the bisulfite converted sequences of the fully methylated form of the (-) strands). The reverse complements of the bisulfite converted methylated (+) stand and (-) stand sequences will be obvious to one of ordinary skill in the art and are also included by implication in mis disclosure. Sequences ofPCR primers used in amplification of the Amplicons are provided as SEQ ID NOs: 12635-12646 and 12665-12670.
Confirmatory analysis of candidate loci was then done using bisulfite sequencing of candidate loci that were amplified using bisulfite specific but methylation independent amplification primers. This employed a new sample set of biopsies from: 23 EACs, 8 HGD, 15 non-dysplastic BE from individuals without known higher grade lesions. In addition, biopsies were obtained from 5 cases of BE adjacent to HGD, and from 11 cases of BE adjacent to an EAC. These are not included in analyses of non-dysplastic BE. In addition biopsies were obtained from 33 normal squamous mucosa samples.
Table 2 describes the performance in the confirmatory sample set using bisulfite sequencing analysis of amplicons from select loci defined as methylated in EACs versus BE and having preferred marker characteristics. In Table 2, Columns B - J disclose the performance of the amplicons in the confirmatory data set. hi this data set, methylation was calculated as the average level of methylation of all CpGs in between the primers for amplifying the amplicon. For each read across each ampHcon the number of CpGs that were methylated was counted and the read was classified as methylated or unmethyled using cutoffs for a required number of methylated CpGs on the amplicon. Table 2, row 3 lists the number of CpGs between the amplification primers for each of the amplicons. Table 2, row 4 lists the number of CpGs that need to be methylated on an individual read to count that read as methylated (e.g. for Up3 there are 36 CpG residues between the primers, and 25+ (meaning >=25) CpGs must be methylated on a read to score it as methylated). Table 2, rows 6 and 7 record the sensitivity for detecting EACs (row 6) and HGD (row 7) using criteria in which a sample was detected if it demonstrated methylation in greater man 10% (0.1) of all DNA reads. Table 2, rows 8 and 9 record the specificity of each amplicon for not detecting non-dysplastic BE (row 8) and for not detecting normal squamous mucosa (row 9) again using criteria in which a sample was detected if it demonstrated methylation in greater than 10% (0.1) of all DNA reads. Table 2, rows 11 and 12 record the specificity of each amplicon for not detecting non-dysplastic BE (row 11) and for not detecting normal squamous mucosa (row 12) using criteria in which a sample was detected if it demonstrated methylation in greater than 1% (0.01) of all DNA reads. Ampllcons (and patches) need not be used individually, but can be combined into panels for detection of esophageal neoplasia. Performance statistics of selected panels of amplicons are provided in Table 2 columns K through V mat provides the sensitivity and specificity of the panels (when the combination is positive if any member of the combination is positive).
Figure imgf000106_0001
Figure imgf000107_0001
-
In addition, RRBS discovery data was analyzed to identify CpG residues that demonstrated: i) at least 3 informative BE, which in every informative BE demonstrated at least 90% methylation; and where ii) no more than 5% of informative normal squamous samples demonstrated methylation level below 90%,;and that iii) demonstrated at least 6 informative EAC, where in these informative EACs the level of methylation was at least 20 percentage points lower than the methylation level of the least methylated BE. CpGs meeting these criteria are defined as unmethylated in EAC versus BE. Such unmethylated CpGs were then aggregated into patches by grouping together unmethylated CpGs that were within 200bp of one another. Unmethylated in EAC patches may consist of 1 CpG up to any number of CpGs that meet the above criteria. Example 3:Identification of esophageal cancer informative locito detect progression of esophageal neoplasia
Biopsy samples ( that overlapped with the confirmatory biopsy sample set) were further analyzed in tests of panels of markers for detecting the progression of Barrett's esophagus to Barrett's esophagus high grade dysplasia (HGD) or to esophageal adenocarcinoma (EAC). For each panel of markers, Figure 1 shows the sensitivity (percentage of samples detected), the specificity (percentage of samples not detected), the total number of samples studied, and the total number of positive samples. Three panels of markers were selected for study. The first marker pane! consisted of detecting at least one of the following four methylated markers: Upl5-1, Up35-1, Up27, and UplO (using bisulfite sequencing analysis of the corresponding amplicons and using the criteria for detection specified in table 2A). The second panel consisted of testing for somatic non- synonymous mutations in TP53 in assays in which TP53 was amplified from genomic DNA using a set of PGR amplicons that spanned the TP53 coding region and in which Next Generation DNA Sequencing was then used to compare TPS 3 sequences from esophageal lesions versus matched normal esophagus tissue. Samples were classified as detected if a TP53 mutant allele frequency of greater than or equal to 10% was identified. The third panel was a combination of detection of methylation in any of Upl 5-1, Up35-1 , Up27, and UplO or detection of mutation in TP53.
Table 3 shows the individual performance of biomarkers in detecting the different sample types of the 1st validation biopsies set using different cut-off criteria for methylation than the analysis of table 2. Shown is the performance for detection of different sample types of 5 different methylated DNA markers tested by bisulfite sequencing analysis of the corresponding amplicons (Up3, Up 10, Up27, Up35-1, Up35-2). Table 3 specifies the number of CpGs required to be methylated on a DNA sequence read to classify that read as methylated for this analysis. Results are presented when samples are considered methylated if greater than or equal to 1% of all DNA sequence reads are classified as methylated, or if greater than or equal to 10% of all DNA sequence reads are classified as methylated. Also shown is the performance for detecting samples of testing for non-synonymous somatic mutation in assays in which TP53 was amplified from genomic DNA using a set of PGR amplicons that spanned the TP53 coding region and in which Next Generation DNA Sequencing was then used to compare TP53 sequences from esophageal lesions versus matched normal esophagus tissue. Shown are rates of sample detection (expressed as sensitivity or specificity) when samples are classified as TP53 mutant if greater than or equal to 3% of TP53 reads are scored as mutant, or if greater than or equal to 10% of TP53 reads are scored as mutant.
Figure imgf000109_0001
Figure imgf000110_0001
Table 4 shows the performance of selected combinations of the methylated DNA markers (Up3, Up 10, Up27, Up35-1 , Up35-2) for detection of different sample types in the experiment presented in Table 3, Samples are scored as methylated if any member of the marker combination panel scores the sample as methylated. Results are presented when the individual markers are considered methylated if greater than or equal to 1% of all DNA sequence reads are classified as methylaied, or if greater than or equal to 10% of all DNA sequence reads are classified as methylated.
TABLE 4A
Figure imgf000111_0001
Figure imgf000112_0001
Samples summarized in Table 3 and Table 4 were additionally tested for non- synonymous somatic mutations in TP53. Table 5 shows performance of selected combinations of methylated DNA markers (Up3, Up 10, Up27, Up35-1 , Up35-2) plus testing for mutations in TP53 (p53) for detection of different sample types. Samples are scored as detected if any member of the marker combination panel scores the sample as methylated or if analysis for TP53 mutations scores the sample as TPS 3 mutant. Shown is the performance of the marker panel in which samples are scored as detected if any methylation marker is detected as methylated at greater than or equal to 1% of DNA reads, or if TPS 3 is detected as mutant at greater than or equal to 10% of the DNA sequence reads. Also shown is the performance of the marker panel in which samples are scored as detected if any methylation marker is detected as methylated at greater than or equal to 10% of DNA reads, or if TP53 is detected as mutant at greater than or equal to 10% of the DNA sequence reads.
Figure imgf000114_0001
Figure imgf000115_0001
DNA was also extracted frora esophageal samples that were also obtained by cytology brushings of the esophagus. The sample set included brushings from 49 esophageal adenocarcinomas (EAC); 14 carcinomas of the gastroesophageal junction (JCA); 8 Barrett's esophagus with low grade dysplasia (LGD); 9 Barrett's esophagus with high-grade dysplasia (HGD); 33 cases of Barrett's esophagus without dysplasia from cases without HGD or EAC, otherwise termed non-dysplastic BE, that included 1.3 cases of short segment Barrett's esophagus (SSBE). Also included were brushings of the gastroesophageal junction (normal GEJ) frora 62 individuals without Barrett's esophagus, without HGD, without EAC This included individuals with gastroesophageal reflux disease, with eosinophilic esophagiiis. or without any disease. Also included were 176 brushings of normal squamous esophagus frora each of the above individuals. These DNA. samples were analyzed for methylation by bisulfite sequencing of selected ampiicons and were also analyzed for non-synonymous somatic mutations in TP53.
Table 6 shows the individual performance of biomarkers in detecting the different sample types of the validation brushings set. Shown is the performance for detection of different sample types of 5 different methylated DNA markers analyzed by bisulfite sequencing of selected ampiicons (Up3, Up 10, Up27, Up35-1, Up35-2). The table specifies the number of CpGs required to be methylated on a DNA sequence read to classify that read as methylated. Results are presented when samples are considered methylated if greater than or equal to 1% of all DNA sequence reads are classified as methylated, or if greater than or equal to 10% of all DNA sequence reads are classified as methylated. Also shown is the performance for detecting samples of testing for somatic mutation in assays in which TP53 was amplified from genomic DNA using a set of PCR ampiicons that spanned the TP53 coding region and in which Next Generation DNA Sequencing was then used to compare TP53 sequences from esophageal lesions versus matched normal esophagus tissue. Shown are rates of sample detection (expressed as sensitivity or specificity) when samples are classified as TP53 mutant if greater than or equal to 3% of TPS 3 reads are scored as mutant, or if greater than or equal to 10% of TPS 3 reads are scored as mutant.
Figure imgf000117_0001
Figure imgf000118_0001
Table 7 shows the performance of selected combinations of the methylated DNA markers (Up35 Up 10, Up27, Up35-1, Up35-2) for detection of different, sample types in the esophageal brush ings samples presented in Table 6. Samples are scored as methylated if any member of the marker combination panel scores the sample as methylated. Results are presented when the individual markers are considered methylated if greater than or equal to 1% of all DNA sequence reads are classified as methylated, or if greater than or equal to 10% of all DNA sequence reads are classified as methylated.
Figure imgf000119_0001
TABLE ?B
Figure imgf000120_0001
Table 8 shows the performance of selected combinations of methylated DNA markers (Up3, Up1O, Up27, Up35~l , Up35-2) plus testing for mutations in TP53 (ρ53) for detection of different sample types in the esophageal brushings samples presented in Tables 6 and 7. Samples are scored as detected if any member of the marker combination pane! scores the sample as methylated or if analysis for TPS 3 mutations scores the sample as TP53 mutant. Shown is the performance of the marker panel in which samples are scored as detected if any methylation marker is detected as methylated at greater than or equal to 1% of DNA reads, or if TPS 3 is detected as mutant at greater than or equal to 3% or at greater than or equal to 10% of the DNA sequence reads. A!so shown is the performance of the marker pane! in which samples are scored as detected if any methylation marker is detected as methylated at greater thars or equal to 10% of DMA reads, or if TP53 is detected as mutant at greater than or equal to 3% or at greater than or equal to 10% of the DNA sequence reads. Marker combination with greater than 90% specificity in all BE without dysplasia are preferred yellow. Marker combinations that additionally show superior sensitivity for EAC are further preferred, Particularly preferred marker combinations are: Up35-2 methylation plus TP53 mutation; Up35-2 methylation plus Up3 methylation plus TPS 3 mutation; Up 10 methylation plus Up3 methylation plus TP53 mutation; Up35-2 methylation plus Up1O methylation plus TP53 mutation; Up1O methylation plus Up27 methylation plus TP53 mutation: Up35-2 methylation plus Up3 methylation plus Up10 methylation plus TP53 mutation.
Figure imgf000122_0001
Figure imgf000123_0001
Example 4: Analysis of esophageal cancer informative loci in formalin fixed paraffin
Additional studies were performed on DNAs extracted from formalin fixed paraffin embedded (EWE) tissue samples of the stomach and esophagus that capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma. Bisulfite converted DNAs from each sample were amplified with bisulfite specific methylation indifferent primers corresponding to selected amplicons and the amplicons were men analyzed by bisulfite sequencing to determine methylation status on (he parental DNA templates.
Table 9 summarizes the side by side comparison of 8 methylated DNA markers in
FFPE tissue samples of the stomach and esophagus that capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma. Intestinal metaplasia is abbreviated as IM. Table 9 denotes for each marker the number of methylated cytosine bases required to be detected on a DNA sequence read to classify that read as methylated. Samples are detected as methylated if greater than or equal to 1% of DNA sequence reads are classified as methylated.
Figure imgf000125_0001
Figure imgf000125_0002
Figure imgf000126_0001
Table 10 summarizes the side by side comparison of 8 methylated DNA markers in FFPE tissue samples of the stomach and esophagus that capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma, intestinal metaplasia is abbreviated as iM. Table 10 denotes for each marker the number of methylated cytosine bases required to be detected on a DNA sequence read to classify that read as methylated. Samples are detected as methylated if greater than or equal to 10% of DNA sequence reads are classified as methylated,
TABLE "I OA
Figure imgf000128_0001
Figure imgf000129_0001
Table 1 1 summarizes performance of different panels comprised of combinations of methylated DNA markers in FFPE tissue samples of the stomach and esophagus that capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma. Intestinal metaplasia is abbreviated as IM. Tables 9 and 10 denote for each marker the number of methylated cytosine bases required to be detected on a DMA sequence read to classify that read as methylated. Samples are detected as methylated if greater than or equal to 1 % of DNA sequence reads are classi fied as methylated for any member of the marker panel.
Figure imgf000131_0001
Figure imgf000132_0001
Table 12 summarizes performance of different panels comprised of combinations of methylated DNA markers in FFPE tissue samples of the stomach and esophagus thai capture different diagnostic categories other than Barrett's with high grade dysplasia and esophageal adenocarcinoma. Intestinal metaplasia is abbreviated as IM. Tables 9 and 10 denote for each marker the number of methylated cytosine bases required to be detected on a DNA sequence read to classify that read as meihylaled. Samples are detected as methylated if greater than or equal to 10% of DN A sequence reads are classified as methylated for any member of the marker panel.
Figure imgf000134_0001
Figure imgf000135_0001
Somatic mutations in TP53 were detected using the following method. TP53 exons 2-11 were amplified using a multiplexed series of primer pairs covering all coding sequences and splice junctions. The primers contained additional 5' end sequences that were then used for secondary amplification that introduced barcode sequences and Illumina 15 and 17 sequences into the final FCR products. FCR products were mixed, purified and analyzed on an Illumina MiSeq instrument. Data analysis was performed using CLCBio software (Qiagen) and VarianiSiudio software (Illumina).

Claims

We claim:
1. A method of detecting the presence of an esophageal neoplasia or metaplasia in the esophagus in a human subject, comprising:
a) obtaining a human sample; and
b) assaying said sample for the presence of methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, or complements or fragments thereof;
wherein methylation of said nucleotide sequence is indicative of an esophageal neoplasia or metaplasia in the esophagus.
2. A method of detecting an esophageal neoplasia or metaplasia in the esophagus, comprising:
a) obtaining a human sample; and
b) assaying said sample for the presence of DN A methylation by assay in a bisulfite converted DNA for retention of a cytosine base at any of (he Y positions present in one or more of the nucleotide sequences having at least 90% identical to the sequence of any one or more of SEQ ID NOs: 857-1284, 1713-2140, 3425-
3852, 4281-4708, 5927-6321, 6717-7111, 7559-7584, 7611-7636, 7715-7740, 7767-7792, 7867-7890, 7915-7938, 7991-8004, 8019-8032, 8075-8088, 8103- 8116, 8157-8169, 8183-8195, 8223-8236, 8265-8278, 8307-8320, 8349-8362, 8410-8414 or 8425-8429;
wherein methylation of said nucleotide sequence is indicative of metaplasia in the esophagus or neoplasia.
3. The method of claim 2, wherein the sample is assayed for the presence of methylation at any of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of SEQ ID NOs: 7991 - 8004, 8019-8032, 8075-8088, 8103-8116, 8157-8169, 8183-8195, 8223-8236, 8265-8278, 8307-8320, 8349-8362, 8410-8414 or 8425-8429.
4. A method of detecting an esophageal neoplasia, comprising:
a) obtaining a human sample; and
b) assaying said sample for the presence of methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence mat is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 11561-11662, 11867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658, or complements or fragments thereof.
5. A method of detecting an esophageal neoplasia, comprising:
a) obtaining a human sample; and
b) assaying said sample for the presence of DNA methylation by assay in a bisulfite converted DNA for retention of a cytosine base at any of the Y positions present in one or more of the nucleotide sequences having at least 90% identical to the sequence of any one or more of SEQ ID NOs: 8819-9004, 9191-9376, 9935- 10120, 10307-10492, 10973-11119, 11267-11413, 11663-11713, 11765-11815, 11969-12019, 12071-12121, 12267-12313, 12361-12407, 12467-12472, 12479- 12484, 12503-12508, 12515-12520, 12539-12544, 1255M2556, 12569-12574, 12587-12592, 12605-12610, 12623-12628, 12650-12652, or 12659-12661;
wherein methylation of said nucleotide sequence is indicative of metaplasia in the esophagus.
6. The method of claim 4 or 5, wherein the sample is assayed for the presence of DNA methylation by assay in a bisulfite converted DNA for retention of a cytosine base at any of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of SEQ ID NOs: J 1663-11713, 11765- 11815, 11969-12019, 12071-12121, 12267-12313, 12361-12407, 12467-12472, 12479- 12484, 12503-12508, 12515-12520, 12539-12544, 12551-12556, 12569-12574, 12587- 12592, 12605-12610, 12623-12628, 12650-12652, or 12659-12661.
7. The method of any one of claims 4-6, wherein said subject has been assayed for having a somatic mutation in TP53.
8. The method of any one of claims 1 to 3, wherein the esophageal neoplasia or metaplasia in the esophagus is selected from the group consisting of Barrett's esophagus (BE), and/or, Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia (HGD), and/or esophageal adenocarcinoma
(EAC).
9. The method of any one of claims 1 to 8, wherein the sample is obtained from a subject suspected of having or is known to have an esophageal neoplasia or metaplasia in the esophagus.
10. The method of any one of claims 1 to 9, wherein the assay is methylation-specific PCR.
11. The method of any one of claims 1-4, further comprising:
a) treating DNA from the sample with a compound that converts a non- methylated cytosine base in the DNA to a different base;
b) amplifying a region of the compound converted nucleotide sequence with a forward primer and a reverse primer; and
c) analyzing the methylation patterns of said nucleotide sequences.
12. The method of any one of claims 1-4, further comprising:
a) treating DNA from the sample with a compound that converts a non- methylated cytosine base in the DNA to a different base;
b) amplifying a region of the compound converted nucleotide sequence with a forward primer and a reverse primer; and
c) detecting the presence and/or amount of the amplified product.
13. The method of claim 11 or 12, wherein the compound used to treat DNA is a bisulfite compound.
14. The method of one any of claims 1 to 13, wherein the assay comprises using a methylation-sensitive restriction enzyme.
15. T15 .ethod of claim 14, wherein said methylation-sensitive restriction enzyme is selected from Hpall, Smal, Sacll, Eagl, BstUl, and Bssliil.
16. The method of any one of claims 1 to 15. wherein the sample is a bodily fluid selected from the group consisting of blood, saliva, spit, an esophageal washing, serum, plasma, a blood-derived fraction, stool, urine, onic effluent.
17. The method of any one of claims 1 to 15, wherein the sample is derived from a tissue.
18. The method of claim 17, wherein the sample is a biopsy.
19 The method of claim 17, wherein the sample is a brushing,
20. A method of monitoring over time an esophageal neoplasia or metaplasia in the esophagus comprising:
a) detecting the presence of methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1 - 856, 2569 -· 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, or complements or fragments thereof;, or complements or fragments thereof; from a sample from a subject for a first time; and
b) detecting the level of methylation of the nucleotide sequence in a sample from the same subject at a later time.
21. A method of monitoring over time an esophageal neoplasia or metaplasia in the esophagu s com prssang:
a) detecting the methylation status of DNA by assay in a bisulfite converted DNA for retention of a cvtosine base at one or more of the Y positions present Ira one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 857-1284, 1713-2140, 3425- 3852, 4281-4708, 5927-6321, 6717-7111, 7559-7584, 7611-7636, 7715-7740, 7767-7792, 7867-7890, 7915-7938, 7991-8004, 8019-8032, 8075-8088, 8103- 8116, 8157-8169, 8183-8195, 8223-8236, 8265-8278, 8307-8320, 8349-8362, 8410-8414 or 8425-8429 from a sample from a subject for a first time; and
b) detecting the methylation status of the nucleotide sequence in a sample from the same subject at a later time;
wherein absence of methylation in the nucleotide sequence taken at a later time and the presence of methylation in the nucleotide sequence taken at the first time is indicative of neoplasia or metaplasia regression; and
wherein presence of methylation in the nucleotide sequence taken at a later time and the absence of methylation in the nucleotide sequence taken at the first time is indicative of neoplasia or metaplasia progression.
22. A method of monitoring over time an esophageal neoplasia comprising:
a) detecting the presence of methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447- 8818, 9563-9934, 10679-10972, 1 1561-11662, 11867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599- 12604, 12617-12622, 12647-12649 or 12656-12658, or complements or fragments thereof; from a sample from a subject for a first time; and
b) detecting the level of methylation of the nucleotide sequence in a sample from the same subject at a later time.
23. A method of monitoring over time an esophageal neoplasia comprising:
a) detecting the methylation status of a DNA molecule by assay in a bisulfite converted DNA for retention of a cytosine base of one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 8819-9004, 9191- 9376, 9935-10120, 10307-10492, 10973-1 1 1 19, 1 1267-1 1413, 1 1663-1 1713, 1 1765-11815, 11969-12019, 12071-12121, 12267-12313, 12361 -12407, 12467- 12472, 12479-12484, 12503-12508, 12515-12520, 12539-12544, 12551-12556, 12569-12574, 12587-12592, 12605-12610, 12623-12628, 12650-12652, or 12659- 12661 from a sample from a subject for a first time; and
b) detecting the methylation status of the nucleotide sequence in a sample from the same subject at a later time;
wherein absence of methylation in the nucleotide sequence taken at a later time and the presence of methylation in the nucleotide sequence taken at the first time is indicative of neoplasia regression; and
wherein presence of methylation in the nucleotide sequence taken at a later time and the absence of methylation in the nucleotide sequence taken at the first time is indicative of neoplasia progression.
24. The method of any one of claims 20-23, wherein the sample is a bodily fluid
selected from the group consisting of blood, saliva, spit or an esophageal washing.
25. The method of any one of claims 20-23, wherein the sample is derived from tissue.
26. A method of treating a subject having a metaplasia in the esophagus or neoplasia, comprising the step of treating the subject with chemotherapy, radiation therapy and/or with the resection of the neoplasia or metaplasia; and/or with ablation of the neoplasia or metaplasia; wherein said subject has been determined to have methylation in a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663
- 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251
- 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, or complements or fragments thereof.
A method of treating a subject having a metaplasia in the esophagus or neoplasia,
27.
comprising the step of treating the subject with chemotherapy, radiation therapy and/or with the resection of the neoplasia or metaplasia; and/or with ablation of the neoplasia or metplasia; wherein said subject has been determined to have DNA methylation by assay in a bisulfite converted DNA for retention of a cytosine base of one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of: SEQ ID NOs: 857-1284, 1713-2140, 3425-3852, 42814708, 5927-6321, 6717- 7111 , 7559-7584, 761 1-7636, 7715-7740, 7767-7792, 7867-7890, 7915-7938, 7993-8004, 8019-8032, 8075-8088, 8103-81 16, 8157-8169, 8183-8195, 8223- 8236, 8265-8278, 8307-8320, 8349-8362, 8410-8414 or 8425-8429.
A method of treating a subject having an esophageal neoplasia, comprising the step
28.
of treating the subject with chemotherapy, radiation therapy, ablation, and/or with neoplasia resection; wherein said subject has been determined to have methylation methylation within a nucleotide sequence spanning one or more of any of the chromosomal loci having a sequence that is at least 90% identical to the sequence of any one or more of: SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 11561-11662, 1 1867-1 1968, 12173-12266, 12455-12466, 12491 -12502, 12527- 12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-32649 or 12656-12658, or complements or fragments thereof.
29. A method of treating a subject having an esophageal neoplasia, comprising the step of treating the subject with chemotherapy, radiation therapy, ablation, and/or with neoplasia resection; wherein said subject has been determined to have methylation of a DNA molecule by assay in a bisulfite converted UNA for retention of a cytosine base of one or more of the Y positions present in one or more of the nucleotide sequences having at least 90% identity to the sequence of any one or more of; SEQ 3D NOs: 8839-9004, 9191-9376, 9935-10120, 10307-10492, 10973- 1 1 119, 1 1267-1 1413, 1 1663-1 1713, 1 1765-1 1815, 1 1969-12019, 12071 -12121 , 12267-12313, 12361-12407, 12467-12472, 32479-12484, 12503-12508, 12515- 12520, 32539-12544, 12551-32556, 12569-12574, 12587-12592, 12605-12610, 12623-12628, 12650-12652, or 12659-12661.
30.
The method of any one of claims 27-29, wherein said subject has been assayed for having a somatic mutation in TP53.
31.
A bisulfite converted sequence having at least 90% identity to any one or more of SEQ ID NOs: 857 -2568, 3425-5136, 5927-7506, 7559 - 7662, 7715 - 7818, 7867 - 7962, 7991 - 8046, 8075 - 8330, 8157 - 8208, 8223 - 8250, 8265 - 8292, 8307 - 8334, 8349 - 8376, 8410 - 8419, 8425 - 8434, and/or fragments thereof and/or the reverse complements thereof including all unique fragments of these sequences and their reverse complements,
A panel of bisulfite converted sequences selected from the sequences of claim 31 ,
32.
The panel of claim 32, wherein the panel corresponds to any combination of the
33.
sequences having any of SEQ ID NOs: 8307-8334, 8349-8376, 8410-8419 or 8425-8434,
An oligonucleotide primer that hybridizes to any of the sequences of claim 31 .
34.
35. The primer of claim 34, wherein said primer is any of the primers having at least 90% identity to any one or more of SEQ ID NOs: 8377-8404 or 8435-8444, or fragments or complements thereof.
36. The primers of claim 34 or 35, wherein said primers are combined as forward and reverse primers for PGR amplification of any of the. bisulfite converted sequences of claim 31.
37. The primer pairs of claim 36, wherein one of the primers in said printer pairs has a nucleotide sequence that is at least 90% identity to any one or more of SEQ ID NOs: 8377-8404 or 8435-8444, or fragments or complements thereof.
38.
A panel of primer pairs selected from the primer pairs of claim 36 or 37.
39.
The panel of claim 38, wherein the panel correspond to a combination of primer pairs for amplifying any of the combinations of SEQ ID NOs: 8307-8334, 8349- 8376, 8410-8419 or 8425-8434.
40.
A bisulfite converted sequence having at least 90% identity to any one or more of SEQ ID NOs: 8819 - 9562, 9935 - 10678, 10973 - 1 1560; 1 3663 - 1 1866, 1 1969 - 12172, 12267 - 12454; 12467 - 12490, 12503 - 12526, 12539 - 12562, 12569 - 12580, 12587 - 12598, 12605 - 12616, 12623 - 12634, 12650 - 32655, and 12659 - 12664, and the reverse complements thereof, including all unique fragments of these sequences and their reverse complements.
A panel of bisulfite converted sequences selected from the sequences of claim 40,
41.
The pane! of claim 41 , wherein the pane! corresponds to any combination of any of
42.
the sequences of SEQ ID NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664.
An oligonucleotide primer that hybridizes to any of the sequences of claim 40.
43.
44. The primers of claim 43, wherein said primers are those that are provided in having at least 90% identity to any one or more of SEQ ID NOs: 12635-12646, and 12665-12670, or fragments or complements thereof.
45. The primers of claim 43, wherein said primers are combined as forward and
reverse primers for PGR amplification of any of the bisulfite converted sequences of claim 40.
46. The primer pairs of claim 45, wherein one of the primers in said primer pairs has a nucleotide sequence that is at least 90% identity to any one or more of SEQ ID NOs: 12635- 12646, and 12665-12670, or fragments or complements thereof.
47.
A pane! of pri mer pairs selected from the primer pairs of claim 45 or the primer pairs of claim 46.
48.
The panel of claim 47, wherein the panel corresponds to the combination of primer pairs for amplifying any combination of any of the sequences of SEQ ID NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664.
49.
A method of selecting an individual to undergo a diagnostic procedure to determine the presence of Barrett's esophagus, of Barrett's esophagus with Sow grade dysplasia, of Barrett's esophagus with high grade dysplasia, or of esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining the presence in DNA from that sample of DNA raethyiatiors present in any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926: 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, or 8420 - 8424, and/or fragments thereof, and/or the reverse complements thereof.
The method of claim 49, further comprising determining the presence of a somatic
50.
mutation in TPS 3,
51. The method of claim 49 or 50, wherein DNA methylation is detected by cutting one of the DNA sequences with a methylation sensitive restriction enzyme.
52. The method of claim 49 or 50, wherein DNA methylation is detected by bisulfite converting DNA from the sample and detecting the presence of any of the bisulfite con verted DNA sequences of claim 31.
53. The method of claim 52, wherein the bisulfite converted sequences are detected using any of: DNA sequencing, next generation sequencing, methylation specific PGR, methylation specific FCR combined with a fluorogenic hybridization probe, and real time methylation specific PCR.
54.
The method of claim 52, wherein the bisul fite converted sequences are detected using PCR amplification employing a PCR primer or primer pair comprising the nucleotide sequence of any of SEQ ID NOs: 8377-8404 or 8435-8444 .
55.
The method of claim 54, wherein the method further comprises the step of determining the nucleotide sequence of the bisulfite converted sequences. The method of any one of claims 49, 54 or 55, wherein the percent of the target
56. sequence that is methylated in any of the individual target sequences is greater than i%, or greater than 5%, or greater than 10%.
The method of any one of claims 49, 54 or 55, wherein the biological sample is a
57.
tissue sample, ineSuding a sample of the esophagus.
The method of claim 57, wherein the tissue sample is a biopsy or a brushing,
58.
including a biopsy or a brushing of the esophagus.
The method of claims 49-54, wherein the biological sample is a body fluid,
59.
60. The method of claim 59, wherein the body fluid is blood, saliva, spit or an
esophageal washing.
61. A method of selecting an individual to undergo a diagnostic procedure to
determine presence of Barrett's esophagus with low grade dysplasia, of Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma; or to undergo a treatment for for Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia or for esophageal adenocarcinoma; or to undergo enhanced surveillance for the development of Barrett's esophagus with low grade dysplasia, of Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma, by obtaining a biological sample from an individual, and determining the presence in DNA from that sample of methvlation present in any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818, 9563-9934, 10679-10972, 1 1561-1 1662, 1 1867-1 1968, 12173- 12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581 -12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658.
62.
A method of treating a subject having Barrett's esophagus with low grade dysplasia, Barrett's esophagus with high grade dysplasia or esophageal adenocarcinoma, comprising the step of treating the subject with chemotherapy, radiation therapy, and/or with resection of an esophageal lesion or with ablation of an esophageal lesion, wherein the subject has been determined to have methylation at any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818, 9563-9934, 10679-10826, 11561-11662, 11867-11968, 12173- 12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658.
63. A method of selecting an individual to undergo enhanced surveillance for the development of Barrett's esophagus with high grade dysplasia or of esophageal adenocarcinoma, wherein the subject has been determined to have methylation at any of the sequences having at least 90% identity to any one or more of SEQ ID NOs: 8447-8818, 9563-9934, 10679-10826, 11561-11662, 11867-11968, 12173- 12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658.
64. The method of any one of claims 61-63, further comprising determining the
presence of a somatic mutation in TP53.
65. The method of any one of claims 61-64, wherein DNA methylation is detected by cutting one of the DNA sequences with a methylation sensitive restriction enzyme.
66. The method of any one of claims 61-64, wherein DNA methylation is detected by bisulfite converting DNA from the sample and detecting the presence of any of the bisulfite converted DNA sequences of claims 40-42, or detecting the presence of uncoverted cytosine residues at any of the Y bases in any of the bisulfite converted DNA sequences of claims 40-42.
67. The method of claim 66, wherein the bisulfite converted sequences or the presence of unconverted cytosine residues at any of the Y bases of claim 66 are detected using any of: DNA sequencing, next generation sequencing, methylation specific PCR, methylation specific PCR combined with a fluorogenic hybridization probe, and real time methylation specific PCR.
68. The method of claim 66, wherein the bisulfite converted sequences or the presence of unconverted cytosine residues in the bisulfite converted sequences are detected using PCR amplification employing any of the PGR primers or primer pairs of claims43-48.
69. The method of any one of claims 61-68, wherein the percent of the target sequence that is methylated in any of the individual target sequences is greater than 1%, or greater than 5%, or greater than 10%.
70. The method of any one of claims 61-69, wherein the biological sample is a tissue sample, including a sample of the esophagus.
71. The method of claims 70, wherein the tissue sample is a biopsy or a brushing, including a biopsy or brushing of the esophagus.
72. The method of any one of claims 61-69, wherein the biological sample is a body fluid.
73. The method of claim 72, wherein the body fluid is blood, saliva, spit or an
esophageal washing.
74. : The method of any one of claims 61-73, wherein the individual is known to have
Barrett's esophagus.
75.. The method of any one of claims 61-73, wherein the individual is known to have Barrett's esophagus with low grade dysplasia.
76, A method of determining the response of an individual with esophageal cancer to therapy by detection in a body fluid of methylation in any of the DNA sequences having at least 90% identity to any one or more ofSEQ ID NOs: 1 - 856, 2569 -
3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 11561-11662, 11867- 11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658 and/or fragments thereof, and/or the reverse complements thereof; wherein increasing levels of methylation over time are indicative of disease progression and a need for a change in therapy, and wherein absence of increase in levels of methylation over time or decrease in levels of methylation over time are indicative that a change in therapy is not required.
77. The method of claim 76, further comprising detecting the presence of a somatic mutation in TP53.
78. The method of claim 76 or 77, wherein the DNA methylation is detected by
bisulfite converting DNA from a body fluid and detecting the presence of any of the bisulfite converted DNA sequences of any one of claims 31-33 or 40-42.
79. A method of monitoring over time an esophageal neoplasia comprising:
a) detecting in a sample from a subject for a first time (i) the methylation status of one or more of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 11561-11662, 11867-11968, 12173-12266, 12455- 12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658 from that sample; and (ii) the somatic mutation status of TP53 from that sample; and
b) detecting in a sample from the same subject at a later time (i) the methylation status of one or more of the DNA sequences having at least 90% identity to any one or more of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 11561-11662, 11867-11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599- 12604, 12617-12622, 12647-12649 or 12656-12658; and (ii) the somatic mutation status of TP53 from the same subject at a later time.
80. The method of claim 79, wherein neoplasia regression is indicated by:
(i) the sample taken at the first time having methylation in any of Upl5-1, Up35-1, Up35-2, Up3, Up27, or UplO, or having a somatic mutation in TP53; and
(ii) the sample at a later time lacking methylation in Upl5-1, Up35-1, Up35-2, Up3, Up27, UplO, and lacking a somatic mutation in TP53.
81 The method of claim 79, wherein neoplasia progression is indicated by:
(i) the sample taken at the first time lacking methylation in Upl 5-1, Up35-1, Up35-2, Up3, Up27, and UplO, and lacking a somatic mutation in TP53; and
(ii) the sample taken at a later time having methylation in any of Upl 5-1 , Up35-1 , Up35-2, Up3, Up27, UplO, or having a somatic mutation in TP53.
82. The method of any one of claims 79-81, wherein the biological sample is a tissue sample, including a sample of the esophagus.
83 The method of claim 82, wherein the tissue sample is a biopsy or a brushing, including a biopsy or a brushing of the esophagus.
84. The method of any one of claims 79-81, wherein the biological sample is a body fluid.
85, The method of claim 84, wherein the body fluid is blood, saliva, spit or an
esophageal washing.
86. A method of detecting die presence of an esophageal neoplasia in a human subject, comprising:
a) obtaining a human sample; and
b) detecting the methylation status of one or more of the DNA sequences having at least 90% identity to any one or more ofSEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424, 8447-8818, 9563-9934, 10679-10826, 11561-11662, 11867- 11968, 12173-12266, 12455-12466, 12491-12502, 12527-12538, 12563-12568, 12581-12586, 12599-12604, 12617-12622, 12647-12649 or 12656-12658 from that sample.
87. The method of claim 86, wherein the method farther comprises the step of detecting the somatic mutation status of TP53 from that sample.
88. The method of claim 87, wherein the TP53 somatic mutation is detected at a later time than the detection of the methylation status of any of the DNA sequences of claim 86,
89. The method of claim 87, wherein:
a) the presence of methylation at any of SEQ ID NOs: 1 - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424; and b) the absence of methylation at any one of the combination of Upl 5-1, Up3S-l, Up35-2, Up3, Up27, and UplO; and
c) the absence of a somatic mutation in TP53
is indicative of non-dysplastic Barret's esophagus.
90. The method of claim 87, wherein:
a) the presence of methylation at any of SEQ ID NOs: I - 856, 2569 - 3424, 5137 - 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 - 8424; and
b) the absence of methylation in all members of an assay panel selected from any or all of Upl5-1, Up35-1, Up35-2, Up3, Up27, and UplO; and
c) the absence of a somatic mutation in TP53
is indicative of non-dysplastic Barret's esophagus.
91. The method of claim 90, wherein* a) the presence of methylaiion at any of SEQ ID NGs: 1 - 856, 2569 - 3424, 5137
- 5926; 7507 - 7558, 7663 - 7714, 7819 - 7866, 7963 - 7990, 8047 - 8074, 8131 - 8156, 8209 - 8222, 8251 - 8264, 8293 - 8306, 8335 - 8348, 8405 - 8409, 8420 ~ 8424; and
b) the absence of methylaiion at Up-3 and Up35~2; and
c) the absence of a somatic mutation in TP53
is indicative of non-dyspiastic Barret's esophagus.
92. The method of claim 87, wherein the presence of raethyjation at any one of Up15- 1, Up35-1, Up35-2, Up3, Up27, and Up1O; or the presence of a somatic mutation in TP53 is indicative of esophageal adenocarcinoma, of Barrett's esophagus with low grade dysplasia, or of Barrett's esophagus with high grade dysplasia.
93. The method of claim 86, wherein the presence of methylation at any one of Up15- 1, Up35~l, Up35-2, Up3, Up27, or Up 10, is indicative of esophageal
adenocarcinoma, of Barrett's esophagus with low grade dysplasia, or of Barrett's esophagus with high grade dysplasia.
94. The method of any of claims 86-93, wherein the biological sample is a tissue sample, including a sample of the esophagus.
95. The method of claim 94, wherein the tissue sample is a biopsy or a brushing, including a biopsy or a brushing of the esophagus.
96. The method of any of claims 86-94, wherein the biological sample is a body fluid.
97. The method of claim 96, wherein the body fluid is blood, saliva, spit or an
esophageal washing.
98. A bisu Ifite-converted nucleotide sequence comprising the bisuifite-converted nucleotide sequence of any one of the following: Up3, Up1O, Upl5-1, Up] 5-2, Up20-1, Up20-2, Up20-2, Up27, Up35-1, Up35-2, SqBE2, SqBE5, SqBE7, SqBE9, SqBElO, SqBEl 1-1, SqBEl 1-2, SqBEB, SqBE14-2, SqBE15, SqBE16-l, SqBE16-2, SqBEl 7-1, SqBEl 8, SqBE22-l, SqBE22-2 or SqBE23.
99. Hie bisulfite-converted nucleotide sequence of claim 98, wherein the sequence comprises the bisulfite-converted nucleotide sequence of any one of the following: Up3, UplO, Upl5-1, UplS-2, Up20-1, Up20-2, Up20-2, Up27, Up35-1, or Up35-2ft.
100. The bisulfite-converted nucleotide sequence of claim 98, wherein the sequence comprises the bisulfite-converted nucleotide sequence of any one of the following: SqBE2, SqBE5, SqBE7, SqBE9, SqBElO, SqBEl 1-1, SqBEl 1-2, SqBEB,
SqBE14-2, SqBEl 5, SqBE16-l, SqBEl 6-2, SqBE17-l, SqBEl 8, SqBE22-l, SqBE22-2 or SqBE23.
101. A bisulfite-converted nucleotide sequence comprising a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8307-8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8410, 8411, 8412, 8414, 8415, 8416, 8417, 8419, 8425, 8426, 8427, 8429, 8430, 8431, 8432, 8434, 12605-12616, 12623-12634, 12650-12655, or 12659-12664.
102. The bisulfite-converted nucleotide sequence of claim 101, wherein the sequence comprises a sequence having at least 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID NOs: 8307-8313, 8315-8327, 8329-8334, 8349-8355, 8357-8369, 8371-8376, 8410, 8411, 8412, 8414, 8415, 8416, 8417, 8419, 8425, 8426, 8427, 8429, 8430,
8431, or 8432, 8434.
103. The bisulfite-converted nucleotide sequence of claim 101, wherein the sequence comprises a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: SEQ ID
NOs: 12605-12616, 12623-12634, 12650-12655, or 12659-12664.
104. The bisulfite-converted nucleotide of claim 103, wherein the sequence comprises a sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to any of the following sequences: 12652, 12655, 12661, and 12664.
PCT/US2015/068131 2014-12-31 2015-12-30 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus WO2016109712A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US15/540,956 US20190136325A1 (en) 2014-12-31 2015-12-30 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
CA2972782A CA2972782A1 (en) 2014-12-31 2015-12-30 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
EP15876289.8A EP3240912A4 (en) 2014-12-31 2015-12-30 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
AU2015374019A AU2015374019B2 (en) 2014-12-31 2015-12-30 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
AU2022201153A AU2022201153A1 (en) 2014-12-31 2022-02-21 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
US18/381,926 US20240068045A1 (en) 2014-12-31 2023-10-19 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462099021P 2014-12-31 2014-12-31
US62/099,021 2014-12-31

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/540,956 A-371-Of-International US20190136325A1 (en) 2014-12-31 2015-12-30 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
US18/381,926 Continuation US20240068045A1 (en) 2014-12-31 2023-10-19 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus

Publications (1)

Publication Number Publication Date
WO2016109712A1 true WO2016109712A1 (en) 2016-07-07

Family

ID=56285051

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2015/068131 WO2016109712A1 (en) 2014-12-31 2015-12-30 Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
PCT/US2015/068252 WO2016109782A2 (en) 2014-12-31 2015-12-31 Methods and compositions for detecting colorectal neoplasias

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2015/068252 WO2016109782A2 (en) 2014-12-31 2015-12-31 Methods and compositions for detecting colorectal neoplasias

Country Status (5)

Country Link
US (3) US20190136325A1 (en)
EP (1) EP3240912A4 (en)
AU (2) AU2015374019B2 (en)
CA (1) CA2972782A1 (en)
WO (2) WO2016109712A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018009535A1 (en) * 2016-07-06 2018-01-11 Case Western Reserve University Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
EP3336197A1 (en) * 2016-12-16 2018-06-20 GATC Biotech AG Epigenetic markers and related methods and means for the detection and management of ovarian cancer
EP3382033A1 (en) * 2017-03-30 2018-10-03 Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen Method for determining blood counts based on dna methylation
IT201700072650A1 (en) * 2017-06-28 2018-12-28 Univ Degli Studi Cagliari METHOD FOR READING AND / OR FOR THE PROGNOSIS OF COLON RECTAL NEOPLASES
US10400286B2 (en) 2003-08-14 2019-09-03 Case Western Reserve University Methods and compositions for detecting gastrointestinal and other cancers
EP3715474A4 (en) * 2017-11-22 2022-02-09 Lepidyne Co., Ltd. Method for assessing prognosis or risk stratification of liver cancer by using cpg methylation variation in gene
US20220349012A1 (en) * 2017-12-29 2022-11-03 City Of Hope Dna methylation diagnostic test for breast cancer
TWI815043B (en) * 2020-04-30 2023-09-11 大陸商廣州康立明生物科技股份有限公司 Tumor detection reagent and kit

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10428388B2 (en) * 2009-11-05 2019-10-01 Genomictree, Inc. Method for detecting the methylation of colorectal-cancer-specific methylation marker genes for colorectal cancer diagnosis
JP6871926B2 (en) 2015-08-31 2021-05-19 マヨ ファウンデーション フォア メディカル エデュケーション アンド リサーチMayo Foundation for Medical Education and Research Detecting gastric neoplasms
WO2018211404A1 (en) * 2017-05-17 2018-11-22 King Abdullah University Of Science And Technology Composite epigenetic biomarkers for accurate screening, diagnosis and prognosis of colorectal cancer
AU2018374176A1 (en) 2017-11-30 2020-06-11 Exact Sciences Corporation Detecting breast cancer
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
US11898199B2 (en) 2019-11-11 2024-02-13 Universal Diagnostics, S.A. Detection of colorectal cancer and/or advanced adenomas
US11530453B2 (en) 2020-06-30 2022-12-20 Universal Diagnostics, S.L. Systems and methods for detection of multiple cancer types
WO2022015860A1 (en) * 2020-07-15 2022-01-20 Rutgers, The State University Of New Jersey Biomarkers for identifying patients at high risk of progressing from barrett's esophagus to esophageal adenocarcinoma

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070161031A1 (en) * 2005-12-16 2007-07-12 The Board Of Trustees Of The Leland Stanford Junior University Functional arrays for high throughput characterization of gene expression regulatory elements
US20100151468A1 (en) * 2007-04-11 2010-06-17 Manel Esteller Epigenetic biomarkers for early detection, therapeutic effectiveness, and relapse monitoring of cancer
US20140206565A1 (en) * 2007-08-01 2014-07-24 University Of Maryland, Baltimore Esophageal Cancer Markers

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2004516821A (en) * 2000-09-01 2004-06-10 エピゲノミクス アーゲー Method for quantifying the degree of methylation of specific cytosine in sequence context 5-CpG-3 on genomic DNA
US20040063108A1 (en) * 2001-12-13 2004-04-01 Eos Biotechnology, Inc. Methods of diagnosis of colorectal cancer, compositions and methods of screening for modulators of colorectal cancer
WO2004041076A2 (en) * 2002-11-04 2004-05-21 Protein Design Labs, Inc. Methods of detecting colorectal cancer
US8415100B2 (en) * 2003-08-14 2013-04-09 Case Western Reserve University Methods and compositions for detecting gastrointestinal and other cancers
JP2008283947A (en) * 2007-04-19 2008-11-27 Tokyo Medical & Dental Univ Method for discriminating esophageal carcinoma
US8642271B2 (en) * 2009-08-27 2014-02-04 Case Western Reserve University Aberrant methylation of C6Orf150 DNA sequences in human colorectal cancer
WO2013033627A2 (en) * 2011-09-01 2013-03-07 The Regents Of The University Of California Diagnosis and treatment of arthritis using epigenetics

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070161031A1 (en) * 2005-12-16 2007-07-12 The Board Of Trustees Of The Leland Stanford Junior University Functional arrays for high throughput characterization of gene expression regulatory elements
US20100151468A1 (en) * 2007-04-11 2010-06-17 Manel Esteller Epigenetic biomarkers for early detection, therapeutic effectiveness, and relapse monitoring of cancer
US20140206565A1 (en) * 2007-08-01 2014-07-24 University Of Maryland, Baltimore Esophageal Cancer Markers

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
KAZ ET AL.: "Differential methylation in gene bodies and promoters in barrett's esophagus and esophageal adenocarcinoma revealed by genome-wide methylation array analysis", GASTROENTEROLOGY, vol. 144, no. 5, May 2013 (2013-05-01), pages S355 - S356 *
KAZ ET AL.: "Unique Methylation Signatures and Molecular Subclasses of Barrett's Esophagus and Esophageal Adenocarcinoma Revealed by Methylation Array Analysis", GASTROENTEROLOGY, vol. 140, no. 5, May 2011 (2011-05-01), pages S818, XP055461397 *
LIN ET AL.: "Deletion or epigenetic silencing of AJAP1 on 1p36 in glioblastoma", MOL CANCER RES., vol. 10, no. 2, 12 January 2012 (2012-01-12), pages 208 - 17, XP055461425 *
See also references of EP3240912A4 *
STOCKDALE ET AL.: "Epigenetic alterations in Barrett's esophagus and esophageal adenocarcinoma: Identification and evaluation of potential biomarkers", CANCER RES, vol. 74, no. 19, 1 October 2014 (2014-10-01), XP009504320 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10400286B2 (en) 2003-08-14 2019-09-03 Case Western Reserve University Methods and compositions for detecting gastrointestinal and other cancers
US10450615B2 (en) 2003-08-14 2019-10-22 Case Western Reserve University Methods and compositions for detecting gastrointestinal and other cancers
US11136629B2 (en) 2003-08-14 2021-10-05 Case Western Reserve University Methods and compositions for detecting gastrointestinal and other cancers
US20220154294A1 (en) * 2016-07-06 2022-05-19 Case Western Reserve University Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
WO2018009535A1 (en) * 2016-07-06 2018-01-11 Case Western Reserve University Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
US20190309372A1 (en) * 2016-07-06 2019-10-10 Case Western Reserve University Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
AU2017292668B2 (en) * 2016-07-06 2023-11-09 Case Western Reserve University Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
EP3336197A1 (en) * 2016-12-16 2018-06-20 GATC Biotech AG Epigenetic markers and related methods and means for the detection and management of ovarian cancer
WO2018109212A1 (en) * 2016-12-16 2018-06-21 Gatc Biotech Ag Epigenetic markers and related methods and means for the detection and management of ovarian cancer
WO2018109217A1 (en) * 2016-12-16 2018-06-21 Gatc Biotech Ag Epigenetic markers and related methods and means for the detection and management of certain cancers
US11920200B2 (en) 2016-12-16 2024-03-05 Eurofins Genomics Europe Sequencing GmbH Epigenetic markers and related methods and means for the detection and management of certain cancers
EP3382033A1 (en) * 2017-03-30 2018-10-03 Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen Method for determining blood counts based on dna methylation
IT201700072650A1 (en) * 2017-06-28 2018-12-28 Univ Degli Studi Cagliari METHOD FOR READING AND / OR FOR THE PROGNOSIS OF COLON RECTAL NEOPLASES
EP4180540A1 (en) * 2017-11-22 2023-05-17 Lepidyne Co., Ltd. Method for assessing prognosis or risk stratification of liver cancer by using cpg methylation variation in gene
EP3715474A4 (en) * 2017-11-22 2022-02-09 Lepidyne Co., Ltd. Method for assessing prognosis or risk stratification of liver cancer by using cpg methylation variation in gene
US20220349012A1 (en) * 2017-12-29 2022-11-03 City Of Hope Dna methylation diagnostic test for breast cancer
TWI815043B (en) * 2020-04-30 2023-09-11 大陸商廣州康立明生物科技股份有限公司 Tumor detection reagent and kit

Also Published As

Publication number Publication date
EP3240912A4 (en) 2018-07-25
CA2972782A1 (en) 2016-07-07
AU2015374019B2 (en) 2022-03-10
US20190136325A1 (en) 2019-05-09
EP3240912A1 (en) 2017-11-08
WO2016109782A2 (en) 2016-07-07
US20170369948A1 (en) 2017-12-28
WO2016109782A3 (en) 2016-09-01
AU2022201153A1 (en) 2022-03-17
AU2015374019A1 (en) 2017-08-03
US20240068045A1 (en) 2024-02-29

Similar Documents

Publication Publication Date Title
US20240068045A1 (en) Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
US7432050B2 (en) Methods and compositions for detecting colon cancers
US8221977B2 (en) Methods and compositions for detecting colon cancers
US20220154294A1 (en) Methods and compositions for detecting esophageal neoplasias and/or metaplasias in the esophagus
US11136629B2 (en) Methods and compositions for detecting gastrointestinal and other cancers
US8642271B2 (en) Aberrant methylation of C6Orf150 DNA sequences in human colorectal cancer

Legal Events

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

Ref document number: 15876289

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2972782

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015876289

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2015374019

Country of ref document: AU

Date of ref document: 20151230

Kind code of ref document: A