WO2014164365A1 - Methods of detecting head and neck cancer - Google Patents

Methods of detecting head and neck cancer Download PDF

Info

Publication number
WO2014164365A1
WO2014164365A1 PCT/US2014/022146 US2014022146W WO2014164365A1 WO 2014164365 A1 WO2014164365 A1 WO 2014164365A1 US 2014022146 W US2014022146 W US 2014022146W WO 2014164365 A1 WO2014164365 A1 WO 2014164365A1
Authority
WO
WIPO (PCT)
Prior art keywords
markers
hnc
subject
levels
cells
Prior art date
Application number
PCT/US2014/022146
Other languages
French (fr)
Inventor
Harry Stylli
Colleen KELLY
Original Assignee
Harry Stylli
Kelly Colleen
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 Harry Stylli, Kelly Colleen filed Critical Harry Stylli
Publication of WO2014164365A1 publication Critical patent/WO2014164365A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/5055Cells of the immune system involving macrophages
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • 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
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/50Determining the risk of developing a disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • This invention relates generally to using biological markers for the diagnosis, prognosis, and monitoring of head and neck cancer.
  • Leukocytes begin as pluripotent hematopoietic stem cells in the bone marrow and develop along either the myeloid lineage (monocytes, macrophages, neutrophils, eosinophils, and basophils) or the lymphoid lineage (T and B lymphocytes and natural killer cells).
  • myeloid lineage e.g., neutrophils and macrophages
  • the major function of the myeloid lineage cells e.g., neutrophils and macrophages
  • Phagocytes from healthy animals do not replicate and are diploid, i.e., have a DNA content of 2n.
  • each cell contains ⁇ 10 ng DNA, ⁇ 20 ng RNA, and ⁇ 300 ng of protein.
  • Non-phagocytic cells are also diploid and are not involved in the internalization of dead cells or infectious organisms and have a DNA index of one.
  • the lifetime of various white blood cell subpopulations varies from a few days (e.g., neutrophils) to several months (e.g., macrophages). Like other cell types, leukocytes age and eventually die.
  • human blood- and tissue-derived phagocytes e.g., neutrophils
  • phagocytes e.g., neutrophils
  • apoptosis markers of programmed cell death
  • caspase activation e.g., caspase activation
  • pyknotic nuclei e.g., pyknotic nuclei
  • chromatin fragmentation e.g., phosphatidylserine, sugars
  • One object of the present invention is to provide diagnostic methods that can facilitate the detection of head and neck cancer markers, e.g., nucleic acids, proteins, and the like by using phagocytic cells alone, or in combination with non- phagocytic cells.
  • Another object of this invention is to provide methods of identifying head and neck cancer markers and further use such markers alone or together with any known markers to diagnose head and neck cancer.
  • the present invention provides methods for detecting or diagnosing head and neck cancer by using at least one or more markers selected from the HNC-Macro 1-100 (Table 1) and/or HNC-Neutro 1-100 (Table 2).
  • Levels e.g., gene expression levels, protein expression levels, or activity levels
  • levels of the selected markers may be measured from macrophages or neutrophils, respectively, and from non-phagocytes, from a subject. Such levels then can be compared, e.g., the levels of the selected markers in the phagocytic cells and in the non-phagocytic cells to identify one or more differences between the measured levels, indicating whether the subject has head and neck cancer.
  • the identified difference(s) can also be used for assessing the risk of developing head and neck cancer, prognosing head and neck cancer, monitoring head and neck cancer progression or regression, assessing the efficacy of a treatment for head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer.
  • the levels of the selected markers in the phagocytic cells may be compared to the levels of the selected markers in a control (e.g., a normal or healthy control subject, or a normal or healthy cell from the subject) to identify one or more differences between the measured levels, indicating whether the subject has head and neck cancer, the prognosis of the cancer and the monitoring of the cancer.
  • the identified difference(s) can also be used for assessing the risk of developing head and neck cancer, prognosing head and neck cancer, monitoring head and neck cancer progression or regression, assessing the efficacy of a treatment for head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer.
  • a method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject comprising the steps of:
  • a method for assessing the risk of developing head and neck cancer in a subject comprising the steps of:
  • a method for prognosing or aiding in the prognosis of head and neck cancer in a subject comprising the steps of:
  • the identified difference is indicative of the prognosis of said head and neck cancer in the subject.
  • a method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising:
  • g) identifying a difference between the first difference and the second difference, wherein the difference identified in g) is indicative of the efficacy of the treatment for said head and neck cancer in the subject.
  • a method for monitoring the progression or regression of head and neck cancer in a subject comprising:
  • a method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising:
  • a method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject comprising the steps of:
  • a method for assessing the risk of developing head and neck cancer in a subject comprising the steps of:
  • the identified difference indicates that the subject has a risk of developing said head and neck cancer.
  • a method for prognosing or aiding in the prognosis of head and neck cancer in a subject comprising the steps of:
  • a method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising:
  • a method for monitoring the progression or regression of head and neck cancer in a subject comprising:
  • a method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before administering the compound to the subject;
  • a method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject comprising the steps of:
  • Macro markers in a population of the subject's non-phagocytic cells and measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells;
  • a method for assessing the risk of developing head and neck cancer in a subject comprising the steps of:
  • a method for prognosing or aiding in the prognosis of head and neck cancer in a subject comprising the steps of: a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells, and
  • a method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising:
  • a method for monitoring the progression or regression of head and neck cancer in a subject comprising:
  • a method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising:
  • the standard parameter is selected from the group consisting of tumor stage, tumor grade, tumor size, tumor visual characteristics, tumor growth, tumor thickness, tumor progression, tumor metastasis tumor distribution within the body, odor, molecular pathology, genomics, or tumor angiograms.
  • HNC-Macro markers comprise one or more markers selected from the group consisting of NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • the selected HNC-Macro markers comprise one or more markers selected from the group consisting of HNC-Macro 8, 9, 10, 11, 12, 13, 15, 18, 22, 24, 27, 33, 35, 36, 37, 38, 40, 41, 42, 43, 44, 45, 47, 49, 51, 52, 54, 55, 58, 59, 60, 62, 63, 64, 65, 68, 69, 71, 73, 74, 75, 77, 80, 82, 83, 85, 86, 87, 88, 89, 91, 92, 94, 95, 97, and 99 and wherein the selected HNC-Macro markers are up-regulated or activated in the macrophage cells compared to the non-phagocytic cells.
  • the selected HNC-Macro markers comprise one or more markers selected from the group consisting of P2RY10 and TNFAIP3 and wherein the selected HNC-Macro markers are up-regulated or activated in the macrophage cells compared to the non-phagocytic cells.
  • the selected HNC-Macro markers comprise one or more markers selected from the group consisting of HNC-Macro 1, 2, 3, 4, 5, 6, 7, 14, 16, 17, 19, 20, 21, 23, 25, 26, 28, 29, 30, 31, 32, 34, 39, 46, 48, 50, 53, 56, 57, 61, 66, 67, 70, 72, 76, 78, 79, 81, 84, 90, 93, 96, 98, and 100 and wherein the selected HNC-Macro markers are down-regulated or inhibited in the macrophage cells compared to the non- phagocytic cells.
  • the selected HNC-Macro markers comprise one or more markers selected from the group consisting of NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, and CLN8 and wherein the selected HNC-Macro markers are down-regulated or inhibited in the macrophage cells compared to the non-phagocytic cells.
  • HNC-Neutro markers comprise one or more HNC-Neutro markers selected from the group consisting of SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
  • the selected FiNC-Neutro markers comprise one or more markers selected from the group consisting of FiNC-Neutro 1-100 and wherein the selected FFNC-Neutro markers are down-regulated or inhibited in the neutrophil cells compared to the non-phagocytic cells.
  • the selected FFNC-Neutro markers comprise one or more markers selected from the group consisting of RPS4X, SLC25A6, RPL3, RPL18, and OBFC2B and wherein the selected FFNC-Neutro markers are down-regulated or inhibited in the neutrophil cells compared to the non-phagocytic cells.
  • non- phagocytic cells are T cells, B cells, null cells, basophils, or mixtures thereof.
  • macrophage cells are isolated from a bodily fluid sample, tissues, or cells of the subject.
  • neutrophil cells are isolated from a bodily fluid sample, tissues, or cells of the subject.
  • the non- phagocytic cells are isolated from a bodily fluid sample, tissues, or cells of the subject.
  • the bodily fluid sample is blood, urine, stool, saliva, lymph fluid, cerebrospinal fluid, synovial fluid, cystic fluid, ascites, pleural effusion, fluid obtained from a pregnant woman in the first trimester, fluid obtained from a pregnant woman in the second trimester, fluid obtained from a pregnant woman in the third trimester, maternal blood, amniotic fluid, chorionic villus sample, fluid from a preimplantation embryo, maternal urine, maternal saliva, placental sample, fetal blood, lavage and cervical vaginal fluid, interstitial fluid, or ocular fluid.
  • neutrophil cells are isolated using antibodies, using a ligand that binds to a molecular receptor expressed on the plasma membranes of white blood cells, or by flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, microfluidics, magnetic separation technique, fluorescent- magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platforms, or a combination thereof.
  • non- phagocytic cells are isolated using antibodies, using a ligand that binds to a molecular receptor expressed on the plasma membranes of white blood cells, or by flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, microfluidics, magnetic separation technique, fluorescent- magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platforms, or a combination thereof.
  • the quantitative assay uses sequencing, direct sequencing, RNA sequencing, whole transcriptome shotgun sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, whole-genome sequencing, sequencing by hybridization, pyrosequencing, capillary electrophoresis, gel electrophoresis, duplex sequencing, cycle sequencing, single- base extension sequencing, solid-phase sequencing, high-throughput sequencing, massively parallel signature sequencing, emulsion PCR, sequencing by reversible dye terminator, paired-end sequencing, near-term sequencing, exonuclease sequencing, sequencing by ligation, short-read sequencing, single-molecule sequencing, sequencing-by-synthesis, real-time sequencing, reverse-terminator sequencing, nanopore sequencing, 454 sequencing, Solexa Genome Analyzer sequencing, SOLiD® sequencing, MS-PET sequencing, mass spectrometry,
  • RNAase mismatch analysis mass spectrometry, tandem mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI- TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole-time of flight (Q-TOF) mass spectrometry, atmospheric
  • kits of embodiment 73 or 75, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of
  • kits of embodiment 74 or 75, wherein the selected HNC-Neutro markers comprise one or more markers selected from the group consisting of
  • a method of treating or preventing head and neck cancer in a subject comprising administering to said subject an agent that modulates the activity or expression of at least one or more markers selected from the group consisting of HNC-Macro 1-100.
  • a method of treating or preventing head and neck cancer in a subject comprising administering to said subject an agent that modulates the activity or expression of at least one or more markers selected from the group consisting of HNC-Neutro 1 - 100.
  • Figure 1 depicts a diagram of a 3 -fold cross validation method.
  • Figure 2 depicts a summary of head and neck cancer markers identified from macrophage vs. T cell and neutrophil vs. T cell comparisons in head and neck cancer patients.
  • Figure 3 depicts a comparison of cancer detection using markers identified from macrophages and neutrophils vs. T cells, as compared to detection when the phagocyte gene expression is not compared to T cell gene expression.
  • Figure 4 depicts a purification method for validating methods of detecting head and neck cancer.
  • Figure 5 depicts a comparison of purification methods in validating a method of detecting head and neck cancer.
  • the present invention provides biological markers and methods of using them to detect a cancer. More specifically, the present invention provides biomarkers that are specific for head and neck cancers.
  • a “biomarker” or “marker” refers to an analyte (e.g., a nucleic acid, DNA, RNA, peptide, protein, or metabolite) that can be objectively measured and evaluated as an indicator for a biological process.
  • a marker is differentially detectable in phagocytes and is indicative of the presence or absence of head and neck cancer.
  • An analyte is differentially detectable if it can be distinguished quantitatively or qualitatively in phagocytes compared to a control, e.g., a normal or healthy control or non-phagocytic cells.
  • the present invention is based on the discovery that one or more markers selected from Table 1 (FiNC-Macro markers) or Table 2 (FTNC-Neutro markers) are useful in diagnosing head and neck cancer.
  • the biomarkers e.g., gene expression levels, protein expression levels, or protein activity levels
  • phagocytes e.g., macrophage or neutrophils
  • the ability of these markers to correlate with head and neck cancer diagnosis may be amplified by using them in combination.
  • a "level" of a marker of this invention can be qualitative (e.g., presence or absence) or quantitative (e.g., amounts, copy numbers, or dosages).
  • a level of a marker at a zero value can indicate the absence of this marker.
  • the levels of any marker of this invention can be measured in various forms.
  • the level can be a gene expression level, a R A transcript level, a protein expression level, a protein activity level, an enzymatic activity level.
  • the markers of this invention can be used in methods for diagnosing or aiding in the diagnosis of head and neck cancer by comparing levels (e.g., gene expression levels, or protein expression levels, or protein activities) of one or more head and neck cancer markers (e.g., nucleic acids or proteins) between phagocytes (e.g., macrophages or neutrophils) and non-phagocytic cells taken from the same individual.
  • This invention also provides methods for assessing the risk of developing head and neck cancer, prognosing said cancer, monitoring said cancer progression or regression, assessing the efficacy of a treatment, or identifying a compound capable of ameliorating or treating said cancer.
  • the methods comprise: a) measuring the levels of one or more markers selected from Table 1 (HNC-Macro markers) in a population of a subject's macrophage cells; b) measuring the levels of one or more of the selected markers in a population of a subject's non-phagocytic cells (e.g., T-cells, B-cells, null cells, basophils or the mixtures of two more non-phagocytic cells); comparing the measured levels in step a) to the measured levels in step b) and further identify a difference between the measured levels of a) and b). The identified difference is indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
  • HNC-Macro markers e.g., HNC-Macro markers
  • the methods comprise: a) measuring the levels of one or more markers selected from Table 1 (HNC-Macro markers) in a population of a subject's macrophage cells; identifying a difference between the measured levels of the selected markers in step a) and the levels of the selected markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject). The identified difference is indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
  • HNC-Macro markers e.g., HNC-Macro markers
  • the selected markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1-9, i.e., NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • the selected markers are up-regulated (see Table 1 for up-regulated markers) in head and neck cancer patients.
  • the selected markers are down-regulated (see Table 1 for down- regulated markers) in head and neck cancer patients.
  • the selected markers comprise at least one HNC-Macro Marker that is up-regulated and at least one HNC-Macro Marker that is down-regulated.
  • the selected markers consist of NR4A3 , PRDM 1 , CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • the methods comprise: a) measuring the levels of one or more markers selected from Table 2 (HNC-Neutro markers) in a population of a subject's neutrophil cells; b) measuring the levels of one or more of the selected markers in a population of a subject's non-phagocytic cells (e.g., T-cells, B-cells, null cells, basophils or the mixtures of two more non-phagocytic cells); comparing the measured levels in step a) to the measured levels in step b) and further identify a difference between the measured levels of a) and b). The identified difference is indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
  • HNC-Neutro markers HNC-Neutro markers
  • the methods comprise: a) measuring the levels of one or more markers selected from Table 2 (HNC-Neutro markers) in a population of a subject's neutrophil cells; identifying a difference between the measured levels of the selected markers in step a) and the levels of the selected markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject). The identified difference is indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
  • HNC-Neutro markers HNC-Neutro markers
  • the selected markers comprise one or more (e.g., two, three, four, five, or six) HNC-Neutro Markers 1-6, e.g., SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
  • the selected markers are up-regulated (see Table 2 for up-regulated markers) in head and neck cancer patients.
  • the selected markers are down-regulated (see Table 2 for down-regulated markers) in head and neck cancer patients.
  • the selected markers comprise at least one HNC-Neutro Marker that is up-regulated and at least one HNC-Neutro Marker that is down-regulated.
  • the selected markers consist of SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
  • the methods comprise: a) measuring the levels of one or more markers selected from Table 2 (HNC-Neutro markers) in a population of a subject's neutrophil cells and the levels of one or more markers selected from Table 1 (HNC-Macro markers) in a population of a subject's macrophage cells; b) measuring the levels of one or more of the selected HNC-Macro markers and the levels one or more of the selected HNC-Neutro markers in a population of a subject's non-phagocytic cells (e.g., T-cells, B-cells, null cells, basophils or the mixtures of two more non-phagocytic cells); identifying a difference between the measured levels of the selected HNC-Neutro markers of steps a) and b) and identifying a
  • the methods comprise: a) measuring the levels of one or more markers selected from Table 2 (HNC-Neutro markers) in a population of a subject's neutrophil cells and the levels of one or more markers selected from Table 1 (HNC-Macro markers) in a population of a subject's macrophage cells; identifying a difference between the measured levels of the selected HNC-Neutro markers of steps a) and the levels of the selected HNC-Neutro markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject) and identifying a difference between the measured levels of the selected HNC-Macro markers of step a) and the levels of the selected HNC-Macro markers in a control (e.g., a healthy control cell, or a control cell from a control cell from a
  • the selected HNC-Neutro markers comprise one or more (e.g., two, three, four, five, or six) HNC-Neutro Markers 1-6, e.g., SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B and the selected HNC-Macro markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1-9, i.e., NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • HNC-Neutro Markers 1-6 e.g., SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B
  • the selected HNC-Macro markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers
  • the selected markers are up-regulated (see Tables 1 and 2 for up-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers are down- regulated (see Tables 1 and 2 for down-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one marker (HNC-Macro or HNC-Neutro marker) that is up-regulated and at least one marker (HNC-Macro or HNC-Neutro marker) that is down-regulated. In some
  • the selected markers consist of SLC25A6, RPS4X, RPL3, RPL18, OBFC2B, NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 (Table 1) in a population of the subject's macrophage cells before the treatment, or at a first time point, or before
  • the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 (Table 1) in a population of the subject's macrophage cells before the treatment, or at a first time point, or before
  • step (a) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in step (a) and the levels of the one or more selected HNC-Macro markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject) before the treatment, or at the first time point, or before administration of the compound, respectively; c) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment, or at a second time point, or after administration of the compound, respectively; d) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in step c) and the levels of the one or more selected HNC-Macro markers in a control after the treatment, or at the second time point, or after administration of the compound, respectively; and e) identifying a difference between the first difference and the second difference, wherein the difference identified in e) is indicative of
  • the selected markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1- 9, i.e., NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • the selected markers are up-regulated (see Table 1 for up-regulated markers) in head and neck cancer patients.
  • the selected markers are down-regulated (see Table 1 for down- regulated markers) in head and neck cancer patients.
  • the selected markers comprise at least one HNC-Macro Marker that is up-regulated and at least one HNC-Macro Marker that is down-regulated.
  • the selected markers consist of NR4A3, PRDMl, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 (Table 2) in a population of the subject's neutrophil cells before the treatment, or at a first time point, or before
  • the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 (Table 2) in a population of the subject's neutrophil cells before the treatment, or at a first time point, or before
  • step (a) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in step (a) and the levels of the one or more selected HNC-Neutro markers in a control (e.g., a control cell from a healthy subject, or a normal or healthy cell from the subject) before the treatment, or at the first time point, or before administration of the compound, respectively; c) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment, or at a second time point, or after administration of the compound, respectively; d) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in step c) and the levels of the one or more selected HNC-Neutro markers in a control after the treatment, or at the second time point, or after administration of the compound, respectively; and e) identifying a difference between the first difference and the second difference, wherein the difference
  • the selected markers comprise one or more (e.g., two, three, four, five, or six) HNC-Neutro Markers 1-6, e.g., SLC25A6,
  • the selected markers are up-regulated (see Table 2 for up-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers are down-regulated (see Table 2 for down-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one HNC-Neutro Marker that is up-regulated and at least one HNC-Neutro Marker that is down-regulated. In some embodiments, the selected markers consist of SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
  • the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising:
  • the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising:
  • the selected HNC-Neutro markers comprise one or more (e.g., two, three, four, five, or six) HNC-Neutro Markers 1- 6, e.g., SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B and the selected HNC- Macro markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1-9, i.e., NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • HNC-Neutro Markers 1- 6 e.g., SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B
  • the selected HNC- Macro markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1-9,
  • the selected markers are up-regulated (see Tables 1 and 2 for up-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers are down-regulated (see Tables 1 and 2 for down-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one marker (HNC-Macro or HNC-Neutro marker) that is up-regulated and at least one marker (HNC-Macro or HNC-Neutro marker) that is down-regulated.
  • the selected markers consist of SLC25A6, RPS4X, RPL3, RPL18, OBFC2B, NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
  • At least one or more of the selected marker may be substituted with a biological marker different from any of the selected marker.
  • such biological marker may be a known marker for head and neck cancer.
  • such biological marker and the selected marker being substituted may belong to the same signaling or biological pathway (e.g., TGF-beta pathway, apoptosis pathway, programmed cell death-associated pathway), or may have similar biological function or activity (e.g., DNA binding activity, rR A binding activity, G-protein coupled receptor activity, insulin-like growth factor receptor binding activity, DNA binding transcription factor activity, ADP antiporter activity, receptor for purines coupled to G-proteins), or may be regulated by a common protein, or may belong to the same protein complex (e.g., ribosome).
  • TGF-beta pathway e.g., TGF-beta pathway, apoptosis pathway, programmed cell death-associated pathway
  • similar biological function or activity e.g., DNA binding activity, rR A binding activity, G-protein coupled receptor activity, insulin-like growth factor receptor binding activity, DNA binding transcription factor activity, ADP antiporter activity, receptor for purines coupled to G-proteins
  • a common protein e
  • a population of the subject's macrophage cells is used as the selected phagocytic cells for measuring the levels of the selected markers (e.g., HNC-Macro markers) and a population of the subject's T-cells is used as the selected non-phagocytic cells for measuring the levels of the selected markers (e.g., HNC-Macro markers).
  • the selected markers e.g., HNC-Macro markers
  • T-cells e.g., HNC-Macro markers
  • a population of the subject's neutrophil cells is used as the selected phagocytic cells for measuring the levels of the selected markers (e.g., HNC-Neutro markers) and a population of the subject's T-cells is used as the selected non-phagocytic cells for measuring the levels of the selected markers (e.g., HNC-Neutro markers).
  • the selected markers e.g., HNC-Neutro markers
  • the levels of two, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, or more markers selected from Table 1 or Table 2 are measured.
  • one or more markers selected from Table 1 and one or more markers selected from Table 2 are measured.
  • the gene names/descriptions provided in Tables 1 and 2 are merely illustrative.
  • the markers of this invention encompass all forms and variants of any specifically described markers, including, but not limited to, polymorphic or allelic variants, isoforms, mutants, derivatives, precursors including nucleic acids and pro-proteins, cleavage products, and structures comprised of any of the markers as constituent subunits of the fully assembled structure.
  • a "patient”, “subject”, or “individual” are used interchangeably and refer to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animals (e.g., bovines, porcines), companion animals (e.g., canines, felines) and rodents (e.g., mice and rats).
  • mammals such as humans, primates, livestock animals (e.g., bovines, porcines), companion animals (e.g., canines, felines) and rodents (e.g., mice and rats).
  • normal control As used herein, the terms "normal control”, “healthy control”, and “not- diseased cells” likewise mean a sample (e.g., cells, serum, tissue) taken from a source (e.g., subject, control subject, cell line) that does not have the condition or disease being assayed and therefore may be used to determine the baseline for the condition or disorder being measured.
  • a control subject refers to any individual that has not been diagnosed as having the disease or condition being assayed. It is also understood that the control subject, normal control, and healthy control, include data obtained and used as a standard, i.e. it can be used over and over again for multiple different subjects.
  • the data from the control sample could have been obtained in a different set of experiments, for example, it could be an average obtained from a number of healthy subjects and not actually obtained at the time the data for the subject was obtained.
  • diagnosis refers to methods by which the skilled artisan can estimate and/or determine whether or not a patient is suffering from a given disease or condition.
  • diagnosis also refers to staging (e.g., Stage I, II, III, or IV) of cancer.
  • staging e.g., Stage I, II, III, or IV
  • the skilled artisan often makes a diagnosis on the basis of one or more diagnostic indicators, e.g., a marker, the presence, absence, amount, or change in amount of which is indicative of the presence, severity, or absence of the condition.
  • prognosis refers to is used herein to refer to the likelihood of head and neck cancer progression, including recurrence of head and neck cancer.
  • PCT/US 11/44969, PCT/US 11/45018, and PCT/US09/31395 and U.S. Provisional Applications 61/660,518 and 61/660,427 are incorporated herein by reference for all purposes.
  • Methods using the head and neck cancer markers described herein provide high specificity, sensitivity, and accuracy in detecting and diagnosing head and neck cancer.
  • the methods also eliminate the "inequality of baseline” that is known to occur among individuals due to intrinsic (e.g., age, gender, ethnic background, health status and the like) and temporal variations in marker expression. Additionally, by using a comparison of phagocytes and non- phagocytes from the same individual, the methods also allow detection, diagnosis, and treatment to be personalized to the individual.
  • the invention provides non-invasive assays for the early detection of head and neck cancer, i.e., before the head and neck cancer can be diagnosed by conventional diagnostic techniques, e.g., imaging techniques, and, therefore, provide a foundation for improved decision-making relative to the needs and strategies for intervention, prevention, and treatment of individuals with such disease or condition.
  • diagnostic techniques e.g., imaging techniques
  • Phagocytic cells that can be used in the methods of this invention include all types of cells that are capable of ingesting various types of substances (e.g., apoptotic cells, infectious agents, dead cells, viable cells, cell-free DNAs, cell-free RNAs, cell-free proteins).
  • the phagocytic cells are neutrophils, macrophages, monocytes, dendritic cells, foam cells, mast cells, eosinophils, or keratinocytes.
  • the phagocytic cells can be a mixture of different types of phagocytic cells.
  • the phagocytic cells can be activated phagocytic cells, e.g., activated macrophages or neutrophils.
  • a phagocyte is a histiocyte, e.g., a Langerhans cell.
  • head and neck cancer refers to a cancer or tumor that occurs in the neck and the head except for the brain and the eyes. In general, it includes oral cancer, paranasal sinus and nasal cancer, labial cancer, pharyngeal cancer, laryngeal cancer, head tumor, cancer of ears, cancer of nasopharynx, oropharynx, or hypopharynx, or cancer of salivary gland. Head and neck cancers may begin in the squamous cells that line the moist, mucosal surfaces inside the head and neck (for example, inside the mouth, the nose, and the throat).
  • squamous cell carcinomas of the head and neck may be referred to as squamous cell carcinomas of the head and neck. Head and neck cancers may also begin in the salivary glands. Salivary glands contain many different types of cells that can become cancerous, so there are many different types of salivary gland cancer.
  • treating head and neck cancer refers to taking steps to obtain beneficial or desired results, including clinical results.
  • beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms associated with diseases or conditions.
  • administering or “administration of a compound or an agent to a subject can be carried out using one of a variety of methods known to those skilled in the art.
  • a compound or an agent can be administered, intravenously, arterially, intradermally, intramuscularly, intraperitonealy, intravenously, subcutaneously, ocularly, sublingually, orally (by ingestion), intranasally (by inhalation), intraspinally, intracerebrally, and transdermally (by absorbtion, e.g., through a skin duct).
  • a compound or agent can also appropriately be introduced by rechargeable or biodegradable polymeric devices or other devices, e.g., patches and pumps, or formulations, which provide for the extended, slow, or controlled release of the compound or agent.
  • Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods.
  • the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug.
  • a physician who instructs a patient to self-administer a drug, or to have the drug administered by another and/or who provides a patient with a prescription for a drug is
  • a compound or an agent is administered orally, e.g., to a subject by ingestion, or intravenously, e.g., to a subject by injection.
  • the orally administered compound or agent is in an extended release or slow release formulation, or administered using a device for such slow or extended release.
  • markers used in the methods of invention are up- regulated or activated in phagocytes (e.g., macrophages or neutrophils) compared to non-phagocytes.
  • markers used in the methods of invention are down-regulated or inhibited in phagocytes (e.g., macrophages or neutrophils) compared to non-phagocytes.
  • up-regulation or up- regulated can refer to an increase in expression levels (e.g., gene expression or protein expression), gene copy numbers, gene dosages, and other qualitative or quantitative detectable state of the markers.
  • down-regulation or down- regulated can refer to a decrease in expression levels, gene copy numbers, gene dosages, and other qualitative or quantitative detectable state of the markers.
  • activation or activated can refer to an active state of the marker, e.g., a phosphorylation state, a DNA methylation state, or a DNA acetylation state.
  • inhibition or inhibited can refer to a repressed state or an inactivated state of the marker, e.g., a de -phosphorylation state, a ubiquitination state, a DNA de-methylation state.
  • methods of this invention also comprise at least one of the following steps before determination of various levels: i) lysing the phagocytic or non-phagocytic cells; and ii) extracting cellular contents from the lysed cells. Any known cell lysis and extraction methods can be used herein.
  • at least one or more head and neck cancer markers are present in the phagocytes. In certain embodiments, there is no marker present in the cellular contents of the non-phagocytic cells.
  • the phagocytic cells and/or non-phagocytic cells are isolated from a bodily fluid sample, tissues, or population of cells.
  • Exemplary bodily fluid samples can be whole blood, urine, stool, saliva, lymph fluid, cerebrospinal fluid, synovial fluid, cystic fluid, ascites, pleural effusion, fluid obtained from a pregnant woman in the first trimester, fluid obtained from a pregnant woman in the second trimester, fluid obtained from a pregnant woman in the third trimester, maternal blood, amniotic fluid, chorionic villus sample, fluid from a preimplantation embryo, maternal urine, maternal saliva, placental sample, fetal blood, lavage and cervical vaginal fluid, interstitial fluid, buccal swab sample, sputum, bronchial lavage, Pap smear sample, or ocular fluid.
  • the phagocytic cells or non-phagocytic cells are isolated from white blood cells.
  • cell separation/isolation/purification methods are used to isolate populations of cells from bodily fluid sample, cells, or tissues of a subject.
  • a skilled worker can use any known cell
  • separation/isolation/purification techniques to isolate phagocytic cells and non- phagocytic cells from a bodily fluid.
  • Exemplary techniques include, but are not limited to, using antibodies, flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, microfluidics, magnetic separation technique, fluorescent-magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platform, or a combination thereof.
  • the phagocytic cells and/or non-phagocytic cells are isolated by using a product secreted by the cells.
  • the phagocytic cells and/or non-phagocytic cells are isolated by using a cell surface target (e.g., receptor protein) on the surface of the cells.
  • the cell surface target is a protein that has been engulfed by phagocytic cells.
  • the cell surface target is expressed by cells on their plasma membranes.
  • the cell surface target is an exogenous protein that is translocated on the plasma membranes, but not expressed by the cells (e.g., the phagocytic cells).
  • the cell surface target is a marker of head and neck cancer.
  • analytes include nucleic acids, proteins, or any combinations thereof.
  • markers include nucleic acids, proteins, or any combinations thereof.
  • nucleic acid is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), DNA-RNA hybrids, and analogs of the DNA or RNA generated using nucleotide analogs.
  • the nucleic acid molecule can be a nucleotide,
  • the nucleic acid is a transrenal nucleic acid.
  • a transrenal nucleic acid is an extracellular nucleic acid that is excreted in the urine. See, e.g., U.S. Patent Publication No. 20100068711 and U.S. Patent Publication No.
  • amino acid includes organic compounds containing both a basic amino group and an acidic carboxyl group. Included within this term are natural amino acids (e.g., L-amino acids), modified and unusual amino acids (e.g., D-amino acids and ⁇ -amino acids), as well as amino acids which are known to occur biologically in free or combined form but usually do not occur in proteins.
  • natural amino acids e.g., L-amino acids
  • modified and unusual amino acids e.g., D-amino acids and ⁇ -amino acids
  • Natural protein occurring amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, serine, threonine, tyrosine, tryptophan, proline, and valine.
  • Natural non-protein amino acids include arginosuccinic acid, citrulline, cysteine sulfuric acid, 3,4-dihydroxyphenylalanine, homocysteine, homoserine, ornithine, 3-monoiodotyrosine, 3,5-diiodotryosine, 3, 5, 5-triiodothyronine, and 3,3 ',5,5'- tetraiodothyronine.
  • Modified or unusual amino acids include D-amino acids, hydroxylysine, 4-hydroxyproline, N-Cbz- protected amino acids, 2,4-diaminobutyric acid, homoarginine, norleucine, N- methylaminobutyric acid, naphthylalanine, phenylglycine, alpha-phenylproline, tert-leucine, 4-aminocyclohexylalanine, N-methyl-norleucine, 3 ,4-dehydroproline, ⁇ , ⁇ -dimethylaminoglycine, N-methylaminoglycine, 4-aminopiperidine-4- carboxylic acid, 6-aminocaproic acid, trans-4-(aminomethyl)- cyclohexanecarboxylic acid, 2-, 3-, and 4-(aminomethyl)- benzoic acid, 1- aminocyclopentanecarboxylic acid, 1-aminocyclopropanecarboxylic acid, and 2-
  • peptide includes compounds that consist of two or more amino acids that are linked by means of a peptide bond. Peptides may have a molecular weight of less than 10,000 Daltons, less than 5,000 Daltons, or less than 2,500 Daltons. The term “peptide” also includes compounds containing both peptide and non-peptide components, such as pseudopeptide or
  • peptidomimetic residues or other non-amino acid components Such compounds containing both peptide and non-peptide components may also be referred to as a "peptide analog.”
  • protein includes compounds that consist of amino acids arranged in a linear chain and joined together by peptide bonds between the carboxyl and amino groups of adjacent amino acid residues. Proteins used in methods of the invention include, but are not limited to, amino acids, peptides, antibodies, antibody fragments, cytokines, lipoproteins, or glycoproteins.
  • antibody includes polyclonal antibodies, monoclonal antibodies (including full length antibodies which have an
  • immunoglobulin Fc region immunoglobulin Fc region
  • multispecific antibodies e.g., bispecific antibodies, diabodies, and single-chain molecules
  • antibody fragments e.g., Fab or F(ab') 2 , and Fv.
  • cytokine refers to a secreted protein or active fragment or mutant thereof that modulates the activity of cells of the immune system.
  • cytokines include, without limitation, interleukins, interferons, chemokines, tumor necrosis factors, colony-stimulating factors for immune cell precursors, and the like.
  • lipoprotein includes negatively charged compositions that comprise a core of hydrophobic cholesteryl esters and triglyceride surrounded by a surface layer of amphipathic phospholipids with which free cholesterol and apolipoproteins are associated.
  • Lipoproteins may be characterized by their density (e.g. very-low-density lipoprotein (VLDL), low- density lipoprotein (LDL) and high density lipoprotein (HDL)), which is determined by their size, the relative amounts of lipid and protein.
  • VLDL very-low-density lipoprotein
  • LDL low- density lipoprotein
  • HDL high density lipoprotein
  • Lipoproteins may also be characterized by the presence or absence of particular modifications (e.g. oxidization, acetylation, or glycation).
  • glycoprotein includes glycosides which have one or more oligo- or polysaccharides covalently attached to a peptide or protein.
  • exemplary glycoproteins can include, without limitation, immunoglobulins, members of the major histocompatibility complex, collagens, mucins, glycoprotein Ilb/IIIa, glycoprotein-41 (gp41) and glycoprotein- 120 (gpl2), follicle-stimulating hormone, alpha-fetoprotein, erythropoietin, transferrins, alkaline phosphatase, and lectins.
  • a sample may comprise one or more stabilizers for a cell or an analyte such as DNA, R A, and/or protein.
  • a sample may comprise a DNA stabilizer, an RNA stabilizer, and/or a protein stabilizer.
  • Stabilizers are well known in the art and include, for example, DNAse inhibitors, RNAse inhibitors, and protease inhibitors or equivalents thereof.
  • levels of at least one or more head and neck cancer markers are compared. This comparison can be quantitative or qualitative. Quantitative measurements can be taken using any of the assays described herein. For example, sequencing, direct sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, targeted sequencing, whole- genome sequencing, sequencing by hybridization, pyrosequencing, capillary electrophoresis, gel electrophoresis, duplex sequencing, cycle sequencing, single- base extension sequencing, solid-phase sequencing, high-throughput sequencing, massively parallel signature sequencing, emulsion PCR, co-amplification at lower denaturation temperature-PCR (COLD-PCR), sequencing by reversible dye terminator, paired-end sequencing, near-term sequencing, exonuclease sequencing, sequencing by ligation, short-read sequencing, single-molecule sequencing, sequencing-by-synthesis, real-time sequencing, reverse-terminator sequencing, nanopore sequencing, 454 sequencing, Solexa Genome Analyzer sequencing, SOLiD® sequencing, MS
  • Quantitative comparisons can include statistical analyses such as t-test, ANOVA, Krustal- Wallis, Wilcoxon, Mann- Whitney, and odds ratio. Quantitative differences can include differences in the levels of markers between levels or differences in the numbers of markers present between levels, and combinations thereof. Examples of levels of the markers can be, without limitation, gene expression levels, nucleic acid levels, and protein levels. Qualitative differences can include, but are not limited to, activation and inactivation, protein degradation, nucleic acid degradation, and covalent modifications.
  • the level is a nucleic acid level or a protein level, or a combination thereof.
  • the level can be qualitatively or quantitatively determined.
  • a nucleic acid level can be, without limitation, a genotypic level, a single nucleotide polymorphism level, a gene mutation level, a gene copy number level, a DNA methylation level, a DNA acetylation level, a chromosome dosage level, a gene expression level, or a combination thereof.
  • the nucleic acid level can be determined by any methods known in the art to detect genotypes, single nucleotide polymorphisms, gene mutations, gene copy numbers, DNA methylation states, DNA acetylation states, chromosome dosages.
  • Exemplary methods include, but are not limited to, polymerase chain reaction (PCR) analysis, sequencing analysis, electrophoretic analysis, restriction fragment length polymorphism (RFLP) analysis, Northern blot analysis, quantitative PCR, reverse-transcriptase-PCR analysis (RT-PCR), allele-specific oligonucleotide hybridization analysis, comparative genomic hybridization, heteroduplex mobility assay (HMA), single strand conformational polymorphism (SSCP), denaturing gradient gel electrophisis (DGGE), RNAase mismatch analysis, mass
  • MALDI-TOF desorption/ionization-time of flight
  • electrospray ionization (ESI) mass spectrometry mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole- time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), surface plasmon resonance, Southern blot analysis, in situ hybridization, fluorescence in situ hybridization (FISH), chromogenic in situ hybridization (CISH), immunohistochemistry (IHC), microarray, comparative genomic hybridization, karyotyping, multiplex ligation-dependent probe amplification (MLPA), Quantitative Multiplex PCR of Short Fluorescent Frag
  • HELP Ligation-mediated PCR
  • sequencing is used in a broad sense and refers to any technique known in the art that allows the order of at least some consecutive nucleotides in at least part of a nucleic acid to be identified, including without limitation at least part of an extension product or a vector insert.
  • exemplary sequencing techniques include targeted sequencing, single molecule real-time sequencing, whole transcriptome shotgun sequencing ("RNA-seq”), electron microscopy-based sequencing, transistor-mediated sequencing, direct sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, exon sequencing, whole-genome sequencing, sequencing by hybridization,
  • sequencing comprises an detecting the sequencing product using an instrument, for example but not limited to an ABI PRISM® 377 DNA Sequencer, an ABI PRISM® 310, 3100, 3100-Avant, 3730, or 3730x1 Genetic Analyzer, an ABI PRISM® 3700 DNA Analyzer, or an Applied Biosystems SOLiDTM System (all from Applied Biosystems), a Genome Sequencer 20 System (Roche Applied Science), or a mass spectrometer.
  • sequencing comprises emulsion PCR.
  • sequencing comprises a high throughput sequencing technique, for example but not limited to, massively parallel signature sequencing (MPSS).
  • MPSS massively parallel signature sequencing
  • phosphorylation state an ubiquitination state, a myristoylation state, or a conformational state of the protein.
  • a protein level can be detected by any methods known in the art for detecting protein expression levels, protein phosphorylation state, protein ubiquitination state, protein myristoylation state, or protein conformational state. In some embodiments, a protein level can be determined by an
  • immunohistochemistry assay an enzyme-linked immunosorbent assay (ELISA), in situ hybridization, chromatography, liquid chromatography, size exclusion chromatography, high performance liquid chromatography (HPLC), gas chromatography, mass spectrometry, tandem mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole- time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), radioimmun
  • the "difference" between different levels detected by the methods of this invention can refer to different gene copy numbers, different DNA, RNA, or protein expression levels, different DNA methylation states, different DNA acetylation states, and different protein modification states.
  • the difference can be a difference greater than 1 fold.
  • the difference is a 1.05-fold, 1.1 -fold, 1.2-fold, 1.3-fold, 1.4-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 4- fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or 10-fold difference.
  • the difference is any fold difference between 1-10, 2-10, 5-10, 10- 20, or 10-100 fold.
  • the difference is differential gene expression (DGE), e.g. DGE of phagocytes vs. non-phagocytes.
  • the DGE may be any number, provided that it is
  • Down-regulated genes have X ⁇ 0, while up-regulated genes have X > 0. See, e.g., Efron, J Am Stat Assoc 104: 1015-1028 (2009).
  • a general principle of assays to detect markers involves preparing a sample or reaction mixture that may contain the marker (e.g., one or more of DNA, RNA, or protein) and a probe under appropriate conditions and for a time sufficient to allow the marker and probe to interact and bind, thus forming a complex that can be removed and/or detected in the reaction mixture.
  • the marker e.g., one or more of DNA, RNA, or protein
  • a probe under appropriate conditions and for a time sufficient to allow the marker and probe to interact and bind, thus forming a complex that can be removed and/or detected in the reaction mixture.
  • one method to conduct such an assay would involve anchoring the marker or probe onto a solid phase support, also referred to as a substrate, and detecting target marker/probe complexes anchored on the solid phase at the end of the reaction.
  • a sample from a subject which is to be assayed for presence and/or concentration of marker, can be anchored onto a carrier or solid phase support.
  • the reverse situation is possible, in which the probe can be anchored to a solid phase and a sample from a subject can be allowed to react as an unanchored component of the assay.
  • biotinylated assay components can be prepared from biotin-NHS(N-hydroxy-succinimide) using techniques known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • biotinylation kit Pierce Chemicals, Rockford, IL
  • streptavidin-coated 96 well plates Piereptavidin-coated 96 well plates
  • the surfaces with immobilized assay components can be prepared in advance and stored.
  • suitable carriers or solid phase supports for such assays include any material capable of binding the class of molecule to which the marker or probe belongs.
  • Well known supports or carriers include, but are not limited to, glass, polystyrene, nylon, polypropylene, nylon, polyethylene, dextran, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • the non- immobilized component is added to the solid phase upon which the second component is anchored.
  • uncomplexed components may be removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized upon the solid phase.
  • the detection of marker/probe complexes anchored to the solid phase can be accomplished in a number of methods outlined herein.
  • the probe when it is the unanchored assay component, can be labeled for the purpose of detection and readout of the assay, either directly or indirectly, with detectable labels discussed herein and which are well-known to one skilled in the art.
  • marker/probe complex formation without further manipulation or labeling of either component (marker or probe), for example by utilizing the technique of fluorescence energy transfer (see, for example, U.S. Patent Nos. 5,631,169 and 4,868,103).
  • a fiuorophore label on the first, 'donor' molecule is selected such that, upon excitation with incident light of appropriate wavelength, its emitted fluorescent energy will be absorbed by a fluorescent label on a second 'acceptor' molecule, which in turn is able to fluoresce due to the absorbed energy.
  • the 'donor' protein molecule may simply utilize the natural fiuorescent energy of tryptophan residues.
  • Labels are chosen that emit different wavelengths of light, such that the 'acceptor' molecule label may be differentiated from that of the 'donor'. Since the efficiency of energy transfer between the labels is related to the distance separating the molecules, spatial relationships between the molecules can be assessed. In a situation in which binding occurs between the molecules, the fluorescent emission of the 'acceptor' molecule label in the assay should be maximal.
  • An FET binding event can be conveniently measured through standard fluorometric detection means well known in the art (e.g., using a fluorimeter).
  • determination of the ability of a probe to recognize a marker can be accomplished without labeling either assay component (probe or marker) by utilizing a technology such as real-time Biomolecular
  • BIA Interaction Analysis
  • analogous diagnostic and prognostic assays can be conducted with marker and probe as solutes in a liquid phase.
  • the complexed marker and probe are separated from uncomplexed components by any of a number of standard techniques, including but not limited to: differential centrifugation, chromatography, electrophoresis and immunoprecipitation.
  • differential centrifugation marker/probe complexes may be separated from uncomplexed assay components through a series of centrifugal steps, due to the different sedimentation equilibria of complexes based on their different sizes and densities (see, for example, Rivas and Minton (1993) Trends Biochem. Sci. 18:284).
  • Standard chromatographic techniques may also be utilized to separate complexed molecules from uncomplexed ones.
  • gel filtration chromatography separates molecules based on size, and through the utilization of an appropriate gel filtration resin in a column format, for example, the relatively larger complex may be separated from the relatively smaller uncomplexed components.
  • the relatively different charge properties of the marker/probe complex as compared to the uncomplexed components may be exploited to differentiate the complex from uncomplexed components, for example through the utilization of ion-exchange chromatography resins.
  • Such resins and chromatographic techniques are well known to one skilled in the art (see, e.g., Heegaard (1998) J. Mol. Recognit. 11 : 141; Hage and Tweed (1997) J.
  • Gel electrophoresis may also be employed to separate complexed assay components from unbound components (see, e.g., Ausubel et al, ed., Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1987 1999). In this technique, protein or nucleic acid complexes are separated based on size or charge, for example. In order to maintain the binding interaction during the electrophoretic process, non-denaturing gel matrix materials and conditions in the absence of reducing agent are typically preferred. Appropriate conditions to the particular assay and components thereof will be well known to one skilled in the art.
  • the level of mRNA corresponding to the marker can be determined either by in situ and/or by in vitro formats in a biological sample using methods known in the art. Many expression detection methods use isolated RNA. For in vitro methods, any RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of RNA from blood cells (see, e.g., Ausubel et al, ed., Current Protocols in Molecular Biology, John Wiley & Sons, New York 1987 1999). Additionally, large numbers of cells and/or samples can readily be processed using techniques well known to those of skill in the art, such as, for example, the single- step RNA isolation process of Chomczynski (1989, U.S. Patent No. 4,843,155).
  • Isolated mRNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses and probe arrays.
  • a diagnostic method for the detection of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to the mRNA encoded by the gene being detected.
  • the nucleic acid probe can be, for example, a full-length cDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to an mRNA or genomic DNA encoding a marker of the present invention.
  • Other suitable probes for use in the diagnostic assays of the invention are described herein. Hybridization of an mRNA with the probe indicates that the marker in question is being expressed.
  • the mRNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose.
  • the probe(s) are immobilized on a solid surface and the mRNA is contacted with the probe(s), for example, in a gene chip array.
  • a skilled artisan can readily adapt known mRNA detection methods for use in detecting the level of mRNA encoded by the markers of the present invention.
  • An alternative method for determining the level of mRNA corresponding to a marker of the present invention in a sample involves the process of nucleic acid amplification, e.g., by RT-PCR (the experimental embodiment set forth in U.S. Patent Nos. 4,683,195 and 4,683,202), COLD-PCR (Li et al. (2008) Nat. Med. 14:579), ligase chain reaction (Barany, 1991, Proc. Natl. Acad. Sci. USA, 88: 189), self sustained sequence replication (Guatelli et al, 1990, Proc. Natl. Acad. Sci. USA 87: 1874), transcriptional amplification system (Kwoh et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173), Q- Beta Replicase (Lizardi et al. (1988) Bio/Technology 6: 1197), rolling circle replication (U.S. Patent No.
  • amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5 ' or 3 ' regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between.
  • amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
  • mRNA does not need to be isolated from the sample ⁇ e.g., a bodily fluid ⁇ e.g., blood cells)) prior to detection.
  • a cell or tissue sample is prepared/processed using known histological methods. The sample is then immobilized on a support, typically a glass slide, and then contacted with a probe that can hybridize to mRNA that encodes the marker.
  • determinations may be based on the normalized expression level of the marker.
  • Expression levels are normalized by correcting the absolute expression level of a marker by comparing its expression to the expression of a gene that is not a marker, e.g., a housekeeping gene that is constitutively expressed. Suitable genes for normalization include housekeeping genes such as the actin gene, or epithelial cell- specific genes. This normalization allows the comparison of the expression level in a patient sample from one source to a patient sample from another source, e.g., to compare a population of phagocytic from an individual to a population of non-phagocytic cells from the individual.
  • an agent for detecting a protein or polypeptide can be an antibody capable of binding to the polypeptide, such as an antibody with a detectable label.
  • label with regard to a probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.
  • Examples of indirect labeling include detection of a primary antibody using a fiuorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fiuorescently labeled streptavidin.
  • Antibodies can be polyclonal or monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used. In one format, antibodies, or antibody fragments, can be used in methods such as Western blots or immunofluorescence techniques to detect the expressed proteins. In such uses, it is generally preferable to immobilize either the antibody or proteins on a solid support.
  • Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody.
  • Well known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, magnetite and the like.
  • a variety of formats can be employed to determine whether a sample contains a protein that binds to a given antibody.
  • formats include, but are not limited to, competitive and non-competitive immunoassay, enzyme immunoassay (EIA), radioimmunoassay (RIA), antigen capture assays, two-antibody sandwich assays, Western blot analysis, enzyme linked
  • Immunoabsorbant assay ELISA
  • a planar array a colorimetric assay, a chemiluminescent assay, a fluorescent assay, and the like.
  • Immunoassays including radioimmmunoassays and enzyme- linked immunoassays, are useful in the methods of the present invention.
  • a skilled artisan can readily adapt known protein/antibody detection methods for use in determining whether cells (e.g., bodily fluid cells such as blood cells) express a marker of the present invention.
  • protein isolated from cells e.g., bodily fluid cells such as blood cells
  • a solid phase support such as nitrocellulose
  • the support can then be washed with suitable buffers followed by treatment with the detectably labeled antibody.
  • the solid phase support can then be washed with the buffer a second time to remove unbound antibody.
  • the amount of bound label on the solid support can then be detected by conventional means.
  • assays are provided for diagnosis, prognosis, assessing the risk of developing head and neck cancer, assessing the efficacy of a treatment, monitoring the progression or regression of head and neck cancer, and identifying a compound capable of ameliorating or treating head and neck cancer.
  • An exemplary method for these methods involves obtaining a bodily fluid sample from a test subject, isolating phagocytes and non-phagocytes, and contacting the phagocytes and non-phagocytes with a compound or an agent capable of detecting one or more of the markers of the disease or condition, e.g., marker nucleic acid (e.g., mRNA, genomic DNA), marker peptide (e.g., polypeptide or protein), marker lipid (e.g., cholesterol), or marker metabolite (e.g., creatinine) such that the presence of the marker is detected.
  • an agent for detecting marker mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to marker mRNA or genomic DNA.
  • the nucleic acid probe can be, for example, a full-length marker nucleic acid or a portion thereof. Other suitable probes for use in the diagnostic assays of the invention are described herein.
  • a compound capable of ameliorating or treating head and neck cancer can include, without limitations, any substance that can improve symptoms or prognosis, prevent progression of the head and neck cancer, promote regression of the head and neck cancer, or eliminate the head and neck cancer.
  • the methods of the invention can also be used to detect genetic alterations in a marker gene, thereby determining if a subject with the altered gene is at risk for developing head and neck cancer characterized by misregulation in a marker protein activity or nucleic acid expression.
  • the methods include detecting, in phagocytes, the presence or absence of a genetic alteration characterized by an alteration affecting the integrity of a gene encoding a marker peptide and/or a marker gene.
  • such genetic alterations can be detected by ascertaining the existence of at least one of: 1) a deletion of one or more nucleotides from one or more markers genes; 2) an addition of one or more nucleotides to one or more markers genes; 3) a substitution of one or more nucleotides of one or more markers genes, 4) a chromosomal rearrangement of one or more markers genes; 5) an alteration in the level of a messenger RNA transcript of one or more markers genes; 6) aberrant modification of one or more markers genes, such as of the methylation pattern of the genomic DNA; 7) the presence of a non-wild type splicing pattern of a messenger RNA transcript of one or more markers genes; 8) a non-wild type level of a one or more markers proteins; 9) allelic loss of one or more markers genes; and 10) inappropriate post-translational modification of one or more markers proteins.
  • assays there are a large number of assays known in the art which can be used for detecting alterations in one or more
  • detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195, 4,683,202 and 5,854,033), such as real-time PCR, COLD-PCR (Li et al. (2008) Nat. Med. 14:579), anchor PCR, recursive PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. (1988) Science 241 : 1077; Prodromou and Pearl (1992) Protein Eng. 5:827; and Nakazawa et al. (1994) Proc.
  • PCR polymerase chain reaction
  • This method can include the steps of collecting a sample of cell free bodily fluid from a subject, isolating nucleic acid (e.g., genomic, mRNA or both) from the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to a marker gene under conditions such that hybridization and amplification of the marker gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
  • nucleic acid e.g., genomic, mRNA or both
  • Alternative amplification methods include: self sustained sequence replication (Guatelli et al, (1990) Proc. Natl. Acad. Sci. USA 87: 1874), transcriptional amplification system (Kwoh et al., (1989) Proc. Natl. Acad. Sci. USA 86:1173), Q Beta Replicase (Lizardi et al. (1988) Bio-Technology 6: 1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.
  • mutations in one or more markers genes from a sample can be identified by alterations in restriction enzyme cleavage patterns.
  • sample and control DNA is isolated, optionally amplified, digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA.
  • sequence specific ribozymes see, for example, U.S. Pat. No. 5,498,531 can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
  • genetic mutations in one or more of the markers described herein can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high density arrays containing hundreds or thousands of oligonucleotides probes (Cronin et al. (1996) Human Mutation 7: 244; Kozal et al. (1996) Nature Medicine 2:753).
  • a marker nucleic acid can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin, M. T. et al. supra. Briefly, a first
  • hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations. This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
  • any of a variety of sequencing reactions known in the art can be used to directly sequence a marker gene and detect mutations by comparing the sequence of the sample marker gene with the corresponding wild-type (control) sequence.
  • Examples of sequencing reactions include those based on techniques developed by Maxam and Gilbert ((1977) Proc. Natl. Acad. Sci. USA 74:560) or Sanger ((1977) Proc. Natl. Acad. Sci. USA 74:5463). It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays ((1995)
  • Biotechniques 19:448) including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen et al. (1996) Adv. Chromatogr. 36: 127-162; and Griffin et al. (1993) Appl. Biochem. Biotechnol. 38:147).
  • RNA/RNA or RNA/DNA heteroduplexes Other methods for detecting mutations in a marker gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al. (1985) Science 230:1242).
  • the art technique of "mismatch cleavage" starts by providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing the wild- type marker sequence with potentially mutant RNA or DNA obtained from a tissue sample.
  • the double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as which will exist due to base pair mismatches between the control and sample strands.
  • RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S 1 nuclease to enzymatically digesting the mismatched regions.
  • either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, for example, Cotton et al. (1988) Proc. Natl. Acad. Sci. USA 85:4397; Saleeba et al. (1992) Methods Enzymol. 217:286.
  • the control DNA or RNA can be labeled for detection.
  • the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in marker cDNAs obtained from samples of cells.
  • DNA mismatch repair enzymes
  • the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. (1994) Carcinogenesis 15: 1657).
  • a probe based on a marker sequence e.g., a wild-type marker sequence
  • a marker sequence e.g., a wild-type marker sequence
  • the duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, for example, U.S. Patent No. 5,459,039.
  • alterations in electrophoretic mobility will be used to identify mutations in marker genes.
  • SSCP single strand conformation polymorphism
  • Single-stranded DNA fragments of sample and control marker nucleic acids will be denatured and allowed to renature.
  • the secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change.
  • the DNA fragments may be labeled or detected with labeled probes.
  • the sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence.
  • the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet. 7:5).
  • the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. (1985) Nature 313:495).
  • DGGE denaturing gradient gel electrophoresis
  • DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR.
  • a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys. Chem. 265: 12753).
  • oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324: 163; Saiki et al. (1989) Proc. Natl. Acad. Sci. USA 86:6230).
  • Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the
  • oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
  • Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al. (1989) Nucl. Acids Res. 17:2437) or at the extreme 3 ' end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner (1993) Tibtech 11 :238).
  • amplification may also be performed using Taq ligase for amplification (Barany (1991) Proc. Natl. Acad. Sci. USA 88: 189). In such cases, ligation will occur only if there is a perfect match at the 3 ' end of the 5 ' sequence making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.
  • An exemplary method for detecting the presence or absence of an analyte (e.g. , DNA, RNA, protein, polypeptide, or the like) corresponding to a marker of the invention in a biological sample involves obtaining a bodily fluid sample (e.g. , blood) from a test subject and contacting the bodily fluid sample with a compound or an agent capable of detecting one or more markers.
  • Detection methods described herein can be used to detect one or more markers in a biological sample in vitro as well as in vivo.
  • in vitro techniques for detection of mRNA include Northern hybridizations and in situ hybridizations.
  • In vitro techniques for detection of a polypeptide corresponding to a marker of the invention include enzyme linked immunosorbent assays (ELISAs), Western blots,
  • in vitro techniques for detection of genomic DNA include Southern hybridizations.
  • in vivo techniques for detection of a polypeptide corresponding to a marker of the invention include introducing into a subject a labeled antibody directed against the polypeptide.
  • the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques. Because each marker is also an analyte, any method described herein to detect the presence or absence of a marker can also be used to detect the presence or absence of an analyte.
  • the markers useful in the methods of the invention can include any mutation in any one of the markers. Mutation sites and sequences can be identified, for example, by databases or repositories of such information, e.g., The Human Gene Mutation Database (www.hgmd.cf.ac.uk), the Single Nucleotide Polymorphism Database (dbSNP, www.ncbi.nlm.nih.gov/projects/SNP), and the Online Mendelian Inheritance in Man (OMIM) website
  • kits that comprise marker detection agents that detect at least one or more of the head and neck cancer markers described herein.
  • the present invention also provides methods of treating or preventing head and neck cancer in a subject comprising administering to said subject an agent that modulates the activity or expression or disrupts the function of at least one or more of the markers of this invention.
  • the one or more markers identified by this invention may be used in the treatment of head and neck cancer.
  • a marker e.g., a protein or gene
  • a marker identified by the invention also may be used in any of the other methods of the invention, e.g., for monitoring the progression or regression of a disease or condition.
  • the one or more markers identified by the methods of this invention may have therapeutic potential.
  • a marker is identified as being up- regulated (or down-regulated), see, for example, the up-regulated (or down- regulated) markers in Tables 1 and 2, or activated (or inhibited) in phagocytic cells from a subject having head and neck cancer
  • a compound or an agent that is capable of down-regulating (or up-regulating) or inhibiting (or activating) said marker may be useful in treating head and neck cancer.
  • a gene protein expression level, a protein expression level, or a combination thereof may be useful in this aspect of the invention.
  • Example 1 Microarray analysis of head and neck cancer patients
  • Blood samples were collected from eight head and neck cancer patients and eight blood donors without head and neck cancer. Approximately 10 ml of blood was collected from each patient into purple top blood collection EDTA tubes (BD Biosciences, CA). Within 3 hours, macrophages, neutrophils and T cells were isolated from each blood sample and total RNA was extracted and purified on the same day. The healthy control blood samples were obtained from apheresis collars of anonymous platelet donors.
  • Gender determination of the blood donors was performed by PCR using two sets of primers, SRY primers (Forward: 5 -CAG TGT GAA ACG GGA GAA AAC AG-3'; Reverse: 5 * -ACT TCG CTG CAG AGT ACC GAA G-3') amplifying a 336 bp fragment on Y chromosome and AR6 primers (Forward: 5 * -CAA TCA GAG ACA TTC CCT CTG G-3 * ; Reverse: 5'- AGT GGT CCT CTC TGA ATC TC-3 * ) amplifying a 267 bp fragment on X chromosome (males have both fragments amplified; females have only one).
  • the PCR (36 cycles) was done under the conditions of 95 °C for 45 seconds, 56 °C for 45 seconds, and 72 °C for 45 seconds.
  • the freshly isolated white blood cell samples (in ⁇ 1 mL PBS) were incubated (25 min, 4°C, constant shaking) first with anti-monocyte coated Dynabeads® (CD 14 - Cat. No. 11149D, Life Technologies), then with anti-neutrophil coated Dynabeads® (CD 15 - Cat. No. 11137D, Life Technologies), and finally with anti- T cell coated
  • Dynabeads® (CD2 Pan T - Cat. No. 11159D, Life Technologies). Following each incubation, the bead-bound cells were separated using a magnet. The purity of these white blood cell subpopulations, which per manufacturer's specifications (Life Technologies) is >95%, was evident from the unique gene expression pattern obtained (cluster analysis). As soon as each white blood cell subpopulation was isolated, the magnetic bead bound cells were washed with lx PBS and lysed in Trizol®. The fractionation and subsequent lysis of all the three types of cells were completed in less than 2 hours after the isolation of the buffy coat.
  • RNA from macrophages, neutrophils, T cells, tumor tissue (TT), and "normal" head and neck tissue (NT) of head and neck cancer patients, and from macrophages, neutrophils, and T cells extracted from healthy male blood donors were used in gene expression profiling.
  • Biotinylated cDNA probes were prepared from 100 ng of each RNA sample, fragmented, and hybridized with Human Gene 1.0 ST chip (Affymetrix). Array signals of fluorescence were scanned, captured and recorded as CEL files. All the processing and analysis of the data were done using R 49 and Bioconductor software packages. To obtain the log2 transformed expression levels, the raw data files obtained in CEL file format were pre-processed using the oligo package and the RMA (robust multichip average algorithm) routine to background correct, quantile normalize and summarize at the core level.
  • RMA robust multichip average algorithm
  • Example 2 Statistical analysis of microarray data
  • FIG. 1 depicts a diagram of a three-fold cross validation, wherein the diagnostic accuracy is averaged from the three splits.
  • the ideal split for cross-validation is 10-fold for accurate and precise estimates of diagnostic accuracy.
  • there are more than 10 splits because there are many choices for which data points go into the folds.
  • Empirical Bayesian method was used as follows:
  • DE differential gene expression
  • the number of genes K to include in the signature was determined by comparing misclassification rates in independent test sets with cross-validation.
  • Errors were calculated using an average of 1-senstivity and 1 -specificity, and the cross-validated error was used to select markers.
  • the markers associated with head and neck cancer in macrophages vs. T cells (Table 1) and the markers associated with head and neck cancer in neutrophils vs. T cells (Table 2) were identified.
  • specific signatures of nine markers (for macrophages) and six markers (for neutrophils) also were identified that give especially high sensitivity and specificity.
  • a nine-marker signature (FiNC-Macro 1-9) from macrophages has a sensitivity of 95.5% and a specificity of 98.8%. The nine identified were
  • Figure 2 shows a summary of the head and neck cancer markers identified from macrophages and from neutrophils, as compared to T cells from the same individuals, for the HNC-Macro 1-100 and HNC-Neutro 1-100 markers.
  • HNC-Neutro 1-100 FiNC-Macro 1-100
  • HNC-Neutro 1-6 HNC-Neutro 1-100
  • the six HNC-Neutro markers include: SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
  • Figure 3 demonstrates the power of a paired within- subject (phagocyte to non-phagocyte) comparison to detect head and neck cancer as compared to phagocytes not paired with T cell data for comparison.
  • the paired approach (comparing macrophage or neutrophils to T cell expression) is better than the phagocyte gene expression alone.
  • Additional validation is performed by validating the gene signature on a new data set of ⁇ 50 head and neck cancer cases and 50 controls.
  • Final validation is performed by estimating the sensitivity and specificity of the final gene signature on a large sample. For example, 195 cases and 195 controls can be used to estimate a sensitivity/specificity of at least 97.5% with a 95% margin of error no more than 5%.
  • a challenge in designing a final validation study is that although cancer patients are pure, controls may have up to 20% false negatives.
  • a statistical issue is that, while estimating sensitivity is not a problem, specificity has an upper bound of 80%.
  • the solution is to purify the control set of patients.
  • a purification method uses secondary screening of all controls, wherein three methylated gene marker tests are used to purify the control test set:
  • HNC Head and Neck Cancer
  • HNC Head and Neck Cancer

Landscapes

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

Abstract

This invention provides a set of biological markers that are useful for detecting head and neck cancer. This invention further provides methods of using those biological markers for the diagnosis, prognosis, or monitoring of head and neck cancer.

Description

METHODS OF DETECTING HEAD AND NECK CANCER
[0001] Related Application
[0002] This application claims priority and benefit from U.S. Provisional Patent Application 61/775,558, filed March 9, 2013, the contents and disclosures of which are incorporated herein by reference in their entirety.
[0003] Field of the Invention
[0004] This invention relates generally to using biological markers for the diagnosis, prognosis, and monitoring of head and neck cancer.
[0005] Background of the Invention
[0006] Early diagnosis of a disease often increases the likelihood of successful treatment or cure of such disease. Current diagnostic methods, however, depend largely on population-derived average values obtained from healthy individuals. Personalized diagnostic methods are needed that enable the diagnosis, especially the early diagnosis, of the presence of a disease or a condition in individuals who are not known to have the disease or who have recurrent disease. This is of particular importance in head and neck cancer, which one of the most common cancer with more than 900,000 cases diagnosed each year.
[0007] Leukocytes begin as pluripotent hematopoietic stem cells in the bone marrow and develop along either the myeloid lineage (monocytes, macrophages, neutrophils, eosinophils, and basophils) or the lymphoid lineage (T and B lymphocytes and natural killer cells). The major function of the myeloid lineage cells (e.g., neutrophils and macrophages) is the phagocytosis of infectious organisms, live unwanted damaged cells, senescent and dead cells (apoptotic and necrotic), as well as the clearing of cellular debris. Phagocytes from healthy animals do not replicate and are diploid, i.e., have a DNA content of 2n. On average, each cell contains <10 ng DNA, <20 ng RNA, and <300 ng of protein. Non-phagocytic cells are also diploid and are not involved in the internalization of dead cells or infectious organisms and have a DNA index of one. [0008] The lifetime of various white blood cell subpopulations varies from a few days (e.g., neutrophils) to several months (e.g., macrophages). Like other cell types, leukocytes age and eventually die. During their aging process, human blood- and tissue-derived phagocytes (e.g., neutrophils) exhibit all the classic markers of programmed cell death (i.e., apoptosis), including caspase activation, pyknotic nuclei, and chromatin fragmentation. These cells also display a number of "eat-me" flags (e.g., phosphatidylserine, sugars) on the extracellular surfaces of their plasma membranes. Consequently, dying and dead cells and subcellular fragments thereof are cleared from tissues and blood by other phagocytic cells.
[0009] One object of the present invention is to provide diagnostic methods that can facilitate the detection of head and neck cancer markers, e.g., nucleic acids, proteins, and the like by using phagocytic cells alone, or in combination with non- phagocytic cells. Another object of this invention is to provide methods of identifying head and neck cancer markers and further use such markers alone or together with any known markers to diagnose head and neck cancer.
[0010] Summary of the Invention
[0011] In one aspect, the present invention provides methods for detecting or diagnosing head and neck cancer by using at least one or more markers selected from the HNC-Macro 1-100 (Table 1) and/or HNC-Neutro 1-100 (Table 2).
Levels (e.g., gene expression levels, protein expression levels, or activity levels) of the selected markers may be measured from macrophages or neutrophils, respectively, and from non-phagocytes, from a subject. Such levels then can be compared, e.g., the levels of the selected markers in the phagocytic cells and in the non-phagocytic cells to identify one or more differences between the measured levels, indicating whether the subject has head and neck cancer. The identified difference(s) can also be used for assessing the risk of developing head and neck cancer, prognosing head and neck cancer, monitoring head and neck cancer progression or regression, assessing the efficacy of a treatment for head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer.
[0012] In yet another aspect, the levels of the selected markers in the phagocytic cells may be compared to the levels of the selected markers in a control (e.g., a normal or healthy control subject, or a normal or healthy cell from the subject) to identify one or more differences between the measured levels, indicating whether the subject has head and neck cancer, the prognosis of the cancer and the monitoring of the cancer. The identified difference(s) can also be used for assessing the risk of developing head and neck cancer, prognosing head and neck cancer, monitoring head and neck cancer progression or regression, assessing the efficacy of a treatment for head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer.
[0013] Some embodiments of this invention are as follows:
1. A method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b),
wherein the identified difference indicates that the subject has said head and neck cancer.
2. A method for assessing the risk of developing head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b), wherein the identified difference indicates that the subject has a risk of developing said head and neck cancer.
3. A method for prognosing or aiding in the prognosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b),
wherein the identified difference is indicative of the prognosis of said head and neck cancer in the subject.
4. A method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before the treatment;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before the treatment;
c) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment;
e) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after the treatment;
f) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference, wherein the difference identified in g) is indicative of the efficacy of the treatment for said head and neck cancer in the subject.
5. A method for monitoring the progression or regression of head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells at a first time point;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells at the first time point;
c) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells at a second time point;
e) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells at the second time point;
f) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) is indicative of the progression or regression of said head and neck cancer in the subject. 6. A method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before administering the compound to the subject;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before administering the compound to the subject; c) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the administration of the compound;
e) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after the
administration of the compound;
f) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) indicates that the compound is capable of ameliorating or treating said head and neck cancer in the subject.
7. A method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells;
b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b),
wherein the identified difference indicates that the subject has said head and neck cancer.
8. A method for assessing the risk of developing head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells; b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b),
wherein the identified difference indicates that the subject has a risk of developing said head and neck cancer.
9. A method for prognosing or aiding in the prognosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells;
b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b),
wherein the identified difference is indicative of the prognosis of said head and neck cancer in the subject. 10. A method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before the treatment;
b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before the treatment;
c) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment;
e) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after the treatment; f) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) is indicative of the efficacy of the treatment for said head and neck cancer in the subject.
11. A method for monitoring the progression or regression of head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells at a first time point;
b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells at the first time point;
c) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells at a second time point;
e) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells at the second time point;
f) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) is indicative of the progression or regression of said head and neck cancer in the subject.
12. A method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before administering the compound to the subject;
b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before administering the compound to the subject;
c) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the administration of the compound;
e) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after the
administration of the compound;
f) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) indicates that the compound is capable of ameliorating or treating said head and neck cancer in the subject.
13. A method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells;
b) measuring the levels of the at least one or more selected HNC-
Macro markers in a population of the subject's non-phagocytic cells; and measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells;
c) identifying a difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
d) identifying a difference between the measured levels or activities the at least one or more selected HNC-Neutro markers in steps a) and b);
wherein the differences identified in c) and d) indicate that the subject has said head and neck cancer. 14. A method for assessing the risk of developing head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells;
b) measuring the levels of the at least one or more selected HNC- Macro markers in a population of the subject's non-phagocytic cells; and
measuring the levels of the at least one or more selected HNC-
Neutro markers in a population of the subject's non-phagocytic cells;
c) identifying a difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
d) identifying a difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
wherein the differences identified in c) and d) indicate that the subject has a risk of developing said head and neck cancer.
15. A method for prognosing or aiding in the prognosis of head and neck cancer in a subject, the method comprising the steps of: a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells;
c) identifying a difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
d) identifying a difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
wherein the differences identified in c) and d) are indicative of the prognosis of said head and neck cancer in the subject.
16. A method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before the treatment, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before the treatment;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before the treatment; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before the treatment; c) identifying a first difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and identifying a second difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment, and measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment;
e) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after the treatment; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after the treatment; f) identifying a third difference between the measured levels of the at least one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a fourth difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps d) and e);
h) identifying a difference between the first and second differences; and i) identifying a difference between the third and fourth differences, wherein the differences identified in h) and i) are indicative of the efficacy of the treatment for said head and neck cancer in the subject.
17. A method for monitoring the progression or regression of head and neck cancer in a subject comprising:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells at a first time point, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells at the first time point;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells at the first time point; and measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells at the first time point; c) identifying a first difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
identifying a second difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's macrophage cells at a second time point, and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells at the second time point; e) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells at the second time point; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells at the second time point;
f) identifying a third difference between the measured levels of the at least one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a fourth difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps d) and e);
h) identifying a difference between the first and second differences; and i) identifying a difference between the third and fourth differences, wherein the differences identified in h) and i) are indicative of the progression or regression of said head and neck cancer in the subject.
18. A method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before administering the compound to the subject, and measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before administering the compound to the subject;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before administering the compound to the subject; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before administering the compound to the subject;
c) identifying a first difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
identifying a second difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's macrophage cells after administering the compound to the subject, and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after administering the compound to the subject;
e) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after administering the compound to the subject; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after administering the compound to the subject;
f) identifying a third difference between the measured levels of the at least one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a fourth difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps d) and e);
h) identifying a difference between the first and second differences; and i) identifying a difference between the third and fourth differences, wherein the differences identified in h) and i) indicate that the compound is capable of ameliorating or treating said head and neck cancer in the subject.
19. The method of any one of the embodiments 1-18, further comprising measuring at least one standard parameter associated with said head and neck cancer.
20. The method of embodiment 19, wherein the standard parameter is selected from the group consisting of tumor stage, tumor grade, tumor size, tumor visual characteristics, tumor growth, tumor thickness, tumor progression, tumor metastasis tumor distribution within the body, odor, molecular pathology, genomics, or tumor angiograms.
21. The method of any one of the embodiments 13-18, wherein the selected HNC-Macro markers and the selected HNC-Neutro markers are measured from the same population of non-phagocytic cells in steps b) or e).
22. The method of any one of the embodiments 13-18, wherein the selected HNC-Macro markers and the selected HNC-Neutro are from different populations of non-phagocytic cells in steps b) or e).
23. The method of any one of the embodiments 1-6 and 13-18, wherein at least two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or fifteen markers are selected from HNC-Macro 1-100.
24. The method of any one of the embodiments 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
25. The method of any one of the embodiments 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of HNC-Macro 8, 9, 10, 11, 12, 13, 15, 18, 22, 24, 27, 33, 35, 36, 37, 38, 40, 41, 42, 43, 44, 45, 47, 49, 51, 52, 54, 55, 58, 59, 60, 62, 63, 64, 65, 68, 69, 71, 73, 74, 75, 77, 80, 82, 83, 85, 86, 87, 88, 89, 91, 92, 94, 95, 97, and 99 and wherein the selected HNC-Macro markers are up-regulated or activated in the macrophage cells compared to the non-phagocytic cells.
26. The method of any one of the embodiments 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of P2RY10 and TNFAIP3 and wherein the selected HNC-Macro markers are up-regulated or activated in the macrophage cells compared to the non-phagocytic cells.
27. The method of any one of the embodiments 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of HNC-Macro 1, 2, 3, 4, 5, 6, 7, 14, 16, 17, 19, 20, 21, 23, 25, 26, 28, 29, 30, 31, 32, 34, 39, 46, 48, 50, 53, 56, 57, 61, 66, 67, 70, 72, 76, 78, 79, 81, 84, 90, 93, 96, 98, and 100 and wherein the selected HNC-Macro markers are down-regulated or inhibited in the macrophage cells compared to the non- phagocytic cells.
28. The method of any one of the embodiments 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, and CLN8 and wherein the selected HNC-Macro markers are down-regulated or inhibited in the macrophage cells compared to the non-phagocytic cells.
29. The method of any one of the embodiments 7-18, wherein at least two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or fifteen markers are selected from HNC-Neutro 1-100.
30. The method of any one of the embodiments 7-18, wherein the selected HNC-Neutro markers comprise one or more HNC-Neutro markers selected from the group consisting of SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
31. The method of any one of the embodiments 7-18, wherein the selected FiNC-Neutro markers comprise one or more markers selected from the group consisting of FiNC-Neutro 1-100 and wherein the selected FFNC-Neutro markers are down-regulated or inhibited in the neutrophil cells compared to the non-phagocytic cells. 32. The method of any one of the embodiments 7-18, wherein the selected FFNC-Neutro markers comprise one or more markers selected from the group consisting of RPS4X, SLC25A6, RPL3, RPL18, and OBFC2B and wherein the selected FFNC-Neutro markers are down-regulated or inhibited in the neutrophil cells compared to the non-phagocytic cells.
33. The method of any one of the embodiments 1-6 and 13-18, further comprising lysing the macrophage cells and the non-phagocytic cells before a).
34. The method of any one of the embodiments 1-6 and 13-18, further comprising extracting the cellular contents from the macrophage cells and the non- phagocytic cells before a).
35. The method of any one of the embodiments 7-18, further comprising lysing the neutrophil cells and the non-phagocytic cells before a).
36. The method of any one of the embodiments 7-18, further comprising extracting the cellular contents from the neutrophil cells and the non- phagocytic cells before a). 37. The method of embodiment 34, wherein the cellular contents of the macrophage cells comprise viable diseased cells, dead diseased cells, apoptotic diseased cells, circulating tumor cells, infectious agents, fetal cells, trophoblasts, or fragments thereof.
38. The method of embodiment 36, wherein the cellular contents of the neutrophil cells comprise viable diseased cells, dead diseased cells, apoptotic diseased cells, circulating tumor cells, infectious agents, fetal cells, trophoblasts, or fragments thereof.
39. The method of embodiment 34, wherein the selected one or more markers are present in the cellular contents of the macrophage cells.
40. The method of embodiment 34, wherein the selected one or more markers are not present in the cellular contents of the non phagocytic cells. 41. The method of any one of the embodiments 1-6 and 13-18, wherein the macrophage cells express the one or more selected HNC-Macro markers.
42. The method of embodiment 36, wherein the selected one or more markers are present in the cellular contents of the neutrophil cells.
43. The method of embodiment 36, wherein the selected one or more markers are not present in the cellular contents of the non phagocytic cells.
44. The method of any one of the embodiments 7-18, wherein the neutrophil cells express the one or more selected HNC-Neutro markers.
45. The method of any one of the embodiments 1-18, wherein the non- phagocytic cells are T cells, B cells, null cells, basophils, or mixtures thereof. 46. The method of any one of the embodiments 1-6 and 13-18, wherein the macrophage cells are isolated from a bodily fluid sample, tissues, or cells of the subject. 47. The method of any one of the embodiments 7-18, wherein the neutrophil cells are isolated from a bodily fluid sample, tissues, or cells of the subject.
48. The method of any one of the embodiments 1-18, wherein the non- phagocytic cells are isolated from a bodily fluid sample, tissues, or cells of the subject. 49. The method of any one of the embodiments 46-48, wherein the bodily fluid sample is blood, urine, stool, saliva, lymph fluid, cerebrospinal fluid, synovial fluid, cystic fluid, ascites, pleural effusion, fluid obtained from a pregnant woman in the first trimester, fluid obtained from a pregnant woman in the second trimester, fluid obtained from a pregnant woman in the third trimester, maternal blood, amniotic fluid, chorionic villus sample, fluid from a preimplantation embryo, maternal urine, maternal saliva, placental sample, fetal blood, lavage and cervical vaginal fluid, interstitial fluid, or ocular fluid.
50. The method of any one of the embodiments 1-6 and 13-18, wherein the macrophage cells are isolated using antibodies, using a ligand that binds to a molecular receptor expressed on the plasma membranes of white blood cells, or by flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, microfluidics, magnetic separation technique, fluorescent- magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platforms, or a combination thereof.
51. The method of any one of the embodiments 7-18, wherein the neutrophil cells are isolated using antibodies, using a ligand that binds to a molecular receptor expressed on the plasma membranes of white blood cells, or by flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, microfluidics, magnetic separation technique, fluorescent- magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platforms, or a combination thereof.
52. The method of any one of the embodiments 1-18, wherein the non- phagocytic cells are isolated using antibodies, using a ligand that binds to a molecular receptor expressed on the plasma membranes of white blood cells, or by flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, microfluidics, magnetic separation technique, fluorescent- magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platforms, or a combination thereof.
53. The method of any one of the embodiments 1-6 and 13-18, wherein the macrophage cells are isolated using a product secreted by the macrophage cells. 54. The method of any one of the embodiments 7-18, wherein the neutrophil cells are isolated by using a product secreted by the neutrophil cells.
55. The method of any one the embodiments 1-6 and 13-18, wherein the macrophage cells are isolated by using a cell surface target on the surface of macrophage cells.
56. The method of any one of the embodiments 7-18, wherein the neutrophil cells are isolated by using a cell surface target on the surface of neutrophil cells.
57. The method of embodiment 55, wherein the target is expressed by the macrophage cells.
58. The method of embodiment 55, wherein the target is not expressed by the macrophage cells. 59. The method of embodiment 56, wherein the target is expressed by the neutrophil cells.
60. The method of embodiment 56, wherein the target is not expressed by the neutrophil cells.
61. The method of any one of the embodiments 55-60, wherein the target is a marker of said head and neck cancer. 62. The method of any one of the embodiments 1-18, wherein the measured levels are gene expression levels.
63. The method of any one of the embodiments 1-18, wherein the measured levels are protein expression levels.
64. The method of any one of the embodiment 1-18, wherein the levels or activities are measured by a qualitative assay, a quantitative assay, or a combination thereof. 65. The method of embodiment 64, wherein the quantitative assay uses sequencing, direct sequencing, RNA sequencing, whole transcriptome shotgun sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, whole-genome sequencing, sequencing by hybridization, pyrosequencing, capillary electrophoresis, gel electrophoresis, duplex sequencing, cycle sequencing, single- base extension sequencing, solid-phase sequencing, high-throughput sequencing, massively parallel signature sequencing, emulsion PCR, sequencing by reversible dye terminator, paired-end sequencing, near-term sequencing, exonuclease sequencing, sequencing by ligation, short-read sequencing, single-molecule sequencing, sequencing-by-synthesis, real-time sequencing, reverse-terminator sequencing, nanopore sequencing, 454 sequencing, Solexa Genome Analyzer sequencing, SOLiD® sequencing, MS-PET sequencing, mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole-time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), polymerase chain reaction (PCR) analysis, quantitative PCR, real-time PCR, fluorescence assay, colorimetric assay, chemiluminescent assay, or a combination thereof.
66. The method of embodiment 62, wherein the gene expression levels are measured by polymerase chain reaction (PCR) analysis, sequencing analysis, electrophoretic analysis, restriction fragment length polymorphism (RFLP) analysis, Northern blot analysis, quantitative PCR, reverse-transcriptase-PCR analysis (RT-PCR), allele-specific oligonucleotide hybridization analysis, comparative genomic hybridization, heteroduplex mobility assay (HMA), single strand conformational polymorphism (SSCP), denaturing gradient gel electrophisis (DGGE), RNAase mismatch analysis, mass spectrometry, tandem mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI- TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole-time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), surface plasmon resonance, Southern blot analysis, in situ hybridization, fluorescence in situ hybridization (FISH), chromogenic in situ hybridization (CISH), immunohistochemistry (IHC), microarray, comparative genomic hybridization, karyotyping, multiplex ligation- dependent probe amplification (MLPA), Quantitative Multiplex PCR of Short Fluorescent Fragments (QMPSF), microscopy, methylation specific PCR (MSP) assay, Hpall tiny fragment Enrichment by Ligation-mediated PCR (HELP) assay, radioactive acetate labeling assays, colorimetric DNA acetylation assay, chromatin immunoprecipitation combined with microarray (ChlP-on-chip) assay, restriction landmark genomic scanning, Methylated DNA immunoprecipitation (MeDIP), molecular break light assay for DNA adenine methyltransferase activity, chromatographic separation, methylation-sensitive restriction enzyme analysis, bisulfite-driven conversion of non-methylated cytosine to uracil, methyl-binding PCR analysis, or a combination thereof.
67. The method of embodiment 62, wherein the gene expression levels are measured by a sequencing technique selected from the group consisting of direct sequencing, RNA sequencing, whole transcriptome shotgun sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, whole- genome sequencing, sequencing by hybridization, pyrosequencing, capillary electrophoresis, gel electrophoresis, duplex sequencing, cycle sequencing, single- base extension sequencing, solid-phase sequencing, high-throughput sequencing, massively parallel signature sequencing, emulsion PCR, sequencing by reversible dye terminator, paired-end sequencing, near-term sequencing, exonuclease sequencing, sequencing by ligation, short-read sequencing, single-molecule sequencing, sequencing-by-synthesis, real-time sequencing, reverse-terminator sequencing, nanopore sequencing, 454 sequencing, Solexa Genome Analyzer sequencing, SOLiD® sequencing, MS-PET sequencing, mass spectrometry, and a combination thereof.
68. The method of embodiment 63, wherein the protein expression levels are measured by an immunohistochemistry assay, an enzyme-linked immunosorbent assay (ELISA), in situ hybridization, chromatography, liquid chromatography, size exclusion chromatography, high performance liquid chromatography (HPLC), gas chromatography, mass spectrometry, tandem mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI- TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole-time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), radioimmunoassays, microscopy, microfluidic chip-based assays, surface plasmon resonance, sequencing, Western blotting assay, or a combination thereof.
69. The method of any one the embodiments 1-68, wherein the subject is a mammal.
70. The method of embodiment 69, wherein the subject is a human.
71. The method of any one the embodiments 1-18, wherein the difference is greater than a 1-fold difference.
72. The method of embodiment 71, wherein the difference is at least 1.05-fold, 1.1 -fold, 1.2-fold, 1.3-fold, 1.4-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 4- fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or 10-fold difference.
73. A kit for measuring the levels of at least one or more markers selected from the group consisting of FiNC-Macro 1-100, comprising reagents for specifically measuring the levels of the selected FiNC-Macro markers.
74. A kit for measuring the levels of at least one or more markers selected from the group consisting of FiNC-Neutro 1-100, comprising reagents for specifically measuring the levels of the selected FFNC-Neutro markers.
75. A kit for measuring the levels of at least one or more markers selected from the group consisting of FFNC-Macro 1-100 and at least one or more markers selected from the group consisting of FFNC-Neutro 1-100, comprising reagents for specifically measuring the levels of the selected FFNC-Macro markers and reagents for specifically measuring the levels of the selected HNC-Neutro markers.
76. The kit of embodiment 73 or 75, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of
NR4A3, PRDMl, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
77. The kit of embodiment 74 or 75, wherein the selected HNC-Neutro markers comprise one or more markers selected from the group consisting of
SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
78. The kit of any one of the embodiments 73-77, wherein the reagents comprise one or more antibodies or fragments thereof, oligonucleotides, or aptamers.
79. A method of treating or preventing head and neck cancer in a subject comprising administering to said subject an agent that modulates the activity or expression of at least one or more markers selected from the group consisting of HNC-Macro 1-100.
80. A method of treating or preventing head and neck cancer in a subject comprising administering to said subject an agent that modulates the activity or expression of at least one or more markers selected from the group consisting of HNC-Neutro 1 - 100.
81. The method of embodiment 79 or 80, wherein the agent is a small molecule modulator, siRNA, or an antibody or fragment thereof. [0014] Brief Description of the Drawings
[0015] Figure 1 depicts a diagram of a 3 -fold cross validation method.
[0016] Figure 2 depicts a summary of head and neck cancer markers identified from macrophage vs. T cell and neutrophil vs. T cell comparisons in head and neck cancer patients.
[0017] Figure 3 depicts a comparison of cancer detection using markers identified from macrophages and neutrophils vs. T cells, as compared to detection when the phagocyte gene expression is not compared to T cell gene expression.
[0018] Figure 4 depicts a purification method for validating methods of detecting head and neck cancer.
[0019] Figure 5 depicts a comparison of purification methods in validating a method of detecting head and neck cancer.
[0020] Detailed Description of the Invention
[0021] The present invention provides biological markers and methods of using them to detect a cancer. More specifically, the present invention provides biomarkers that are specific for head and neck cancers.
[0022] As used here in, a "biomarker" or "marker" refers to an analyte (e.g., a nucleic acid, DNA, RNA, peptide, protein, or metabolite) that can be objectively measured and evaluated as an indicator for a biological process. In some embodiments, a marker is differentially detectable in phagocytes and is indicative of the presence or absence of head and neck cancer. An analyte is differentially detectable if it can be distinguished quantitatively or qualitatively in phagocytes compared to a control, e.g., a normal or healthy control or non-phagocytic cells.
[0023] The present invention is based on the discovery that one or more markers selected from Table 1 (FiNC-Macro markers) or Table 2 (FTNC-Neutro markers) are useful in diagnosing head and neck cancer. By measuring the levels of the biomarkers (e.g., gene expression levels, protein expression levels, or protein activity levels) in a population of phagocytes (e.g., macrophage or neutrophils) from a human subject, one can provide a reliable diagnosis for head and neck cancer. The ability of these markers to correlate with head and neck cancer diagnosis may be amplified by using them in combination. [0024] As used herein, a "level" of a marker of this invention can be qualitative (e.g., presence or absence) or quantitative (e.g., amounts, copy numbers, or dosages). In some embodiments, a level of a marker at a zero value can indicate the absence of this marker. The levels of any marker of this invention can be measured in various forms. For example, the level can be a gene expression level, a R A transcript level, a protein expression level, a protein activity level, an enzymatic activity level.
[0025] The markers of this invention can be used in methods for diagnosing or aiding in the diagnosis of head and neck cancer by comparing levels (e.g., gene expression levels, or protein expression levels, or protein activities) of one or more head and neck cancer markers (e.g., nucleic acids or proteins) between phagocytes (e.g., macrophages or neutrophils) and non-phagocytic cells taken from the same individual. This invention also provides methods for assessing the risk of developing head and neck cancer, prognosing said cancer, monitoring said cancer progression or regression, assessing the efficacy of a treatment, or identifying a compound capable of ameliorating or treating said cancer.
[0026] In a first aspect, the methods (e.g., diagnosis of head and neck cancer, prognosis of head and neck cancer, or assessing the risk of developing head and neck cancer) provided in the invention comprise: a) measuring the levels of one or more markers selected from Table 1 (HNC-Macro markers) in a population of a subject's macrophage cells; b) measuring the levels of one or more of the selected markers in a population of a subject's non-phagocytic cells (e.g., T-cells, B-cells, null cells, basophils or the mixtures of two more non-phagocytic cells); comparing the measured levels in step a) to the measured levels in step b) and further identify a difference between the measured levels of a) and b). The identified difference is indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
[0027] In a second aspect, the methods (e.g., d diagnosis of head and neck cancer, prognosis of head and neck cancer, or assessing the risk of developing head and neck cancer) provided in the invention comprise: a) measuring the levels of one or more markers selected from Table 1 (HNC-Macro markers) in a population of a subject's macrophage cells; identifying a difference between the measured levels of the selected markers in step a) and the levels of the selected markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject). The identified difference is indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
[0028] In the first and second aspects, the selected markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1-9, i.e., NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3. In some embodiments, the selected markers are up-regulated (see Table 1 for up-regulated markers) in head and neck cancer patients. In some
embodiments, the selected markers are down-regulated (see Table 1 for down- regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one HNC-Macro Marker that is up-regulated and at least one HNC-Macro Marker that is down-regulated. In some
embodiments, the selected markers consist of NR4A3 , PRDM 1 , CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
[0029] In a third aspect, the methods (e.g., diagnosis of head and neck cancer, prognosis of head and neck cancer, or assessing the risk of developing head and neck cancer) provided in the invention comprise: a) measuring the levels of one or more markers selected from Table 2 (HNC-Neutro markers) in a population of a subject's neutrophil cells; b) measuring the levels of one or more of the selected markers in a population of a subject's non-phagocytic cells (e.g., T-cells, B-cells, null cells, basophils or the mixtures of two more non-phagocytic cells); comparing the measured levels in step a) to the measured levels in step b) and further identify a difference between the measured levels of a) and b). The identified difference is indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
[0030] In a fourth aspect, the methods (e.g., diagnosis of head and neck cancer, prognosis of head and neck cancer, or assessing the risk of developing head and neck cancer) provided in the invention comprise: a) measuring the levels of one or more markers selected from Table 2 (HNC-Neutro markers) in a population of a subject's neutrophil cells; identifying a difference between the measured levels of the selected markers in step a) and the levels of the selected markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject). The identified difference is indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
[0031] In the third and fourth aspects, the selected markers comprise one or more (e.g., two, three, four, five, or six) HNC-Neutro Markers 1-6, e.g., SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B. In some embodiments, the selected markers are up-regulated (see Table 2 for up-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers are down-regulated (see Table 2 for down-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one HNC-Neutro Marker that is up-regulated and at least one HNC-Neutro Marker that is down-regulated. In some embodiments, the selected markers consist of SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
[0032] In a fifth aspect, the methods (e.g., diagnosis of head and neck cancer, prognosis of head and neck cancer, or assessing the risk of developing head and neck cancer) provided in the invention comprise: a) measuring the levels of one or more markers selected from Table 2 (HNC-Neutro markers) in a population of a subject's neutrophil cells and the levels of one or more markers selected from Table 1 (HNC-Macro markers) in a population of a subject's macrophage cells; b) measuring the levels of one or more of the selected HNC-Macro markers and the levels one or more of the selected HNC-Neutro markers in a population of a subject's non-phagocytic cells (e.g., T-cells, B-cells, null cells, basophils or the mixtures of two more non-phagocytic cells); identifying a difference between the measured levels of the selected HNC-Neutro markers of steps a) and b) and identifying a difference between the measured levels of the selected HNC-Macro markers of steps a) and b). The identified differences are indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
[0033] In a sixth aspect, the methods (e.g., diagnosis of head and neck cancer, prognosis of head and neck cancer, or assessing the risk of developing head and neck cancer) provided in the invention comprise: a) measuring the levels of one or more markers selected from Table 2 (HNC-Neutro markers) in a population of a subject's neutrophil cells and the levels of one or more markers selected from Table 1 (HNC-Macro markers) in a population of a subject's macrophage cells; identifying a difference between the measured levels of the selected HNC-Neutro markers of steps a) and the levels of the selected HNC-Neutro markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject) and identifying a difference between the measured levels of the selected HNC-Macro markers of step a) and the levels of the selected HNC-Macro markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject). The identified differences are indicative of the diagnosis (e.g., presence or absence), prognosis (e.g., lethal outcome, or tumor stage), or the risk of developing head and neck cancer.
[0034] In the fifth and sixth aspects, the selected HNC-Neutro markers comprise one or more (e.g., two, three, four, five, or six) HNC-Neutro Markers 1-6, e.g., SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B and the selected HNC-Macro markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1-9, i.e., NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3. In some embodiments, the selected markers are up-regulated (see Tables 1 and 2 for up-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers are down- regulated (see Tables 1 and 2 for down-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one marker (HNC-Macro or HNC-Neutro marker) that is up-regulated and at least one marker (HNC-Macro or HNC-Neutro marker) that is down-regulated. In some
embodiments, the selected markers consist of SLC25A6, RPS4X, RPL3, RPL18, OBFC2B, NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
[0035] In a seventh aspect, the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 (Table 1) in a population of the subject's macrophage cells before the treatment, or at a first time point, or before
administration of the compound, respectively; b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before the treatment, or at the first time point, or before administration of the compound, respectively; c) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b); d) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment, or at a second time point, or after administration of the compound, respectively; e) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after the treatment, or at the second time point, or after administration of the compound, respectively; f) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in steps d) and e); and g) identifying a difference between the first difference and the second difference, wherein the difference identified in g) is indicative of the efficacy of the treatment for the head and neck cancer, or the progression or regression of the head and neck cancer, or whether the compound is capable of ameliorating or treating the head and neck cancer, respectively, in the subject.
[0036] In a eighth aspect, the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 (Table 1) in a population of the subject's macrophage cells before the treatment, or at a first time point, or before
administration of the compound, respectively; b) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in step (a) and the levels of the one or more selected HNC-Macro markers in a control (e.g., a healthy control cell, or a control cell from a healthy subject) before the treatment, or at the first time point, or before administration of the compound, respectively; c) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment, or at a second time point, or after administration of the compound, respectively; d) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in step c) and the levels of the one or more selected HNC-Macro markers in a control after the treatment, or at the second time point, or after administration of the compound, respectively; and e) identifying a difference between the first difference and the second difference, wherein the difference identified in e) is indicative of the efficacy of the treatment for the head and neck cancer, or the progression or regression of the head and neck cancer, or whether the compound is capable of ameliorating or treating the head and neck cancer, respectively, in the subject.
[0037] In the seventh and eighth aspects, the selected markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1- 9, i.e., NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3. In some embodiments, the selected markers are up-regulated (see Table 1 for up-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers are down-regulated (see Table 1 for down- regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one HNC-Macro Marker that is up-regulated and at least one HNC-Macro Marker that is down-regulated. In some
embodiments, the selected markers consist of NR4A3, PRDMl, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
[0038] In a ninth aspect, the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 (Table 2) in a population of the subject's neutrophil cells before the treatment, or at a first time point, or before
administration of the compound, respectively; b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non- phagocytic cells before the treatment, or at the first time point, or before administration of the compound, respectively; c) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b); d) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment, or at a second time point, or after administration of the compound, respectively; e) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after the treatment, or at the second time point, or after administration of the compound, respectively; f) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in steps d) and e); and g) identifying a difference between the first difference and the second difference, wherein the difference identified in g) is indicative of the efficacy of the treatment for the head and neck cancer, or the progression or regression of the head and neck cancer, or whether the compound is capable of ameliorating or treating the head and neck cancer, respectively, in the subject.
[0039] In a tenth aspect, the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising: a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 (Table 2) in a population of the subject's neutrophil cells before the treatment, or at a first time point, or before
administration of the compound, respectively; b) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in step (a) and the levels of the one or more selected HNC-Neutro markers in a control (e.g., a control cell from a healthy subject, or a normal or healthy cell from the subject) before the treatment, or at the first time point, or before administration of the compound, respectively; c) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment, or at a second time point, or after administration of the compound, respectively; d) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in step c) and the levels of the one or more selected HNC-Neutro markers in a control after the treatment, or at the second time point, or after administration of the compound, respectively; and e) identifying a difference between the first difference and the second difference, wherein the difference identified in e) is indicative of the efficacy of the treatment for the head and neck cancer, or the progression or regression of the head and neck cancer, or whether the compound is capable of ameliorating or treating the head and neck cancer, respectively, in the subject.
[0040] In the ninth and tenth aspects, the selected markers comprise one or more (e.g., two, three, four, five, or six) HNC-Neutro Markers 1-6, e.g., SLC25A6,
RPS4X, RPL3, RPL18, and OBFC2B. In some embodiments, the selected markers are up-regulated (see Table 2 for up-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers are down-regulated (see Table 2 for down-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one HNC-Neutro Marker that is up-regulated and at least one HNC-Neutro Marker that is down-regulated. In some embodiments, the selected markers consist of SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
[0041] In an eleventh aspect, the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before the treatment, at a first time point, or before administration of the compound, respectively, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before the treatment, at the first time point, or before administration of the compound, respectively; b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before the treatment, at the first time point, or before administration of the compound, respectively; and measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before the treatment, at the first time point, or before administration of the compound, respectively; c) identifying a first difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
identifying a second difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment, at a second time point, or after administration of the compound, respectively, and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment, at the second time point, or after administration of the compound, respectively;
e) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after the treatment, at the second time point, or after administration of the compound, respectively; and measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after the treatment, at the second time point, or after administration of the compound, respectively;
f) identifying a third difference between the measured levels of the at least one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a fourth difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps d) and e);
h) identifying a difference between the first and second differences; and i) identifying a difference between the third and fourth differences, wherein the differences identified in h) and i) are indicative of the efficacy of the treatment for the head and neck cancer, or the progression or regression of the head and neck cancer, or whether the compound is capable of ameliorating or treating the head and neck cancer, respectively, in the subject. [0042] In an twelfth aspect, the methods provided in this invention for assessing the efficacy of a treatment for head and neck cancer, monitoring the progression or regression of head and neck cancer, or identifying a compound capable of ameliorating or treating head and neck cancer, respectively, in a subject comprising:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before the treatment, at a first time point, or before administration of the compound, respectively, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before the treatment, at the first time point, or before administration of the compound, respectively;
b) identifying a first difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and the levels of the at least one or more selected HNC-Macro markers in a control before the treatment, at the first time point, or before administration of the compound, respectively; and
identifying a second difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and the levels of the at least one or more selected HNC-Neutro markers in a control before the treatment, at the first time point, or before administration of the compound, respectively;
c) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment, at a second time point, or after administration of the compound, respectively, and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment, at the second time point, or after administration of the compound, respectively;
d) identifying a third difference between the measured levels of the at least one or more selected HNC-Macro markers in steps c) and the levels of the at least one or more selected HNC-Macro markers in a control after the treatment, at the second time point, or after administration of the compound, respectively; and e) identifying a fourth difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps c) and the levels of the at least one or more selected HNC-Neutro markers in a control after the treatment, at the second time point, or after administration of the compound, respectively; f) identifying a difference between the first and second differences; and g) identifying a difference between the third and fourth differences, wherein the differences identified in f) and g) are indicative of the efficacy of the treatment for the head and neck cancer, or the progression or regression of the head and neck cancer, or whether the compound is capable of ameliorating or treating the head and neck cancer, respectively, in the subject.
[0043] In the eleventh and twelfth aspects, the selected HNC-Neutro markers comprise one or more (e.g., two, three, four, five, or six) HNC-Neutro Markers 1- 6, e.g., SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B and the selected HNC- Macro markers comprise one or more (e.g., two, three, four, five, six, seven, eight or nine) HNC-Macro Markers 1-9, i.e., NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3. In some embodiments, the selected markers are up-regulated (see Tables 1 and 2 for up-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers are down-regulated (see Tables 1 and 2 for down-regulated markers) in head and neck cancer patients. In some embodiments, the selected markers comprise at least one marker (HNC-Macro or HNC-Neutro marker) that is up-regulated and at least one marker (HNC-Macro or HNC-Neutro marker) that is down-regulated. In some embodiments, the selected markers consist of SLC25A6, RPS4X, RPL3, RPL18, OBFC2B, NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
[0044] In various embodiments of the present invention, at least one or more of the selected marker (HNC-Macro or HNC-Neutro marker) may be substituted with a biological marker different from any of the selected marker. In some embodiments, such biological marker may be a known marker for head and neck cancer. In some embodiments, such biological marker and the selected marker being substituted may belong to the same signaling or biological pathway (e.g., TGF-beta pathway, apoptosis pathway, programmed cell death-associated pathway), or may have similar biological function or activity (e.g., DNA binding activity, rR A binding activity, G-protein coupled receptor activity, insulin-like growth factor receptor binding activity, DNA binding transcription factor activity, ADP antiporter activity, receptor for purines coupled to G-proteins), or may be regulated by a common protein, or may belong to the same protein complex (e.g., ribosome).
[0045] In various embodiments of the present invention, a population of the subject's macrophage cells is used as the selected phagocytic cells for measuring the levels of the selected markers (e.g., HNC-Macro markers) and a population of the subject's T-cells is used as the selected non-phagocytic cells for measuring the levels of the selected markers (e.g., HNC-Macro markers).
[0046] In various embodiments of the present invention, a population of the subject's neutrophil cells is used as the selected phagocytic cells for measuring the levels of the selected markers (e.g., HNC-Neutro markers) and a population of the subject's T-cells is used as the selected non-phagocytic cells for measuring the levels of the selected markers (e.g., HNC-Neutro markers).
[0047] In some embodiments, two sub-populations of phagocytic cells are used in the methods of this invention, i.e., phagocytic cells that have a DNA content greater than 2n (the >2n phagocytic cells) and phagocytic cells that have a DNA content of 2n (the =2n phagocytic cells). In those embodiments, the levels of the selected markers in the >2n phagocytic cells are compared to the =2n phagocytic cells to identify one or more difference. The identified difference indicate whether the subject has head and neck cancer, or has a risk of developing head and neck cancer, or has a progressing or progressive head and neck cancer.
[0048] In some embodiments, the levels of two, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, or more markers selected from Table 1 or Table 2 are measured. In some
embodiments, one or more markers selected from Table 1 and one or more markers selected from Table 2 are measured.
[0049] The gene names/descriptions provided in Tables 1 and 2 are merely illustrative. The markers of this invention encompass all forms and variants of any specifically described markers, including, but not limited to, polymorphic or allelic variants, isoforms, mutants, derivatives, precursors including nucleic acids and pro-proteins, cleavage products, and structures comprised of any of the markers as constituent subunits of the fully assembled structure.
[0050] A "patient", "subject", or "individual" are used interchangeably and refer to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animals (e.g., bovines, porcines), companion animals (e.g., canines, felines) and rodents (e.g., mice and rats).
[0051] As used herein, the terms "normal control", "healthy control", and "not- diseased cells" likewise mean a sample (e.g., cells, serum, tissue) taken from a source (e.g., subject, control subject, cell line) that does not have the condition or disease being assayed and therefore may be used to determine the baseline for the condition or disorder being measured. A control subject refers to any individual that has not been diagnosed as having the disease or condition being assayed. It is also understood that the control subject, normal control, and healthy control, include data obtained and used as a standard, i.e. it can be used over and over again for multiple different subjects. In other words, for example, when comparing a subject sample to a control sample, the data from the control sample could have been obtained in a different set of experiments, for example, it could be an average obtained from a number of healthy subjects and not actually obtained at the time the data for the subject was obtained.
[0052] The term "diagnosis" as used herein refers to methods by which the skilled artisan can estimate and/or determine whether or not a patient is suffering from a given disease or condition. In some embodiments, the term "diagnosis" also refers to staging (e.g., Stage I, II, III, or IV) of cancer. The skilled artisan often makes a diagnosis on the basis of one or more diagnostic indicators, e.g., a marker, the presence, absence, amount, or change in amount of which is indicative of the presence, severity, or absence of the condition.
[0053] The term "prognosis" as used herein refers to is used herein to refer to the likelihood of head and neck cancer progression, including recurrence of head and neck cancer. [0054] The disclosure of the International Applications PCT/US 11/44969, PCT/US 11/45018, and PCT/US09/31395 and U.S. Provisional Applications 61/660,518 and 61/660,427 are incorporated herein by reference for all purposes.
[0055] Each embodiment described herein may be combined with any other embodiment described herein.
[0056] Methods using the head and neck cancer markers described herein provide high specificity, sensitivity, and accuracy in detecting and diagnosing head and neck cancer. The methods also eliminate the "inequality of baseline" that is known to occur among individuals due to intrinsic (e.g., age, gender, ethnic background, health status and the like) and temporal variations in marker expression. Additionally, by using a comparison of phagocytes and non- phagocytes from the same individual, the methods also allow detection, diagnosis, and treatment to be personalized to the individual. Accordingly, in some embodiments, the invention provides non-invasive assays for the early detection of head and neck cancer, i.e., before the head and neck cancer can be diagnosed by conventional diagnostic techniques, e.g., imaging techniques, and, therefore, provide a foundation for improved decision-making relative to the needs and strategies for intervention, prevention, and treatment of individuals with such disease or condition.
[0057] The methods described herein are supported by whole genome microarray data of total RNA samples isolated from macrophages and neutrophils and from non-phagocytic T cells. The samples were obtained from human subjects with and without head and neck cancer. The data from these microarray experiments demonstrate that macrophage-T cell and neutrophil-T cell comparisons easily and accurately differentiate between head and neck cancer patients and human subjects without head and neck cancer.
[0058] The methods of this invention can be used together with any known diagnostic methods, such as physical inspection, visual inspection, biopsy, scanning, histology, radiology, imaging, ultrasound, use of a commercial kit, genetic testing, immunological testing, analysis of bodily fluids, or monitoring neural activity. [0059] Phagocytic cells that can be used in the methods of this invention include all types of cells that are capable of ingesting various types of substances (e.g., apoptotic cells, infectious agents, dead cells, viable cells, cell-free DNAs, cell-free RNAs, cell-free proteins). In some embodiments, the phagocytic cells are neutrophils, macrophages, monocytes, dendritic cells, foam cells, mast cells, eosinophils, or keratinocytes. In some embodiments, the phagocytic cells can be a mixture of different types of phagocytic cells. In some embodiments, the phagocytic cells can be activated phagocytic cells, e.g., activated macrophages or neutrophils. In some embodiments, a phagocyte is a histiocyte, e.g., a Langerhans cell.
[0060] The methods of this invention can be applied to head and neck cancer. As used herein, "head and neck cancer" refers to a cancer or tumor that occurs in the neck and the head except for the brain and the eyes. In general, it includes oral cancer, paranasal sinus and nasal cancer, labial cancer, pharyngeal cancer, laryngeal cancer, head tumor, cancer of ears, cancer of nasopharynx, oropharynx, or hypopharynx, or cancer of salivary gland. Head and neck cancers may begin in the squamous cells that line the moist, mucosal surfaces inside the head and neck (for example, inside the mouth, the nose, and the throat). These squamous cell cancers may be referred to as squamous cell carcinomas of the head and neck. Head and neck cancers may also begin in the salivary glands. Salivary glands contain many different types of cells that can become cancerous, so there are many different types of salivary gland cancer.
[0061] As used herein, "treating" head and neck cancer refers to taking steps to obtain beneficial or desired results, including clinical results. Beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms associated with diseases or conditions.
[0062] As used herein, "administering" or "administration of a compound or an agent to a subject can be carried out using one of a variety of methods known to those skilled in the art. For example, a compound or an agent can be administered, intravenously, arterially, intradermally, intramuscularly, intraperitonealy, intravenously, subcutaneously, ocularly, sublingually, orally (by ingestion), intranasally (by inhalation), intraspinally, intracerebrally, and transdermally (by absorbtion, e.g., through a skin duct). A compound or agent can also appropriately be introduced by rechargeable or biodegradable polymeric devices or other devices, e.g., patches and pumps, or formulations, which provide for the extended, slow, or controlled release of the compound or agent. Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods. In some aspects, the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug. For example, as used herein, a physician who instructs a patient to self-administer a drug, or to have the drug administered by another and/or who provides a patient with a prescription for a drug is
administering the drug to the patient. In some embodiments, a compound or an agent is administered orally, e.g., to a subject by ingestion, or intravenously, e.g., to a subject by injection. In some embodiments, the orally administered compound or agent is in an extended release or slow release formulation, or administered using a device for such slow or extended release.
[0063] In certain embodiments, markers used in the methods of invention are up- regulated or activated in phagocytes (e.g., macrophages or neutrophils) compared to non-phagocytes. In certain embodiments, markers used in the methods of invention are down-regulated or inhibited in phagocytes (e.g., macrophages or neutrophils) compared to non-phagocytes. As used herein, "up-regulation or up- regulated" can refer to an increase in expression levels (e.g., gene expression or protein expression), gene copy numbers, gene dosages, and other qualitative or quantitative detectable state of the markers. Similarly, "down-regulation or down- regulated" can refer to a decrease in expression levels, gene copy numbers, gene dosages, and other qualitative or quantitative detectable state of the markers. As used herein, "activation or activated" can refer to an active state of the marker, e.g., a phosphorylation state, a DNA methylation state, or a DNA acetylation state. Similarly, "inhibition or inhibited" can refer to a repressed state or an inactivated state of the marker, e.g., a de -phosphorylation state, a ubiquitination state, a DNA de-methylation state.
[0064] In certain embodiments, methods of this invention also comprise at least one of the following steps before determination of various levels: i) lysing the phagocytic or non-phagocytic cells; and ii) extracting cellular contents from the lysed cells. Any known cell lysis and extraction methods can be used herein. In certain embodiments, at least one or more head and neck cancer markers are present in the phagocytes. In certain embodiments, there is no marker present in the cellular contents of the non-phagocytic cells.
[0065] In certain embodiments, the phagocytic cells and/or non-phagocytic cells are isolated from a bodily fluid sample, tissues, or population of cells. Exemplary bodily fluid samples can be whole blood, urine, stool, saliva, lymph fluid, cerebrospinal fluid, synovial fluid, cystic fluid, ascites, pleural effusion, fluid obtained from a pregnant woman in the first trimester, fluid obtained from a pregnant woman in the second trimester, fluid obtained from a pregnant woman in the third trimester, maternal blood, amniotic fluid, chorionic villus sample, fluid from a preimplantation embryo, maternal urine, maternal saliva, placental sample, fetal blood, lavage and cervical vaginal fluid, interstitial fluid, buccal swab sample, sputum, bronchial lavage, Pap smear sample, or ocular fluid. In some
embodiments, the phagocytic cells or non-phagocytic cells are isolated from white blood cells.
[0066] In the methods of this invention, cell separation/isolation/purification methods are used to isolate populations of cells from bodily fluid sample, cells, or tissues of a subject. A skilled worker can use any known cell
separation/isolation/purification techniques to isolate phagocytic cells and non- phagocytic cells from a bodily fluid. Exemplary techniques include, but are not limited to, using antibodies, flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, microfluidics, magnetic separation technique, fluorescent-magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platform, or a combination thereof.
[0067] In certain embodiments, the phagocytic cells and/or non-phagocytic cells are isolated by using a product secreted by the cells. In certain embodiments, the phagocytic cells and/or non-phagocytic cells are isolated by using a cell surface target (e.g., receptor protein) on the surface of the cells. In some embodiments, the cell surface target is a protein that has been engulfed by phagocytic cells. In some embodiments, the cell surface target is expressed by cells on their plasma membranes. In some embodiments, the cell surface target is an exogenous protein that is translocated on the plasma membranes, but not expressed by the cells (e.g., the phagocytic cells). In some embodiments, the cell surface target is a marker of head and neck cancer.
[0068] In certain aspects of the methods described herein, analytes include nucleic acids, proteins, or any combinations thereof. In certain aspects of the methods described herein, markers include nucleic acids, proteins, or any combinations thereof. As used herein, the term "nucleic acid" is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), DNA-RNA hybrids, and analogs of the DNA or RNA generated using nucleotide analogs. The nucleic acid molecule can be a nucleotide,
oligonucleotide, double-stranded DNA, single-stranded DNA, multi-stranded DNA, complementary DNA, genomic DNA, non-coding DNA, messenger RNA (mRNAs), microRNA (miRNAs), small nucleolar RNA (snoRNAs), ribosomal RNA (rRNA), transfer RNA (tRNA), small interfering RNA (siRNA), heterogeneous nuclear RNAs (hnRNA), or small hairpin RNA (shRNA). In some embodiments, the nucleic acid is a transrenal nucleic acid. A transrenal nucleic acid is an extracellular nucleic acid that is excreted in the urine. See, e.g., U.S. Patent Publication No. 20100068711 and U.S. Patent Publication No.
20120021404.
[0069] As used herein, the term "amino acid" includes organic compounds containing both a basic amino group and an acidic carboxyl group. Included within this term are natural amino acids (e.g., L-amino acids), modified and unusual amino acids (e.g., D-amino acids and β-amino acids), as well as amino acids which are known to occur biologically in free or combined form but usually do not occur in proteins. Natural protein occurring amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, serine, threonine, tyrosine, tryptophan, proline, and valine. Natural non-protein amino acids include arginosuccinic acid, citrulline, cysteine sulfuric acid, 3,4-dihydroxyphenylalanine, homocysteine, homoserine, ornithine, 3-monoiodotyrosine, 3,5-diiodotryosine, 3, 5, 5-triiodothyronine, and 3,3 ',5,5'- tetraiodothyronine. Modified or unusual amino acids include D-amino acids, hydroxylysine, 4-hydroxyproline, N-Cbz- protected amino acids, 2,4-diaminobutyric acid, homoarginine, norleucine, N- methylaminobutyric acid, naphthylalanine, phenylglycine, alpha-phenylproline, tert-leucine, 4-aminocyclohexylalanine, N-methyl-norleucine, 3 ,4-dehydroproline, Ν,Ν-dimethylaminoglycine, N-methylaminoglycine, 4-aminopiperidine-4- carboxylic acid, 6-aminocaproic acid, trans-4-(aminomethyl)- cyclohexanecarboxylic acid, 2-, 3-, and 4-(aminomethyl)- benzoic acid, 1- aminocyclopentanecarboxylic acid, 1-aminocyclopropanecarboxylic acid, and 2- benzyl-5-aminopentanoic acid.
[0070] As used herein, the term "peptide" includes compounds that consist of two or more amino acids that are linked by means of a peptide bond. Peptides may have a molecular weight of less than 10,000 Daltons, less than 5,000 Daltons, or less than 2,500 Daltons. The term "peptide" also includes compounds containing both peptide and non-peptide components, such as pseudopeptide or
peptidomimetic residues or other non-amino acid components. Such compounds containing both peptide and non-peptide components may also be referred to as a "peptide analog."
[0071] As used herein, the term "protein" includes compounds that consist of amino acids arranged in a linear chain and joined together by peptide bonds between the carboxyl and amino groups of adjacent amino acid residues. Proteins used in methods of the invention include, but are not limited to, amino acids, peptides, antibodies, antibody fragments, cytokines, lipoproteins, or glycoproteins.
[0072] As used herein, the term "antibody" includes polyclonal antibodies, monoclonal antibodies (including full length antibodies which have an
immunoglobulin Fc region), antibody compositions with polyepitopic specificity, multispecific antibodies (e.g., bispecific antibodies, diabodies, and single-chain molecules, and antibody fragments (e.g., Fab or F(ab')2, and Fv). For the structure and properties of the different classes of antibodies, see e.g., Basic and Clinical Immunology, 8th Edition, Daniel P. Sties, Abba I. Terr and Tristram G. Parsolw (eds), Appleton & Lange, Norwalk, Conn., 1994, page 71 and Chapter 6.
[0073] As used herein, the term "cytokine" refers to a secreted protein or active fragment or mutant thereof that modulates the activity of cells of the immune system. Examples of cytokines include, without limitation, interleukins, interferons, chemokines, tumor necrosis factors, colony-stimulating factors for immune cell precursors, and the like.
[0074] As used herein, the term "lipoprotein" includes negatively charged compositions that comprise a core of hydrophobic cholesteryl esters and triglyceride surrounded by a surface layer of amphipathic phospholipids with which free cholesterol and apolipoproteins are associated. Lipoproteins may be characterized by their density (e.g. very-low-density lipoprotein (VLDL), low- density lipoprotein (LDL) and high density lipoprotein (HDL)), which is determined by their size, the relative amounts of lipid and protein. Lipoproteins may also be characterized by the presence or absence of particular modifications (e.g. oxidization, acetylation, or glycation).
[0075] As used herein, the term "glycoprotein" includes glycosides which have one or more oligo- or polysaccharides covalently attached to a peptide or protein. Exemplary glycoproteins can include, without limitation, immunoglobulins, members of the major histocompatibility complex, collagens, mucins, glycoprotein Ilb/IIIa, glycoprotein-41 (gp41) and glycoprotein- 120 (gpl2), follicle-stimulating hormone, alpha-fetoprotein, erythropoietin, transferrins, alkaline phosphatase, and lectins.
[0076] In some embodiments of the invention, a sample may comprise one or more stabilizers for a cell or an analyte such as DNA, R A, and/or protein. For example, a sample may comprise a DNA stabilizer, an RNA stabilizer, and/or a protein stabilizer. Stabilizers are well known in the art and include, for example, DNAse inhibitors, RNAse inhibitors, and protease inhibitors or equivalents thereof.
[0077] In some embodiments of the invention, levels of at least one or more head and neck cancer markers are compared. This comparison can be quantitative or qualitative. Quantitative measurements can be taken using any of the assays described herein. For example, sequencing, direct sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, targeted sequencing, whole- genome sequencing, sequencing by hybridization, pyrosequencing, capillary electrophoresis, gel electrophoresis, duplex sequencing, cycle sequencing, single- base extension sequencing, solid-phase sequencing, high-throughput sequencing, massively parallel signature sequencing, emulsion PCR, co-amplification at lower denaturation temperature-PCR (COLD-PCR), sequencing by reversible dye terminator, paired-end sequencing, near-term sequencing, exonuclease sequencing, sequencing by ligation, short-read sequencing, single-molecule sequencing, sequencing-by-synthesis, real-time sequencing, reverse-terminator sequencing, nanopore sequencing, 454 sequencing, Solexa Genome Analyzer sequencing, SOLiD® sequencing, MS-PET sequencing, mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole- time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), polymerase chain reaction (PCR) analysis, quantitative PCR, real-time PCR, fluorescence assay, colorimetric assay, chemiluminescent assay, or a combination thereof.
[0078] Quantitative comparisons can include statistical analyses such as t-test, ANOVA, Krustal- Wallis, Wilcoxon, Mann- Whitney, and odds ratio. Quantitative differences can include differences in the levels of markers between levels or differences in the numbers of markers present between levels, and combinations thereof. Examples of levels of the markers can be, without limitation, gene expression levels, nucleic acid levels, and protein levels. Qualitative differences can include, but are not limited to, activation and inactivation, protein degradation, nucleic acid degradation, and covalent modifications.
[0079] In certain embodiments of the invention, the level is a nucleic acid level or a protein level, or a combination thereof. The level can be qualitatively or quantitatively determined.
[0080] A nucleic acid level can be, without limitation, a genotypic level, a single nucleotide polymorphism level, a gene mutation level, a gene copy number level, a DNA methylation level, a DNA acetylation level, a chromosome dosage level, a gene expression level, or a combination thereof.
[0081] The nucleic acid level can be determined by any methods known in the art to detect genotypes, single nucleotide polymorphisms, gene mutations, gene copy numbers, DNA methylation states, DNA acetylation states, chromosome dosages. Exemplary methods include, but are not limited to, polymerase chain reaction (PCR) analysis, sequencing analysis, electrophoretic analysis, restriction fragment length polymorphism (RFLP) analysis, Northern blot analysis, quantitative PCR, reverse-transcriptase-PCR analysis (RT-PCR), allele-specific oligonucleotide hybridization analysis, comparative genomic hybridization, heteroduplex mobility assay (HMA), single strand conformational polymorphism (SSCP), denaturing gradient gel electrophisis (DGGE), RNAase mismatch analysis, mass
spectrometry, tandem mass spectrometry, matrix assisted laser
desorption/ionization-time of flight (MALDI-TOF) mass spectrometry,
electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole- time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), surface plasmon resonance, Southern blot analysis, in situ hybridization, fluorescence in situ hybridization (FISH), chromogenic in situ hybridization (CISH), immunohistochemistry (IHC), microarray, comparative genomic hybridization, karyotyping, multiplex ligation-dependent probe amplification (MLPA), Quantitative Multiplex PCR of Short Fluorescent Fragments (QMPSF), microscopy, methylation specific PCR (MSP) assay, Hpall tiny fragment
Enrichment by Ligation-mediated PCR (HELP) assay, radioactive acetate labeling assays, colorimetric DNA acetylation assay, chromatin immunoprecipitation combined with microarray (ChlP-on-chip) assay, restriction landmark genomic scanning, Methylated DNA immunoprecipitation (MeDIP), molecular break light assay for DNA adenine methyltransferase activity, chromatographic separation, methylation-sensitive restriction enzyme analysis, bisulfite-driven conversion of non-methylated cytosine to uracil, co-amplification at lower denaturation temperature -PCR (COLD-PCR), multiplex PCR, methyl-binding PCR analysis, or a combination thereof.
[0082] As used herein, the term "sequencing" is used in a broad sense and refers to any technique known in the art that allows the order of at least some consecutive nucleotides in at least part of a nucleic acid to be identified, including without limitation at least part of an extension product or a vector insert. Exemplary sequencing techniques include targeted sequencing, single molecule real-time sequencing, whole transcriptome shotgun sequencing ("RNA-seq"), electron microscopy-based sequencing, transistor-mediated sequencing, direct sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, exon sequencing, whole-genome sequencing, sequencing by hybridization,
pyrosequencing, capillary electrophoresis, gel electrophoresis, duplex sequencing, cycle sequencing, single-base extension sequencing, solid-phase sequencing, high- throughput sequencing, massively parallel signature sequencing, emulsion PCR, co-amplification at lower denaturation temperature -PCR (COLD-PCR), multiplex PCR, sequencing by reversible dye terminator, paired-end sequencing, near-term sequencing, exonuclease sequencing, sequencing by ligation, short-read sequencing, single-molecule sequencing, sequencing-by-synthesis, real-time sequencing, reverse-terminator sequencing, nanopore sequencing, 454 sequencing, Solexa Genome Analyzer sequencing, SOLiD® sequencing, MS-PET sequencing, mass spectrometry, and a combination thereof . In some embodiments, sequencing comprises an detecting the sequencing product using an instrument, for example but not limited to an ABI PRISM® 377 DNA Sequencer, an ABI PRISM® 310, 3100, 3100-Avant, 3730, or 3730x1 Genetic Analyzer, an ABI PRISM® 3700 DNA Analyzer, or an Applied Biosystems SOLiD™ System (all from Applied Biosystems), a Genome Sequencer 20 System (Roche Applied Science), or a mass spectrometer. In certain embodiments, sequencing comprises emulsion PCR. In certain embodiments, sequencing comprises a high throughput sequencing technique, for example but not limited to, massively parallel signature sequencing (MPSS). [0083] In further embodiments of the invention, a protein level can be a protein expression level, a protein activation level, or a combination thereof. In some embodiments, a protein activation level can comprise determining a
phosphorylation state, an ubiquitination state, a myristoylation state, or a conformational state of the protein.
[0084] A protein level can be detected by any methods known in the art for detecting protein expression levels, protein phosphorylation state, protein ubiquitination state, protein myristoylation state, or protein conformational state. In some embodiments, a protein level can be determined by an
immunohistochemistry assay, an enzyme-linked immunosorbent assay (ELISA), in situ hybridization, chromatography, liquid chromatography, size exclusion chromatography, high performance liquid chromatography (HPLC), gas chromatography, mass spectrometry, tandem mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole- time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), radioimmunoassays, microscopy, microfluidic chip-based assays, surface plasmon resonance, sequencing, Western blotting assay, or a combination thereof.
[0085] As used herein, the "difference" between different levels detected by the methods of this invention can refer to different gene copy numbers, different DNA, RNA, or protein expression levels, different DNA methylation states, different DNA acetylation states, and different protein modification states. The difference can be a difference greater than 1 fold. In some embodiments, the difference is a 1.05-fold, 1.1 -fold, 1.2-fold, 1.3-fold, 1.4-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 4- fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or 10-fold difference. In some embodiments, the difference is any fold difference between 1-10, 2-10, 5-10, 10- 20, or 10-100 fold. [0086] In some embodiments, the difference is differential gene expression (DGE), e.g. DGE of phagocytes vs. non-phagocytes. DGE can be measured as X = log2(Yp) - log2(YNp). The DGE may be any number, provided that it is
significantly different between the phagocytes and the non-phagocytes. For example, a 2-fold increased in gene expression could be represented as X = log2(Yp) - log2(YNp) = log2(Yp/Y NP) =log2(2) = 1 , while a 2-fold decrease in gene expression could be represented as X = log2(Yp) - log2(YNp) = log2(Yp/Y NP) =log2(l/2) = -1. Down-regulated genes have X < 0, while up-regulated genes have X > 0. See, e.g., Efron, J Am Stat Assoc 104: 1015-1028 (2009).
[0087] A general principle of assays to detect markers involves preparing a sample or reaction mixture that may contain the marker (e.g., one or more of DNA, RNA, or protein) and a probe under appropriate conditions and for a time sufficient to allow the marker and probe to interact and bind, thus forming a complex that can be removed and/or detected in the reaction mixture. These assays can be conducted in a variety of ways.
[0088] For example, one method to conduct such an assay would involve anchoring the marker or probe onto a solid phase support, also referred to as a substrate, and detecting target marker/probe complexes anchored on the solid phase at the end of the reaction. In one embodiment of such a method, a sample from a subject, which is to be assayed for presence and/or concentration of marker, can be anchored onto a carrier or solid phase support. In another embodiment, the reverse situation is possible, in which the probe can be anchored to a solid phase and a sample from a subject can be allowed to react as an unanchored component of the assay.
[0089] There are many established methods for anchoring assay components to a solid phase. These include, without limitation, marker or probe molecules which are immobilized through conjugation of biotin and streptavidin. Such biotinylated assay components can be prepared from biotin-NHS(N-hydroxy-succinimide) using techniques known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). In certain embodiments, the surfaces with immobilized assay components can be prepared in advance and stored. [0090] Other suitable carriers or solid phase supports for such assays include any material capable of binding the class of molecule to which the marker or probe belongs. Well known supports or carriers include, but are not limited to, glass, polystyrene, nylon, polypropylene, nylon, polyethylene, dextran, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
[0091] In order to conduct assays with the above mentioned approaches, the non- immobilized component is added to the solid phase upon which the second component is anchored. After the reaction is complete, uncomplexed components may be removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized upon the solid phase. The detection of marker/probe complexes anchored to the solid phase can be accomplished in a number of methods outlined herein.
[0092] In certain exemplary embodiments, the probe, when it is the unanchored assay component, can be labeled for the purpose of detection and readout of the assay, either directly or indirectly, with detectable labels discussed herein and which are well-known to one skilled in the art.
[0093] It is also possible to directly detect marker/probe complex formation without further manipulation or labeling of either component (marker or probe), for example by utilizing the technique of fluorescence energy transfer (see, for example, U.S. Patent Nos. 5,631,169 and 4,868,103). A fiuorophore label on the first, 'donor' molecule is selected such that, upon excitation with incident light of appropriate wavelength, its emitted fluorescent energy will be absorbed by a fluorescent label on a second 'acceptor' molecule, which in turn is able to fluoresce due to the absorbed energy. Alternately, the 'donor' protein molecule may simply utilize the natural fiuorescent energy of tryptophan residues. Labels are chosen that emit different wavelengths of light, such that the 'acceptor' molecule label may be differentiated from that of the 'donor'. Since the efficiency of energy transfer between the labels is related to the distance separating the molecules, spatial relationships between the molecules can be assessed. In a situation in which binding occurs between the molecules, the fluorescent emission of the 'acceptor' molecule label in the assay should be maximal. An FET binding event can be conveniently measured through standard fluorometric detection means well known in the art (e.g., using a fluorimeter).
[0094] In another embodiment, determination of the ability of a probe to recognize a marker can be accomplished without labeling either assay component (probe or marker) by utilizing a technology such as real-time Biomolecular
Interaction Analysis (BIA) (see, e.g., Sjolander, S. and Urbaniczky, C, 1991, Anal. Chem. 63:2338 2345 and Szabo et al, 1995, Curr. Opin. Struct. Biol. 5:699 705). As used herein, "BIA" or "surface plasmon resonance" is a technology for studying biospecific interactions in real time, without labeling any of the interactants (e.g., BlAcore). Changes in the mass at the binding surface (indicative of a binding event) result in alterations of the refractive index of light near the surface (the optical phenomenon of surface plasmon resonance (SPR)), resulting in a detectable signal which can be used as an indication of real-time reactions between biological molecules.
[0095] Alternatively, in another embodiment, analogous diagnostic and prognostic assays can be conducted with marker and probe as solutes in a liquid phase. In such an assay, the complexed marker and probe are separated from uncomplexed components by any of a number of standard techniques, including but not limited to: differential centrifugation, chromatography, electrophoresis and immunoprecipitation. In differential centrifugation, marker/probe complexes may be separated from uncomplexed assay components through a series of centrifugal steps, due to the different sedimentation equilibria of complexes based on their different sizes and densities (see, for example, Rivas and Minton (1993) Trends Biochem. Sci. 18:284). Standard chromatographic techniques may also be utilized to separate complexed molecules from uncomplexed ones. For example, gel filtration chromatography separates molecules based on size, and through the utilization of an appropriate gel filtration resin in a column format, for example, the relatively larger complex may be separated from the relatively smaller uncomplexed components. Similarly, the relatively different charge properties of the marker/probe complex as compared to the uncomplexed components may be exploited to differentiate the complex from uncomplexed components, for example through the utilization of ion-exchange chromatography resins. Such resins and chromatographic techniques are well known to one skilled in the art (see, e.g., Heegaard (1998) J. Mol. Recognit. 11 : 141; Hage and Tweed (1997) J.
Chromatogr. B. Biomed. Sci. Appl. 12:499). Gel electrophoresis may also be employed to separate complexed assay components from unbound components (see, e.g., Ausubel et al, ed., Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1987 1999). In this technique, protein or nucleic acid complexes are separated based on size or charge, for example. In order to maintain the binding interaction during the electrophoretic process, non-denaturing gel matrix materials and conditions in the absence of reducing agent are typically preferred. Appropriate conditions to the particular assay and components thereof will be well known to one skilled in the art.
[0096] In certain exemplary embodiments, the level of mRNA corresponding to the marker can be determined either by in situ and/or by in vitro formats in a biological sample using methods known in the art. Many expression detection methods use isolated RNA. For in vitro methods, any RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of RNA from blood cells (see, e.g., Ausubel et al, ed., Current Protocols in Molecular Biology, John Wiley & Sons, New York 1987 1999). Additionally, large numbers of cells and/or samples can readily be processed using techniques well known to those of skill in the art, such as, for example, the single- step RNA isolation process of Chomczynski (1989, U.S. Patent No. 4,843,155).
[0097] Isolated mRNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses and probe arrays. In certain exemplary embodiments, a diagnostic method for the detection of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to the mRNA encoded by the gene being detected. The nucleic acid probe can be, for example, a full-length cDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to an mRNA or genomic DNA encoding a marker of the present invention. Other suitable probes for use in the diagnostic assays of the invention are described herein. Hybridization of an mRNA with the probe indicates that the marker in question is being expressed.
[0098] In one format, the mRNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose. In an alternative format, the probe(s) are immobilized on a solid surface and the mRNA is contacted with the probe(s), for example, in a gene chip array. A skilled artisan can readily adapt known mRNA detection methods for use in detecting the level of mRNA encoded by the markers of the present invention.
[0099] An alternative method for determining the level of mRNA corresponding to a marker of the present invention in a sample involves the process of nucleic acid amplification, e.g., by RT-PCR (the experimental embodiment set forth in U.S. Patent Nos. 4,683,195 and 4,683,202), COLD-PCR (Li et al. (2008) Nat. Med. 14:579), ligase chain reaction (Barany, 1991, Proc. Natl. Acad. Sci. USA, 88: 189), self sustained sequence replication (Guatelli et al, 1990, Proc. Natl. Acad. Sci. USA 87: 1874), transcriptional amplification system (Kwoh et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173), Q- Beta Replicase (Lizardi et al. (1988) Bio/Technology 6: 1197), rolling circle replication (U.S. Patent No.
5,854,033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. As used herein, amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5 ' or 3 ' regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between. In general, amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
[0100] For in situ methods, mRNA does not need to be isolated from the sample {e.g., a bodily fluid {e.g., blood cells)) prior to detection. In such methods, a cell or tissue sample is prepared/processed using known histological methods. The sample is then immobilized on a support, typically a glass slide, and then contacted with a probe that can hybridize to mRNA that encodes the marker.
[0101] As an alternative to making determinations based on the absolute expression level of the marker, determinations may be based on the normalized expression level of the marker. Expression levels are normalized by correcting the absolute expression level of a marker by comparing its expression to the expression of a gene that is not a marker, e.g., a housekeeping gene that is constitutively expressed. Suitable genes for normalization include housekeeping genes such as the actin gene, or epithelial cell- specific genes. This normalization allows the comparison of the expression level in a patient sample from one source to a patient sample from another source, e.g., to compare a population of phagocytic from an individual to a population of non-phagocytic cells from the individual.
[0102] In one embodiment of this invention, a protein or polypeptide
corresponding to a marker is detected. In certain embodiments, an agent for detecting a protein or polypeptide can be an antibody capable of binding to the polypeptide, such as an antibody with a detectable label. As used herein, the term "labeled," with regard to a probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fiuorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fiuorescently labeled streptavidin. Antibodies can be polyclonal or monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used. In one format, antibodies, or antibody fragments, can be used in methods such as Western blots or immunofluorescence techniques to detect the expressed proteins. In such uses, it is generally preferable to immobilize either the antibody or proteins on a solid support. Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody. Well known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, magnetite and the like.
[0103] A variety of formats can be employed to determine whether a sample contains a protein that binds to a given antibody. Examples of such formats include, but are not limited to, competitive and non-competitive immunoassay, enzyme immunoassay (EIA), radioimmunoassay (RIA), antigen capture assays, two-antibody sandwich assays, Western blot analysis, enzyme linked
immunoabsorbant assay (ELISA), a planar array, a colorimetric assay, a chemiluminescent assay, a fluorescent assay, and the like. Immunoassays, including radioimmmunoassays and enzyme- linked immunoassays, are useful in the methods of the present invention. A skilled artisan can readily adapt known protein/antibody detection methods for use in determining whether cells (e.g., bodily fluid cells such as blood cells) express a marker of the present invention.
[0104] One skilled in the art will know many other suitable carriers for binding antibody or antigen, and will be able to adapt such support for use with the present invention. For example, protein isolated from cells (e.g., bodily fluid cells such as blood cells) can be run on a polyacrylamide gel electrophoresis and immobilized onto a solid phase support such as nitrocellulose. The support can then be washed with suitable buffers followed by treatment with the detectably labeled antibody. The solid phase support can then be washed with the buffer a second time to remove unbound antibody. The amount of bound label on the solid support can then be detected by conventional means.
[0105] In certain exemplary embodiments, assays are provided for diagnosis, prognosis, assessing the risk of developing head and neck cancer, assessing the efficacy of a treatment, monitoring the progression or regression of head and neck cancer, and identifying a compound capable of ameliorating or treating head and neck cancer. An exemplary method for these methods involves obtaining a bodily fluid sample from a test subject, isolating phagocytes and non-phagocytes, and contacting the phagocytes and non-phagocytes with a compound or an agent capable of detecting one or more of the markers of the disease or condition, e.g., marker nucleic acid (e.g., mRNA, genomic DNA), marker peptide (e.g., polypeptide or protein), marker lipid (e.g., cholesterol), or marker metabolite (e.g., creatinine) such that the presence of the marker is detected. In one embodiment, an agent for detecting marker mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to marker mRNA or genomic DNA. The nucleic acid probe can be, for example, a full-length marker nucleic acid or a portion thereof. Other suitable probes for use in the diagnostic assays of the invention are described herein.
[0106] As used herein, a compound capable of ameliorating or treating head and neck cancer can include, without limitations, any substance that can improve symptoms or prognosis, prevent progression of the head and neck cancer, promote regression of the head and neck cancer, or eliminate the head and neck cancer.
[0107] The methods of the invention can also be used to detect genetic alterations in a marker gene, thereby determining if a subject with the altered gene is at risk for developing head and neck cancer characterized by misregulation in a marker protein activity or nucleic acid expression. In certain embodiments, the methods include detecting, in phagocytes, the presence or absence of a genetic alteration characterized by an alteration affecting the integrity of a gene encoding a marker peptide and/or a marker gene. For example, such genetic alterations can be detected by ascertaining the existence of at least one of: 1) a deletion of one or more nucleotides from one or more markers genes; 2) an addition of one or more nucleotides to one or more markers genes; 3) a substitution of one or more nucleotides of one or more markers genes, 4) a chromosomal rearrangement of one or more markers genes; 5) an alteration in the level of a messenger RNA transcript of one or more markers genes; 6) aberrant modification of one or more markers genes, such as of the methylation pattern of the genomic DNA; 7) the presence of a non-wild type splicing pattern of a messenger RNA transcript of one or more markers genes; 8) a non-wild type level of a one or more markers proteins; 9) allelic loss of one or more markers genes; and 10) inappropriate post-translational modification of one or more markers proteins. As described herein, there are a large number of assays known in the art which can be used for detecting alterations in one or more markers genes.
[0108] In certain embodiments, detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195, 4,683,202 and 5,854,033), such as real-time PCR, COLD-PCR (Li et al. (2008) Nat. Med. 14:579), anchor PCR, recursive PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. (1988) Science 241 : 1077; Prodromou and Pearl (1992) Protein Eng. 5:827; and Nakazawa et al. (1994) Proc. Natl. Acad. Sci. USA 91 :360), the latter of which can be particularly useful for detecting point mutations in a marker gene (see Abravaya et al. (1995) Nucleic Acids Res. 23:675). This method can include the steps of collecting a sample of cell free bodily fluid from a subject, isolating nucleic acid (e.g., genomic, mRNA or both) from the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to a marker gene under conditions such that hybridization and amplification of the marker gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
[0109] Alternative amplification methods include: self sustained sequence replication (Guatelli et al, (1990) Proc. Natl. Acad. Sci. USA 87: 1874), transcriptional amplification system (Kwoh et al., (1989) Proc. Natl. Acad. Sci. USA 86:1173), Q Beta Replicase (Lizardi et al. (1988) Bio-Technology 6: 1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.
[0110] In an alternative embodiment, mutations in one or more markers genes from a sample can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, optionally amplified, digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, for example, U.S. Pat. No. 5,498,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
[0111] In other embodiments, genetic mutations in one or more of the markers described herein can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high density arrays containing hundreds or thousands of oligonucleotides probes (Cronin et al. (1996) Human Mutation 7: 244; Kozal et al. (1996) Nature Medicine 2:753). For example, genetic mutations in a marker nucleic acid can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin, M. T. et al. supra. Briefly, a first
hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations. This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
[0112] In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence a marker gene and detect mutations by comparing the sequence of the sample marker gene with the corresponding wild-type (control) sequence. Examples of sequencing reactions include those based on techniques developed by Maxam and Gilbert ((1977) Proc. Natl. Acad. Sci. USA 74:560) or Sanger ((1977) Proc. Natl. Acad. Sci. USA 74:5463). It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays ((1995)
Biotechniques 19:448), including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen et al. (1996) Adv. Chromatogr. 36: 127-162; and Griffin et al. (1993) Appl. Biochem. Biotechnol. 38:147).
[0113] Other methods for detecting mutations in a marker gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al. (1985) Science 230:1242). In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing the wild- type marker sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as which will exist due to base pair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S 1 nuclease to enzymatically digesting the mismatched regions. In other
embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, for example, Cotton et al. (1988) Proc. Natl. Acad. Sci. USA 85:4397; Saleeba et al. (1992) Methods Enzymol. 217:286. In one embodiment, the control DNA or RNA can be labeled for detection.
[0114] In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in marker cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. (1994) Carcinogenesis 15: 1657). According to an exemplary embodiment, a probe based on a marker sequence, e.g., a wild-type marker sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, for example, U.S. Patent No. 5,459,039.
[0115] In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in marker genes. For example, single strand conformation polymorphism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita et al. (1989) Proc. Natl. Acad. Sci. USA 86:2766, see also Cotton (1993) Mutat. Res. 285: 125; and Hayashi (1992) Genet. Anal. Tech. Appl. 9:73). Single-stranded DNA fragments of sample and control marker nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In one embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet. 7:5).
[0116] In yet another embodiment the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. (1985) Nature 313:495). When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys. Chem. 265: 12753).
[0117] Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324: 163; Saiki et al. (1989) Proc. Natl. Acad. Sci. USA 86:6230). Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the
oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
[0118] Alternatively, allele specific amplification technology which depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al. (1989) Nucl. Acids Res. 17:2437) or at the extreme 3 ' end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner (1993) Tibtech 11 :238). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection (Gasparini et al. (1992) Mol. Cell Probes 6:1). It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification (Barany (1991) Proc. Natl. Acad. Sci. USA 88: 189). In such cases, ligation will occur only if there is a perfect match at the 3 ' end of the 5 ' sequence making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.
[0119] An exemplary method for detecting the presence or absence of an analyte (e.g. , DNA, RNA, protein, polypeptide, or the like) corresponding to a marker of the invention in a biological sample involves obtaining a bodily fluid sample (e.g. , blood) from a test subject and contacting the bodily fluid sample with a compound or an agent capable of detecting one or more markers. Detection methods described herein can be used to detect one or more markers in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of a polypeptide corresponding to a marker of the invention include enzyme linked immunosorbent assays (ELISAs), Western blots,
immunoprecipitations and immunofluorescence. In vitro techniques for detection of genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of a polypeptide corresponding to a marker of the invention include introducing into a subject a labeled antibody directed against the polypeptide. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques. Because each marker is also an analyte, any method described herein to detect the presence or absence of a marker can also be used to detect the presence or absence of an analyte.
[0120] The markers useful in the methods of the invention can include any mutation in any one of the markers. Mutation sites and sequences can be identified, for example, by databases or repositories of such information, e.g., The Human Gene Mutation Database (www.hgmd.cf.ac.uk), the Single Nucleotide Polymorphism Database (dbSNP, www.ncbi.nlm.nih.gov/projects/SNP), and the Online Mendelian Inheritance in Man (OMIM) website
(www.ncbi.nlm.nih.gov/omim).
[0121] The present invention also provides kits that comprise marker detection agents that detect at least one or more of the head and neck cancer markers described herein.
[0122] The present invention also provides methods of treating or preventing head and neck cancer in a subject comprising administering to said subject an agent that modulates the activity or expression or disrupts the function of at least one or more of the markers of this invention.
[0123] The one or more markers identified by this invention (e.g., markers in Tables 1 and 2) may be used in the treatment of head and neck cancer. For example, a marker (e.g., a protein or gene) identified by the present invention may be used as a molecular target for a therapeutic agent. A marker identified by the invention also may be used in any of the other methods of the invention, e.g., for monitoring the progression or regression of a disease or condition. In certain embodiments, the one or more markers identified by the methods of this invention may have therapeutic potential. For example, if a marker is identified as being up- regulated (or down-regulated), see, for example, the up-regulated (or down- regulated) markers in Tables 1 and 2, or activated (or inhibited) in phagocytic cells from a subject having head and neck cancer, a compound or an agent that is capable of down-regulating (or up-regulating) or inhibiting (or activating) said marker may be useful in treating head and neck cancer. Similarly, a gene protein expression level, a protein expression level, or a combination thereof may be useful in this aspect of the invention.
[0124] Unless otherwise defined herein, scientific and technical terms used in this application shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclature used in connection with, and techniques of, cell and tissue culture, molecular biology, cell and cancer biology, neurobiology, neurochemistry, virology, immunology, microbiology, pharmacology, genetics and protein and nucleic acid chemistry, described herein, are those well known and commonly used in the art.
[0125] All of the above, and any other publications, patents and published patent applications referred to in this application are specifically incorporated by reference herein. In case of conflict, the present specification, including its specific definitions, will control.
[0126] 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 components) or group of integers (or components), but not the exclusion of any other integer (or components) or group of integers (or
components).
[0127] The singular forms "a," "an," and "the" include the plurals unless the context clearly dictates otherwise.
[0128] The term "including" is used to mean "including but not limited to." "Including" and "including but not limited to" are used interchangeably.
[0129] It is to be understood that the embodiments of the present invention which have been described are merely illustrative of some of the applications of the principles of the present invention. Numerous modifications may be made by those skilled in the art based upon the teachings presented herein without departing from the true spirit and scope of the invention.
[0130] The following examples are set forth as being representative of the present invention. These examples are not to be construed as limiting the scope of the invention as these and other equivalent embodiments will be apparent in view of the present disclosure and accompanying claims.
[0131] Examples
[0132] Example 1: Microarray analysis of head and neck cancer patients
[0133] Study Population
[0134] Blood samples were collected from eight head and neck cancer patients and eight blood donors without head and neck cancer. Approximately 10 ml of blood was collected from each patient into purple top blood collection EDTA tubes (BD Biosciences, CA). Within 3 hours, macrophages, neutrophils and T cells were isolated from each blood sample and total RNA was extracted and purified on the same day. The healthy control blood samples were obtained from apheresis collars of anonymous platelet donors. Gender determination of the blood donors was performed by PCR using two sets of primers, SRY primers (Forward: 5 -CAG TGT GAA ACG GGA GAA AAC AG-3'; Reverse: 5*-ACT TCG CTG CAG AGT ACC GAA G-3') amplifying a 336 bp fragment on Y chromosome and AR6 primers (Forward: 5*-CAA TCA GAG ACA TTC CCT CTG G-3*; Reverse: 5'- AGT GGT CCT CTC TGA ATC TC-3*) amplifying a 267 bp fragment on X chromosome (males have both fragments amplified; females have only one). The PCR (36 cycles) was done under the conditions of 95 °C for 45 seconds, 56 °C for 45 seconds, and 72 °C for 45 seconds.
[0135] Isolation of Macrophages (M), Neutrophils (N), and T Cells (TC) from Whole Blood
[0136] 7 mL of IX PBS containing 2% FBS and 2 mM EDTA were added to approximately 5 mL of whole blood, the sample centrifuged (2,000 RPM, 10 minutes at 20°C). The buffy coat was removed and centrifuged (2,000 RPM, 10 minutes at 20°C). The cell pellet was then suspended in ~1 mL of PBS and transferred to a 1.5 mL microfuge tube. Next, macrophages, neutrophils, and T cells were isolated using magnetic beads coated with antibodies specific to each of the three cell types (positive cell depletion). Cells were separated from the buffy coat always in the following sequence: 1) macrophages; 2) neutrophils; and 3) T cells (changing the order did not alter the RNA yield and quality). The freshly isolated white blood cell samples (in ~1 mL PBS) were incubated (25 min, 4°C, constant shaking) first with anti-monocyte coated Dynabeads® (CD 14 - Cat. No. 11149D, Life Technologies), then with anti-neutrophil coated Dynabeads® (CD 15 - Cat. No. 11137D, Life Technologies), and finally with anti- T cell coated
Dynabeads® (CD2 Pan T - Cat. No. 11159D, Life Technologies). Following each incubation, the bead-bound cells were separated using a magnet. The purity of these white blood cell subpopulations, which per manufacturer's specifications (Life Technologies) is >95%, was evident from the unique gene expression pattern obtained (cluster analysis). As soon as each white blood cell subpopulation was isolated, the magnetic bead bound cells were washed with lx PBS and lysed in Trizol®. The fractionation and subsequent lysis of all the three types of cells were completed in less than 2 hours after the isolation of the buffy coat.
[0137] Total RNA Isolation
[0138] Total RNA was extracted from cells and tissues with Trizol® and the Pure-Link RNA isolation kit (Cat. # 12183018A, Life Technologies). The quantity and purity of the RNA samples were determined on a Bioanalyzer 2100 (Agilent Technologies) and the Degradometer software (version 1.41). In general, the RIN and 28s/18s ratios were always found to be in the satisfactory range, >9 and >1.9, respectively.
[0139] Whole Genome Microarray Data Analysis
[0140] Total RNA from macrophages, neutrophils, T cells, tumor tissue (TT), and "normal" head and neck tissue (NT) of head and neck cancer patients, and from macrophages, neutrophils, and T cells extracted from healthy male blood donors were used in gene expression profiling. Biotinylated cDNA probes were prepared from 100 ng of each RNA sample, fragmented, and hybridized with Human Gene 1.0 ST chip (Affymetrix). Array signals of fluorescence were scanned, captured and recorded as CEL files. All the processing and analysis of the data were done using R 49 and Bioconductor software packages. To obtain the log2 transformed expression levels, the raw data files obtained in CEL file format were pre-processed using the oligo package and the RMA (robust multichip average algorithm) routine to background correct, quantile normalize and summarize at the core level.
[0141] Example 2: Statistical analysis of microarray data
[0142] Working with microarray data can be challenging because large numbers of genes can increase the likelihood of false positives, while a small number of samples can lead to overfitting. These issues can be overcome by using statistical methods to reduce the false rate of positives and using independent training and test data sets (e.g., cross-validation) to avoid overfitting. In particular, instead of using a "typical" 5% significance level, the false discovery rate (FDR) can be controlled to ensure that only 5% of the genes that are discovered are false positives, and Empirical Bayesian estimates can be used to improve test statistics. [0143] Because an overfit model will perform poorly on an independent test set, a good test of the fit of a model is how well is performs on an independent test set. For small sample sizes, splitting data into test and training sets may leave too small of a data set for good training. This issue can be solved by using cross-validation, which splits the data into K- folds, trains the method on K-l of the folds, and tests the method on the last fold. Figure 1 depicts a diagram of a three-fold cross validation, wherein the diagnostic accuracy is averaged from the three splits. The ideal split for cross-validation is 10-fold for accurate and precise estimates of diagnostic accuracy. In a 10-fold cross validation, however, there are more than 10 splits because there are many choices for which data points go into the folds. For example, with the microarray data collected as described above, there are
50,979,600 ways to form 90% training/10% testing data sets.
[0144] The Empirical Bayesian method was used as follows:
1. The differential gene expression (DE) of phagocytes (macrophages or neutrophils) vs. T cells was calculated for each gene. DE is expressed as the log of the ratio of phagocyte to T cell expression: DE = log(GEP/GETc), where GEP is phagocyte gene expression and GETC is T cell gene expression.
2. The mean DE was compared in cancer and control patients with a two- sample t-test. Empirical Bayes estimates of the test statistics "shrink" these toward zero.
3. Calculate a diagnostic signature with K genes:
Figure imgf000069_0001
i=i
If S>0, then the patient was diagnosed with cancer.
4. The number of genes K to include in the signature was determined by comparing misclassification rates in independent test sets with cross-validation.
Errors were calculated using an average of 1-senstivity and 1 -specificity, and the cross-validated error was used to select markers.
[0145] Using the above methods, the markers associated with head and neck cancer in macrophages vs. T cells (Table 1) and the markers associated with head and neck cancer in neutrophils vs. T cells (Table 2) were identified. Of these, specific signatures of nine markers (for macrophages) and six markers (for neutrophils) also were identified that give especially high sensitivity and specificity. For example, a nine-marker signature (FiNC-Macro 1-9) from macrophages has a sensitivity of 95.5% and a specificity of 98.8%. The nine identified were
1. NR4A3
2. PRDM1
3. CCR 4L
4. ICOSLG
5. GPR183
6. NR4A1
7. CLN8
8. P2RY10
9. TNFAIP3
[0146] Figure 2 shows a summary of the head and neck cancer markers identified from macrophages and from neutrophils, as compared to T cells from the same individuals, for the HNC-Macro 1-100 and HNC-Neutro 1-100 markers.
Specifically, average error, sensitivity, and specificity values are given for a nine marker panel from FiNC-Macro 1-100 (FiNC-Macro 1-9), a six marker panel from HNC-Neutro 1-100 (HNC-Neutro 1-6). The six HNC-Neutro markers include: SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
[0147] Figure 3 demonstrates the power of a paired within- subject (phagocyte to non-phagocyte) comparison to detect head and neck cancer as compared to phagocytes not paired with T cell data for comparison. The paired approach (comparing macrophage or neutrophils to T cell expression) is better than the phagocyte gene expression alone.
[0148] Example 3: Additional validation
[0149] Additional validation is performed by validating the gene signature on a new data set of ~50 head and neck cancer cases and 50 controls. Final validation is performed by estimating the sensitivity and specificity of the final gene signature on a large sample. For example, 195 cases and 195 controls can be used to estimate a sensitivity/specificity of at least 97.5% with a 95% margin of error no more than 5%. A challenge in designing a final validation study is that although cancer patients are pure, controls may have up to 20% false negatives. A statistical issue is that, while estimating sensitivity is not a problem, specificity has an upper bound of 80%. The solution is to purify the control set of patients. A purification method uses secondary screening of all controls, wherein three methylated gene marker tests are used to purify the control test set:
1. GST-Pi (sensitivity = 95%, specificity =85%)
2. RAR-2b (sensitivity = 95%, specificity =48%)
3. APC (sensitivity = 95%, specificity =50%)
in men with two serial negative biopsies. A second purification method is depicted in Figure 4, and a comparison of purification methods is shown in Figure 5.
Table 1. Head and Neck Cancer (HNC)-Macro Markers
(DR = down-regulated; UR= up-regulated)
Figure imgf000072_0001
HNC-Macro Transcript Gene Name Cancer Mean Control Mean Pattern Cluster ID
28 8010778 CSNK1D 2.680154683 5.642429036 Cancer DR
29 8063382 SNAI1 1.247181424 2.622340984 Cancer DR
30 8114511 MGC29506 0.906642744 1.145372798 Cancer DR
31 8075316 OSM 1.343709985 4.288513257 Cancer DR
32 8013068 PLD6 0.648663556 0.968522866 Cancer DR
33 7904465 HIST2H2BA 0.859351662 0.646585111 Cancer UR
34 7955578 GRASP 0.935585573 1.898020047 Cancer DR
35 8145244 TNFRSF10C 4.678293122 1.636470575 Cancer UR
36 7948908 SNORD26 0.364396063 0.146903833 Cancer UR
37 7952036 MPZL3 1.066300046 0.510432796 Cancer UR
38 7926807 PDSS1 1.810161132 0.934438856 Cancer UR
39 8095728 EREG 2.332232946 9.289917749 Cancer DR
40 7961524 ERP27 0.604222101 0.28544135 Cancer UR
41 8130499 DYNLT1 1.498869141 0.809192399 Cancer UR
42 7898693 ALPL 6.066605704 1.217724578 Cancer UR
43 7963760 NFE2 8.048464113 3.411350058 Cancer UR
44 8009502 KCNJ2 6.481285901 1.335377626 Cancer UR
45 8015133 KRT23 4.410727028 1.38015993 Cancer UR
46 8144279 CLN8 1.153905598 1.765163723 Cancer DR
47 8008795 C17orf71 1.408388834 0.853909752 Cancer UR
48 8075052 LRP5L 1.13839651 1.677416605 Cancer DR
49 8161701 TMEM2 1.197717741 0.583857466 Cancer UR
50 8116484 MGAT1 1.457482638 2.461800143 Cancer DR
51 8046124 DHRS9 3.630396735 1.892608925 Cancer UR
52 7948904 SNORD28 0.323404567 0.12953683 Cancer UR
53 7928308 DDIT4 0.380751555 0.992065875 Cancer DR
54 8105229 PELO 1.086584949 0.68849556 Cancer UR
55 8174361 TSC22D3 0.93836709 0.603416748 Cancer UR
56 7945663 LOC402778 1.397846337 1.98533205 Cancer DR HNC-Macro Transcript Gene Name Cancer Mean Control Mean Pattern Cluster ID
57 7968035 SPATA13 0.577404665 1.105643134 Cancer DR
58 8016540 PHOSPHOl 3.22860536 1.183964537 Cancer UR
59 8101992 SLC39A8 0.479178554 0.236077843 Cancer UR
60 8113356 GPAM 1.147292709 0.401284591 Cancer UR
61 8031939 ZNF584 0.782152279 1.095717273 Cancer DR
62 8034837 DNAJB1 0.565411784 0.318274105 Cancer UR
63 7898655 CDA 24.4383885 11.26683891 Cancer UR
64 7935421 FRAT2 2.681524883 1.539424633 Cancer UR
65 8092691 BCL6 7.531381863 3.712937052 Cancer UR
66 8018209 NAT9 1.071090681 1.87926894 Cancer DR
67 8003068 MPHOSPH6 0.954119316 1.725428278 Cancer DR
68 8146159 AP3M2 0.229343977 0.133413067 Cancer UR
69 7937335 IFITM1 0.396140095 0.125735987 Cancer UR
70 7974870 SNAP 1.131562239 1.869828249 Cancer DR
71 7961371 DUSP16 0.252251134 0.140251452 Cancer UR
72 8024518 ZNF555 0.960954856 1.445381736 Cancer DR
73 8132399 C7or06 0.483849886 0.295171863 Cancer UR
74 8083494 MME 5.58032934 1.035844153 Cancer UR
75 7902227 GADD45A 1.099884499 0.730728937 Cancer UR
76 8119898 VEGFA 3.331404529 7.571005261 Cancer DR
77 8109086 ADRB2 1.103982342 0.654871142 Cancer UR
78 7952126 RPL23AP64 0.852575862 1.213790774 Cancer DR
79 8126839 TNFRSF21 1.04364885 2.062951875 Cancer DR
80 7947917 KBTBD4 0.705599895 0.516256819 Cancer UR
81 8156826 TGFBR1 0.830409512 1.21250039 Cancer DR
82 7914184 PTAFR 4.344834176 2.460101693 Cancer UR
83 7906400 IFI16 1.008070795 0.706673006 Cancer UR
84 7898623 UBXN10 0.796334518 1.177014512 Cancer DR
85 7903592 KIAA1324 0.817230793 0.147091728 Cancer UR HNC-Macro Transcript Gene Name Cancer Mean Control Mean Pattern Cluster ID
86 7996100 GPR97 3.950747888 1.060295116 Cancer UR
87 8072328 SEC14L2 0.770722224 0.402689014 Cancer UR
88 7942832 Cl lorfS 3.274797863 1.775613408 Cancer UR
89 8044766 INSIG2 0.86796442 0.639550862 Cancer UR
90 8038815 LIM2 0.902069895 1.170353321 Cancer DR
91 8121277 AIM1 1.014548905 0.592201197 Cancer UR
92 8137257 GIMAP5 0.280198715 0.141348859 Cancer UR
93 8039766 ZNF837 0.882027802 1.191574697 Cancer DR
94 7926836 RAB18 1.354116821 0.880314428 Cancer UR
95 8095986 ANXA3 4.332741626 0.961208097 Cancer UR
96 7935660 DNMBP 1.815175659 3.068331691 Cancer DR
97 8137240 GIMAP7 0.505696085 0.233371366 Cancer UR
98 8019939 TGIF1 0.948812106 1.341688192 Cancer DR
99 7927775 NRBF2 2.179436854 1.277314305 Cancer UR
100 7975203 MPP5 0.791788689 1.090273574 Cancer DR
Table 2. Head and Neck Cancer (HNC)-Neutro Markers
(DR = down-regulated; UR= up-regulated)
Figure imgf000076_0001
HNC-Neutro Transcript Gene Name Cancer Control Mean Pattern Cluster ID Mean
28 8154359 RPL18A 0.220289746 0.692386399 Cancer DR
29 7990965 RPS17 0.080312588 0.361830845 Cancer DR
30 7990916 RPS17 0.080312588 0.361830845 Cancer DR
31 7966996 RPLPO 0.103369275 0.375397366 Cancer DR
32 8180410 Unknown 0.165331625 0.598216703 Cancer DR
33 8091806 RPL23A 0.157667504 0.599130606 Cancer DR
34 7948679 EEF1G 0.145228665 0.54330531 Cancer DR
35 8179519 HLA-DPB1 0.087377773 1.178062313 Cancer DR
36 8059222 DNPEP 0.310072475 0.575887828 Cancer DR
37 8180297 Unknown 0.084078262 0.482251383 Cancer DR
38 8133721 HSPB1 0.283521593 0.864458238 Cancer DR
39 7937802 CD81 0.062728225 0.252465514 Cancer DR
40 7948667 AHNAK 0.059941241 0.505720697 Cancer DR
41 8012464 LOC100128288 0.509259275 1.012811377 Cancer DR
42 8034416 RPL10 0.1305109 0.425975568 Cancer DR
43 8177669 SMA5 0.25098158 0.450737883 Cancer DR
44 8180086 HLA-DMA 0.323867466 2.134934323 Cancer DR
45 8178884 HLA-DMA 0.323867466 2.134934323 Cancer DR
46 8107470 PTMA 0.270189984 0.785811795 Cancer DR
47 8125537 HLA-DMA 0.327821118 2.084216901 Cancer DR
48 8112564 GUSBP3 0.260427769 0.444812785 Cancer DR
49 7946812 RPS13 0.11713807 0.513285234 Cancer DR
50 8178220 HLA-DPB1 0.085063774 1.002289043 Cancer DR
51 7986323 GLTSCR2 0.099005816 0.397706799 Cancer DR
52 7920123 S100A1 0.065476841 0.825667054 Cancer DR
53 8158714 EXOSC2 0.111363025 0.347602396 Cancer DR
54 8085026 RPL35A 0.133431594 0.434831338 Cancer DR
55 7899160 CD52 0.08212946 0.613244823 Cancer DR
56 8112560 SMA5 0.246259487 0.441979303 Cancer DR HNC-Neutro Transcript Gene Name Cancer Control Mean Pattern Cluster ID Mean
57 8180355 Unknown 0.170389269 0.553935753 Cancer DR
58 8013348 RPS2 0.202567803 0.529896514 Cancer DR
59 8112469 GUSBP3 0.2616985 0.445030893 Cancer DR
60 7998655 RPS2 0.204530799 0.527597121 Cancer DR
61 8154727 LOC138412 0.147403774 0.59298426 Cancer DR
62 8105991 GUSBP3 0.247386942 0.438576556 Cancer DR
63 7920725 SCAMP3 0.387977245 0.639026348 Cancer DR
64 8005943 RPL23A 0.143752364 0.541805743 Cancer DR
65 8000284 GGA2 0.17320102 0.578480516 Cancer DR
66 7936100 CALHM2 0.384878562 0.972201149 Cancer DR
67 7973056 APEX1 0.079756496 0.392519161 Cancer DR
68 7901601 MRPL37 0.187070078 0.565051539 Cancer DR
69 8036602 ECH1 0.135446742 0.539329935 Cancer DR
70 8109750 RPLPO 0.038000029 0.3010213 Cancer DR
71 8155849 ANXA1 0.428783765 0.824136121 Cancer DR
72 8180100 HLA-DPA1 0.100245039 1.320933362 Cancer DR
73 8164373 CIZ1 0.403354804 0.681161422 Cancer DR
74 8104621 GUSBP1 0.244790421 0.436104228 Cancer DR
75 8066964 TMEM189 0.295758103 0.729715645 Cancer DR
76 8152215 KLF10 0.587477731 2.30511617 Cancer DR
77 8079334 LIMD1 0.322722967 0.832749853 Cancer DR
78 8009561 RPL38 0.242618725 0.577387836 Cancer DR
79 7996677 NUTF2 0.363473911 0.764111119 Cancer DR
80 8176360 CD99 0.096001869 0.319705642 Cancer DR
81 8165794 CD99 0.096001869 0.319705642 Cancer DR
82 7956152 PA2G4 0.181807121 0.437956091 Cancer DR
83 8092457 ALG3 0.174491575 0.68243038 Cancer DR
84 8043197 VAMP 8 0.14950648 0.787069469 Cancer DR
85 7986685 DEXI 0.152019315 0.324214931 Cancer DR HNC-Neutro Transcript Gene Name Cancer Control Mean Pattern Cluster ID Mean
86 7982185 DEXI 0.152019315 0.324214931 Cancer DR
87 7952129 RPS25 0.128100213 0.417013041 Cancer DR
88 8007441 RPL27 0.287479466 0.675608661 Cancer DR
89 8014454 MY019 0.397985849 0.558046525 Cancer DR
90 8158446 NUP188 0.148057449 0.35409186 Cancer DR
91 8146649 MTFR1 0.397482411 0.785051201 Cancer DR
92 8115158 RPS14 0.172845774 0.490931474 Cancer DR
93 8024299 RPS15 0.168124325 0.545126073 Cancer DR
94 8178891 HLA-DPA1 0.112019416 1.319233515 Cancer DR
95 8125556 HLA-DPA1 0.112019416 1.319233515 Cancer DR
96 7961022 PTMA 0.29114269 0.731385932 Cancer DR
97 7963750 CBX5 0.070817947 0.202669582 Cancer DR
98 8152764 MTSS1 0.191403501 1.370902827 Cancer DR
99 8076511 RPL5 0.09775886 0.440315484 Cancer DR
100 7994565 RRN3 0.117822 0.351763164 Cancer DR

Claims

What is Claimed is:
1. A method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells; b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b),
wherein the identified difference indicates that the subject has said head and neck cancer.
2. A method for assessing the risk of developing head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells; b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b),
wherein the identified difference indicates that the subject has a risk of developing said head and neck cancer.
3. A method for prognosing or aiding in the prognosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells; b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b), wherein the identified difference is indicative of the prognosis of said head and neck cancer in the subject.
4. A method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before the treatment;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before the treatment;
c) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment;
e) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after the treatment;
f) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) is indicative of the efficacy of the treatment for said head and neck cancer in the subject.
5. A method for monitoring the progression or regression of head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells at a first time point;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells at the first time point;
c) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b); d) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells at a second time point;
e) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells at the second time point;
f) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) is indicative of the progression or regression of said head and neck cancer in the subject.
6. A method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before administering the compound to the subject;
b) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before administering the compound to the subject;
c) identifying a first difference between the measured levels of the one or more selected HNC-Macro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the administration of the compound;
e) measuring the levels of the one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after the administration of the compound;
f) identifying a second difference between the measured levels of the one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference, wherein the difference identified in g) indicates that the compound is capable of ameliorating or treating said head and neck cancer in the subject.
7. A method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells; b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b),
wherein the identified difference indicates that the subject has said head and neck cancer.
8. A method for assessing the risk of developing head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells; b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells; and
c) identifying a difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b),
wherein the identified difference indicates that the subject has a risk of developing said head and neck cancer.
9. A method for prognosing or aiding in the prognosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells; b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells; and c) identifying a difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b),
wherein the identified difference is indicative of the prognosis of said head and neck cancer in the subject.
10. A method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before the treatment;
b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before the treatment;
c) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment;
e) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after the treatment;
f) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) is indicative of the efficacy of the treatment for said head and neck cancer in the subject.
11. A method for monitoring the progression or regression of head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells at a first time point;
b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells at the first time point; c) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells at a second time point;
e) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells at the second time point;
f) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in steps d) and e); and
g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) is indicative of the progression or regression of said head and neck cancer in the subject.
12. A method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising:
a) measuring the levels of one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before administering the compound to the subject;
b) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before administering the compound to the subject;
c) identifying a first difference between the measured levels of the one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the administration of the compound;
e) measuring the levels of the one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after the administration of the compound;
f) identifying a second difference between the measured levels of the one or more selected HNC-Neutro markers in steps d) and e); and g) identifying a difference between the first difference and the second difference,
wherein the difference identified in g) indicates that the compound is capable of ameliorating or treating said head and neck cancer in the subject.
13. A method for diagnosing or aiding in the diagnosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells;
c) identifying a difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
d) identifying a difference between the measured levels or activities the at least one or more selected HNC-Neutro markers in steps a) and b);
wherein the differences identified in c) and d) indicate that the subject has said head and neck cancer.
14. A method for assessing the risk of developing head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells; b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells;
c) identifying a difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
d) identifying a difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
wherein the differences identified in c) and d) indicate that the subject has a risk of developing said head and neck cancer.
15. A method for prognosing or aiding in the prognosis of head and neck cancer in a subject, the method comprising the steps of:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells;
c) identifying a difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
d) identifying a difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
wherein the differences identified in c) and d) are indicative of the prognosis of said head and neck cancer in the subject.
16. A method for assessing the efficacy of a treatment for head and neck cancer in a subject comprising: a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before the treatment, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before the treatment;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before the treatment; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before the treatment; c) identifying a first difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
identifying a second difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's macrophage cells after the treatment, and measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after the treatment;
e) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after the treatment; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after the treatment; f) identifying a third difference between the measured levels of the at least one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a fourth difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps d) and e);
h) identifying a difference between the first and second differences; and i) identifying a difference between the third and fourth differences, wherein the differences identified in h) and i) are indicative of the efficacy of the treatment for said head and neck cancer in the subject.
17. A method for monitoring the progression or regression of head and neck cancer in a subject comprising:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells at a first time point, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells at the first time point;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells at the first time point; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells at the first time point; c) identifying a first difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
identifying a second difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's macrophage cells at a second time point, and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells at the second time point; e) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells at the second time point; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells at the second time point;
f) identifying a third difference between the measured levels of the at least one or more selected HNC-Macro markers in steps d) and e); and g) identifying a fourth difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps d) and e);
h) identifying a difference between the first and second differences; and i) identifying a difference between the third and fourth differences, wherein the differences identified in h) and i) are indicative of the progression or regression of said head and neck cancer in the subject.
18. A method for identifying a compound capable of ameliorating or treating head and neck cancer in a subject comprising:
a) measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 in a population of the subject's macrophage cells before administering the compound to the subject, and
measuring the levels of at least one or more markers selected from the group consisting of HNC-Neutro 1-100 in a population of the subject's neutrophil cells before administering the compound to the subject;
b) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells before administering the compound to the subject; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells before administering the compound to the subject;
c) identifying a first difference between the measured levels of the at least one or more selected HNC-Macro markers in steps a) and b); and
identifying a second difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps a) and b);
d) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's macrophage cells after administering the compound to the subject, and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's neutrophil cells after administering the compound to the subject; e) measuring the levels of the at least one or more selected HNC-Macro markers in a population of the subject's non-phagocytic cells after administering the compound to the subject; and
measuring the levels of the at least one or more selected HNC-Neutro markers in a population of the subject's non-phagocytic cells after administering the compound to the subject;
f) identifying a third difference between the measured levels of the at least one or more selected HNC-Macro markers in steps d) and e); and
g) identifying a fourth difference between the measured levels of the at least one or more selected HNC-Neutro markers in steps d) and e);
h) identifying a difference between the first and second differences; and i) identifying a difference between the third and fourth differences, wherein the differences identified in h) and i) indicate that the compound is capable of ameliorating or treating said head and neck cancer in the subject.
19. The method of any one of claims 1-18, further comprising measuring at least one standard parameter associated with said head and neck cancer.
20. The method of claim 19, wherein the standard parameter is selected from the group consisting of tumor stage, tumor grade, tumor size, tumor visual characteristics, tumor growth, tumor thickness, tumor progression, tumor metastasis tumor distribution within the body, odor, molecular pathology, genomics, or tumor angiograms.
21. The method of any one of claims 13-18, wherein the selected HNC-Macro markers and the selected HNC-Neutro markers are measured from the same population of non-phagocytic cells in steps b) or e).
22. The method of any one of claims 13-18, wherein the selected HNC-Macro markers and the selected HNC-Neutro are from different populations of non- phagocytic cells in steps b) or e).
23. The method of any one of claims 1-6 and 13-18, wherein at least two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or fifteen markers are selected from HNC-Macro 1-100.
24. The method of any one of claims 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of NR4A3, PRDM1, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
25. The method of any one of claims 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of HNC-Macro 8, 9, 10, 11, 12, 13, 15, 18, 22, 24, 27, 33, 35, 36, 37, 38, 40, 41, 42, 43, 44, 45, 47, 49, 51, 52, 54, 55, 58, 59, 60, 62, 63, 64, 65, 68, 69, 71, 73, 74, 75, 77, 80, 82, 83, 85, 86, 87, 88, 89, 91, 92, 94, 95, 97, and 99 and wherein the selected HNC-Macro markers are up-regulated or activated in the macrophage cells compared to the non-phagocytic cells.
26. The method of any one of claims 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of P2RY10 and TNFAIP3 and wherein the selected HNC-Macro markers are up-regulated or activated in the macrophage cells compared to the non-phagocytic cells.
27. The method of any one of claims 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of HNC-Macro 1, 2, 3, 4, 5, 6, 7, 14, 16, 17, 19, 20, 21, 23, 25, 26, 28, 29, 30, 31, 32, 34, 39, 46, 48, 50, 53, 56, 57, 61, 66, 67, 70, 72, 76, 78, 79, 81, 84, 90, 93, 96, 98, and 100 and wherein the selected HNC-Macro markers are down- regulated or inhibited in the macrophage cells compared to the non-phagocytic cells.
28. The method of any one of claims 1-6 and 13-18, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of NR4A3, PRDMl, CCRN4L, ICOSLG, GPR183, NR4A1, and CLN8 and wherein the selected HNC-Macro markers are down-regulated or inhibited in the macrophage cells compared to the non-phagocytic cells.
29. The method of any one of claims 7-18, wherein at least two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or fifteen markers are selected from HNC-Neutro 1-100.
30. The method of any one of claims 7-18, wherein the selected HNC-Neutro markers comprise one or more HNC-Neutro markers selected from the group consisting of SLC25A6, RPS4X, RPL3, RPL18, and OBFC2B.
31. The method of any one of claims 7-18, wherein the selected HNC-Neutro markers comprise one or more markers selected from the group consisting of HNC-Neutro 1-100 and wherein the selected HNC-Neutro markers are down- regulated or inhibited in the neutrophil cells compared to the non-phagocytic cells.
32. The method of any one of claims 7-18, wherein the selected HNC-Neutro markers comprise one or more markers selected from the group consisting of RPS4X, SLC25A6, RPL3, RPL18, and OBFC2B and wherein the selected HNC- Neutro markers are down-regulated or inhibited in the neutrophil cells compared to the non-phagocytic cells.
33. The method of any one of claims 1-6 and 13-18, further comprising lysing the macrophage cells and the non-phagocytic cells before a).
34. The method of any one of claims 1-6 and 13-18, further comprising extracting the cellular contents from the macrophage cells and the non-phagocytic cells before a).
35. The method of any one of claims 7-18, further comprising lysing the neutrophil cells and the non-phagocytic cells before a).
36. The method of any one of claims 7-18, further comprising extracting the cellular contents from the neutrophil cells and the non-phagocytic cells before a).
37. The method of claim 34, wherein the cellular contents of the macrophage cells comprise viable diseased cells, dead diseased cells, apoptotic diseased cells, circulating tumor cells, infectious agents, fetal cells, trophoblasts, or fragments thereof.
38. The method of claim 36, wherein the cellular contents of the neutrophil cells comprise viable diseased cells, dead diseased cells, apoptotic diseased cells, circulating tumor cells, infectious agents, fetal cells, trophoblasts, or fragments thereof.
39. The method of claim 34, wherein the selected one or more markers are present in the cellular contents of the macrophage cells.
40. The method of claim 34, wherein the selected one or more markers are not present in the cellular contents of the non-phagocytic cells.
41. The method of any one of claims 1-6 and 13-18, wherein the macrophage cells express the one or more selected HNC-Macro markers.
42. The method of claim 36, wherein the selected one or more markers are present in the cellular contents of the neutrophil cells.
43. The method of claim 36, wherein the selected one or more markers are not present in the cellular contents of the non-phagocytic cells.
44. The method of any one of claims 7-18, wherein the neutrophil cells express the one or more selected HNC-Neutro markers.
45. The method of any one of claims 1-18, wherein the non-phagocytic cells are T cells, B cells, null cells, basophils, or mixtures thereof.
46. The method of any one of claims 1-6 and 13-18, wherein the macrophage cells are isolated from a bodily fluid sample, tissues, or cells of the subject.
47. The method of any one of claims 7-18, wherein the neutrophil cells are isolated from a bodily fluid sample, tissues, or cells of the subject.
48. The method of any one of claims 1-18, wherein the non-phagocytic cells are isolated from a bodily fluid sample, tissues, or cells of the subject.
49. The method of any one of claims 46-48, wherein the bodily fluid sample is blood, urine, stool, saliva, lymph fluid, cerebrospinal fluid, synovial fluid, cystic fluid, ascites, pleural effusion, fluid obtained from a pregnant woman in the first trimester, fluid obtained from a pregnant woman in the second trimester, fluid obtained from a pregnant woman in the third trimester, maternal blood, amniotic fluid, chorionic villus sample, fluid from a preimplantation embryo, maternal urine, maternal saliva, placental sample, fetal blood, lavage and cervical vaginal fluid, interstitial fluid, or ocular fluid.
50. The method of any one of claims 1-6 and 13-18, wherein the macrophage cells are isolated using antibodies, using a ligand that binds to a molecular receptor expressed on the plasma membranes of white blood cells, or by flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, micro fluidics, magnetic separation technique, fluorescent-magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platforms, or a combination thereof.
51. The method of any one of claims 7-18, wherein the neutrophil cells are isolated using antibodies, using a ligand that binds to a molecular receptor expressed on the plasma membranes of white blood cells, or by flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, micro fluidics, magnetic separation technique, fluorescent-magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platforms, or a combination thereof.
52. The method of any one of claims 1-18, wherein the non-phagocytic cells are isolated using antibodies, using a ligand that binds to a molecular receptor expressed on the plasma membranes of white blood cells, or by flow cytometry, fluorescence activated cell sorting, filtration, gradient-based centrifugation, elution, micro fluidics, magnetic separation technique, fluorescent-magnetic separation technique, nanostructure, quantum dots, high throughput microscope-based platforms, or a combination thereof.
53. The method of any one of claims 1-6 and 13-18, wherein the macrophage cells are isolated using a product secreted by the macrophage cells.
54. The method of any one of claims 7-18, wherein the neutrophil cells are isolated by using a product secreted by the neutrophil cells.
55. The method of any one of claims 1-6 and 13-18, wherein the macrophage cells are isolated by using a cell surface target on the surface of macrophage cells.
56. The method of any one of claims 7-18, wherein the neutrophil cells are isolated by using a cell surface target on the surface of neutrophil cells.
57. The method of claim 55, wherein the target is expressed by the
macrophage cells.
58. The method of claim 55, wherein the target is not expressed by the macrophage cells.
59. The method of claim 56, wherein the target is expressed by the neutrophil cells.
60. The method of claim 56, wherein the target is not expressed by the neutrophil cells.
61. The method of any one of claims 55-60, wherein the target is a marker of said head and neck cancer.
62. The method of any one of claims 1-18, wherein the measured levels are gene expression levels.
63. The method of any one of the claims 1-18, wherein the measured levels are protein expression levels.
64. The method of any one of claim 1-18, wherein the levels or activities are measured by a qualitative assay, a quantitative assay, or a combination thereof.
65. The method of claim 64, wherein the quantitative assay uses sequencing, direct sequencing, RNA sequencing, whole transcriptome shotgun sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, whole- genome sequencing, sequencing by hybridization, pyrosequencing, capillary electrophoresis, gel electrophoresis, duplex sequencing, cycle sequencing, single- base extension sequencing, solid-phase sequencing, high-throughput sequencing, massively parallel signature sequencing, emulsion PCR, sequencing by reversible dye terminator, paired-end sequencing, near-term sequencing, exonuclease sequencing, sequencing by ligation, short-read sequencing, single-molecule sequencing, sequencing-by-synthesis, real-time sequencing, reverse-terminator sequencing, nanopore sequencing, 454 sequencing, Solexa Genome Analyzer sequencing, SOLiD® sequencing, MS-PET sequencing, mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI-TOF) mass
spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole-time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), polymerase chain reaction (PCR) analysis, quantitative PCR, real-time PCR, fluorescence assay, colorimetric assay, chemiluminescent assay, or a combination thereof.
66. The method of claim 62, wherein the gene expression levels are measured by polymerase chain reaction (PCR) analysis, sequencing analysis, electrophoretic analysis, restriction fragment length polymorphism (RFLP) analysis, Northern blot analysis, quantitative PCR, reverse-transcriptase-PCR analysis (RT-PCR), allele- specific oligonucleotide hybridization analysis, comparative genomic
hybridization, heteroduplex mobility assay (HMA), single strand conformational polymorphism (SSCP), denaturing gradient gel electrophisis (DGGE), RNAase mismatch analysis, mass spectrometry, tandem mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole- time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), surface plasmon resonance, Southern blot analysis, in situ hybridization, fluorescence in situ hybridization (FISH), chromogenic in situ hybridization (CISH), immunohistochemistry (IHC), microarray, comparative genomic hybridization, karyotyping, multiplex ligation-dependent probe amplification (MLPA), Quantitative Multiplex PCR of Short Fluorescent Fragments (QMPSF), microscopy, methylation specific PCR (MSP) assay, Hpall tiny fragment
Enrichment by Ligation-mediated PCR (HELP) assay, radioactive acetate labeling assays, colorimetric DNA acetylation assay, chromatin immunoprecipitation combined with microarray (ChlP-on-chip) assay, restriction landmark genomic scanning, Methylated DNA immunoprecipitation (MeDIP), molecular break light assay for DNA adenine methyltransferase activity, chromatographic separation, methylation-sensitive restriction enzyme analysis, bisulfite-driven conversion of non-methylated cytosine to uracil, methyl-binding PCR analysis, or a combination thereof.
67. The method of claim 62, wherein the gene expression levels are measured by a sequencing technique selected from the group consisting of direct sequencing, RNA sequencing, whole transcriptome shotgun sequencing, random shotgun sequencing, Sanger dideoxy termination sequencing, whole-genome sequencing, sequencing by hybridization, pyrosequencing, capillary electrophoresis, gel electrophoresis, duplex sequencing, cycle sequencing, single-base extension sequencing, solid-phase sequencing, high-throughput sequencing, massively parallel signature sequencing, emulsion PCR, sequencing by reversible dye terminator, paired-end sequencing, near-term sequencing, exonuclease sequencing, sequencing by ligation, short-read sequencing, single-molecule sequencing, sequencing-by-synthesis, real-time sequencing, reverse-terminator sequencing, nanopore sequencing, 454 sequencing, Solexa Genome Analyzer sequencing, SOLiD® sequencing, MS-PET sequencing, mass spectrometry, and a combination thereof.
68. The method of claim 63, wherein the protein expression levels are measured by an immunohistochemistry assay, an enzyme-linked immunosorbent assay (ELISA), in situ hybridization, chromatography, liquid chromatography, size exclusion chromatography, high performance liquid chromatography (HPLC), gas chromatography, mass spectrometry, tandem mass spectrometry, matrix assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry, electrospray ionization (ESI) mass spectrometry, surface-enhanced laser deorption/ionization-time of flight (SELDI-TOF) mass spectrometry, quadrupole- time of flight (Q-TOF) mass spectrometry, atmospheric pressure photoionization mass spectrometry (APPI-MS), Fourier transform mass spectrometry (FTMS), matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance (MALDI-FT-ICR) mass spectrometry, secondary ion mass spectrometry (SIMS), radioimmunoassays, microscopy, microfluidic chip-based assays, surface plasmon resonance, sequencing, Western blotting assay, or a combination thereof.
69. The method of any one of claims 1-68, wherein the subject is a mammal.
70. The method of claim 69, wherein the subject is a human.
71. The method of any one of claims 1-18, wherein the difference is greater than a 1-fold difference.
72. The method of claim 71 , wherein the difference is at least 1.05 -fold, 1.1- fold, 1.2-fold, 1.3-fold, 1.4-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 4-fold, 5-fold, 6- fold, 7-fold, 8-fold, 9-fold, or 10-fold difference.
73. A kit for measuring the levels of at least one or more markers selected from the group consisting of FiNC-Macro 1-100, comprising reagents for specifically measuring the levels of the selected FiNC-Macro markers.
74. A kit for measuring the levels of at least one or more markers selected from the group consisting of FiNC-Neutro 1-100, comprising reagents for specifically measuring the levels of the selected FiNC-Neutro markers.
75. A kit for measuring the levels of at least one or more markers selected from the group consisting of HNC-Macro 1-100 and at least one or more markers selected from the group consisting of HNC-Neutro 1-100, comprising reagents for specifically measuring the levels of the selected HNC-Macro markers and reagents for specifically measuring the levels of the selected HNC-Neutro markers.
76. The kit of claim 73 or 75, wherein the selected HNC-Macro markers comprise one or more markers selected from the group consisting of NR4A3, PRDMl, CCRN4L, ICOSLG, GPR183, NR4A1, CLN8, P2RY10, and TNFAIP3.
77. The kit of claim 74 or 75, wherein the selected FiNC-Neutro markers comprise one or more markers selected from the group consisting of SLC25 A6, RPS4X, RPL3, RPL18, and OBFC2B.
78. The kit of any one of claims 73-77, wherein the reagents comprise one or more antibodies or fragments thereof, oligonucleotides, or aptamers.
79. A method of treating or preventing head and neck cancer in a subject comprising administering to said subject an agent that modulates the activity or expression of at least one or more markers selected from the group consisting of HNC-Macro 1-100.
80. A method of treating or preventing head and neck cancer in a subject comprising administering to said subject an agent that modulates the activity or expression of at least one or more markers selected from the group consisting of HNC-Neutro 1-100.
81. The method of claim 79 or 80, wherein the agent is a small molecule modulator, siRNA, or an antibody or fragment thereof.
PCT/US2014/022146 2013-03-09 2014-03-07 Methods of detecting head and neck cancer WO2014164365A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361775558P 2013-03-09 2013-03-09
US61/775,558 2013-03-09

Publications (1)

Publication Number Publication Date
WO2014164365A1 true WO2014164365A1 (en) 2014-10-09

Family

ID=51658859

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/022146 WO2014164365A1 (en) 2013-03-09 2014-03-07 Methods of detecting head and neck cancer

Country Status (1)

Country Link
WO (1) WO2014164365A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104792687A (en) * 2015-04-29 2015-07-22 辽宁医学院附属第一医院 Identification method of primed state of basophil

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040110221A1 (en) * 2002-11-21 2004-06-10 Wyeth Methods for diagnosing RCC and other solid tumors
US20080261258A1 (en) * 2004-07-09 2008-10-23 Amaox, Inc. Immune Cell Biosensors and Methods of Using Same
US20110104062A1 (en) * 2008-02-07 2011-05-05 K. W. Michael Siu Biomarkers for Head-And-Neck Cancers and Precancers
WO2012115885A1 (en) * 2011-02-22 2012-08-30 Caris Life Sciences Luxembourg Holdings, S.A.R.L. Circulating biomarkers

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040110221A1 (en) * 2002-11-21 2004-06-10 Wyeth Methods for diagnosing RCC and other solid tumors
US20080261258A1 (en) * 2004-07-09 2008-10-23 Amaox, Inc. Immune Cell Biosensors and Methods of Using Same
US20110104062A1 (en) * 2008-02-07 2011-05-05 K. W. Michael Siu Biomarkers for Head-And-Neck Cancers and Precancers
WO2012115885A1 (en) * 2011-02-22 2012-08-30 Caris Life Sciences Luxembourg Holdings, S.A.R.L. Circulating biomarkers

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104792687A (en) * 2015-04-29 2015-07-22 辽宁医学院附属第一医院 Identification method of primed state of basophil

Similar Documents

Publication Publication Date Title
US20230280346A1 (en) Methods of detecting prostate cancer
US20200332366A1 (en) Methods of detecting cancer
US20200318201A1 (en) Methods of detecting prostate cancer
US11111537B2 (en) Methods of detecting autoimmune or immune-related diseases or conditions
EP2596117B1 (en) Methods of detecting kidney-associated diseases or conditions
EP2596349B1 (en) Methods of detecting cardiovascular diseases or conditions
US20200040404A1 (en) Compositions and methods for detecting prostate cancer
WO2014164365A1 (en) Methods of detecting head and neck cancer
NZ771630B2 (en) Methods of detecting prostate cancer
WO2023014933A2 (en) Compositions and methods for detecting prostate 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: 14780198

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14780198

Country of ref document: EP

Kind code of ref document: A1