US20200332364A1 - Compositions and methods for detection of diseases related to exposure to inhaled carcinogens - Google Patents

Compositions and methods for detection of diseases related to exposure to inhaled carcinogens Download PDF

Info

Publication number
US20200332364A1
US20200332364A1 US16/611,628 US201816611628A US2020332364A1 US 20200332364 A1 US20200332364 A1 US 20200332364A1 US 201816611628 A US201816611628 A US 201816611628A US 2020332364 A1 US2020332364 A1 US 2020332364A1
Authority
US
United States
Prior art keywords
interleukin
disease
receptor
nucleic acid
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/611,628
Inventor
Walter Joseph Jessen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Laboratory Corp of America Holdings
Original Assignee
Laboratory Corp of America Holdings
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 Laboratory Corp of America Holdings filed Critical Laboratory Corp of America Holdings
Priority to US16/611,628 priority Critical patent/US20200332364A1/en
Assigned to LABORATORY CORPORATION OF AMERICA HOLDINGS reassignment LABORATORY CORPORATION OF AMERICA HOLDINGS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JESSEN, Walter Joseph
Publication of US20200332364A1 publication Critical patent/US20200332364A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06FELECTRIC DIGITAL DATA PROCESSING
    • G06F16/00Information retrieval; Database structures therefor; File system structures therefor
    • G06F16/20Information retrieval; Database structures therefor; File system structures therefor of structured data, e.g. relational data
    • G06F16/21Design, administration or maintenance of databases
    • G06F16/215Improving data quality; Data cleansing, e.g. de-duplication, removing invalid entries or correcting typographical errors
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06FELECTRIC DIGITAL DATA PROCESSING
    • G06F16/00Information retrieval; Database structures therefor; File system structures therefor
    • G06F16/20Information retrieval; Database structures therefor; File system structures therefor of structured data, e.g. relational data
    • G06F16/24Querying
    • G06F16/245Query processing
    • G06F16/2458Special types of queries, e.g. statistical queries, fuzzy queries or distributed queries
    • G06F16/2465Query processing support for facilitating data mining operations in structured databases
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06FELECTRIC DIGITAL DATA PROCESSING
    • G06F16/00Information retrieval; Database structures therefor; File system structures therefor
    • G06F16/90Details of database functions independent of the retrieved data types
    • G06F16/901Indexing; Data structures therefor; Storage structures
    • G06F16/9024Graphs; Linked lists
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B5/00ICT specially adapted for modelling or simulations in systems biology, e.g. gene-regulatory networks, protein interaction networks or metabolic networks
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B50/00ICT programming tools or database systems specially adapted for bioinformatics
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B50/00ICT programming tools or database systems specially adapted for bioinformatics
    • G16B50/30Data warehousing; Computing architectures
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/12Pulmonary diseases
    • G01N2800/122Chronic or obstructive airway disorders, e.g. asthma COPD
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A90/00Technologies having an indirect contribution to adaptation to climate change
    • Y02A90/10Information and communication technologies [ICT] supporting adaptation to climate change, e.g. for weather forecasting or climate simulation

Definitions

  • LC lung cancer
  • COPD chronic obstructive pulmaonary disease
  • CVD cardiovascular disease
  • LC lung cancer
  • COPD chronic obstructive pulmaonary disease
  • CVD cardiovascular disease
  • a method to detect biomarkers associated with at least one of lung cancer (LC) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount, or a mutation in, a nucleic acid (e.g.
  • genomic DNA or mRNA that encodes for, or regulates expression of, the gene for at least one of anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRC5), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAM5), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNA5), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine
  • ABO blood group transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase
  • ABO angiotensin I converting enzyme 2
  • ACE2 angiotensinogen
  • APLN apelin
  • APOA1 apolipoprotein A1
  • APOA2 apolipoprotein A2
  • APOB apolipoprotein B
  • APOE apolipoprotein E
  • caspase 1 caspase 1
  • CD36 molecule CD36
  • C-reactive protein C-reactive protein
  • CRP pentraxin-related
  • CRP pentraxin-related
  • ELL elongation factor for RNA polymerase II
  • F2 intercellular adhesion molecule 1
  • IAM1 interleukin 1 beta
  • the method may include measurement of at least one normalization (e.g., housekeeping gene). Or, measurement of various combinations of these markers may be performed.
  • at least one normalization e.g., housekeeping gene
  • a typical immunoglobulin (antibody) structural unit is known to comprise a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD).
  • the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the terms “variable light chain” (VL) and “variable heavy chain” (VH) refer to these light and heavy chains respectively.
  • An antibody can be specific for a particular antigen.
  • the antibody or its antigen can be either an analyte or a binding partner.
  • Antibodies exist as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases.
  • pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)′2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond.
  • the F(ab)′2 may be reduced under mild conditions to break the disulfide linkage in the hinge region thereby converting the (Fab′)2 dimer into an Fab′ monomer.
  • the Fab′ monomer is essentially an Fab with part of the hinge region (see, Fundamental Immunology, W. E. Paul, ed., Raven Press, N.Y.
  • a single chain Fv (“scFv”) polypeptide is a covalently linked VH::VL heterodimer which may be expressed from a nucleic acid including VH- and VL-encoding sequences either joined directly or joined by a peptide-encoding linker.
  • VH- and VL-encoding sequences either joined directly or joined by a peptide-encoding linker.
  • Allele specific primer extension refers to a mutation detection method utilizing primers which hybridize to a corresponding DNA sequence and which are extended depending on the successful hybridization of the 3′ terminal nucleotide of such primer.
  • extension primers that possess a 3′ terminal nucleotide which form a perfect match with the target sequence are extended to form extension products.
  • Modified nucleotides can be incorporated into the extension product, such nucleotides effectively labeling the extension products for detection purposes.
  • an extension primer may instead comprise a 3′ terminal nucleotide which forms a mismatch with the target sequence. In this instance, primer extension does not occur unless the polymerase used for extension inadvertently possesses exonuclease activity.
  • asymmetric PCR may be used to amplify predominantly or exclusively a single stranded product as is well known in the art (e.g., Poddar, Molec. And Cell. Probes 14:25-32 (2000)). This can be achieved using each pair of primers by reducing the concentration of one primer significantly relative to the other primer of the pair (e.g., 100 fold difference). Amplification by asymmetric PCR is generally linear. A skilled artisan will understand that different amplification methods may be used together.
  • control has its art-understood meaning of being a standard against which results are compared. Typically, controls are used to augment integrity in experiments by isolating variables in order to make a conclusion about such variables.
  • a control is a reaction or assay that is performed simultaneously with a test reaction or assay to provide a comparator. In one experiment, the “test” (i.e., the variable being tested) is applied. In the second experiment, the “control,” the variable being tested is not applied.
  • a control is a historical control (i.e., of a test or assay performed previously, or an amount or result that is previously known).
  • a control is or comprises a printed or otherwise saved record. A control may be a positive control or a negative control.
  • deletion encompasses a mutation that removes one or more nucleotides from a naturally-occurring nucleic acid.
  • Epitope refers to a fragment or portion of a molecule or a molecule compound (e.g., a polypeptide or a protein complex) that makes contact with a particular antibody or antibody like proteins.
  • exon is a nucleic acid sequence that is found in mature or processed RNA after other portions of the RNA (e.g., intervening regions known as introns) have been removed by RNA splicing. As such, exon sequences generally encode for proteins or portions of proteins.
  • An intron is the portion of the RNA that is removed from surrounding exon sequences by RNA splicing.
  • a reagent may be directly or indirectly labeled with a detectable substance.
  • the detectable substance may be, for example, selected, e.g., from a group consisting of radioisotopes, fluorescent compounds, enzymes, and enzyme co-factor. Methods of labeling antibodies are well known in the art.
  • Flanking is meant that a primer hybridizes to a target nucleic acid adjoining a region of interest sought to be amplified on the target.
  • preferred primers are pairs of primers that hybridize 3′ from a region of interest, one on each strand of a target double stranded DNA molecule, such that nucleotides may be add to the 3′ end of the primer by a suitable DNA polymerase.
  • primers that flank mutant sequences do not actually anneal to the mutant sequence but rather anneal to a sequence that adjoins the mutant sequence.
  • primers that flank an exon are generally designed not to anneal to the exon sequence but rather to anneal to sequence that adjoins the exon (e.g. intron sequence). However, in some cases, amplification primer may be designed to anneal to the exon sequence.
  • identity refers to sequence identity between two amino acid sequences or between two nucleic acid sequences. Percent identity can be determined by aligning two sequences and refers to the number of identical residues (i.e., amino acid or nucleotide) at positions shared by the compared sequences. Sequence alignment and comparison may be conducted using the algorithms standard in the art (e.g. Smith and Waterman, 1981, Adv. Appl. Math. 2:482; Needleman and Wunsch, 1970, J. Mol. Biol. 48:443; Pearson and Lipman, 1988, Proc. Natl. Acad.
  • in vivo refers to events that occur within a multi-cellular organism such as a human or a non-human animal.
  • Labeled proteins and peptides can be prepared by incorporation of, or conjugation to, a label that is detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical, chemical or other means.
  • a label or labeling moiety may be directly detectable (i.e., it does not require any further reaction or manipulation to be detectable, e.g., a fluorophore is directly detectable) or it may be indirectly detectable (i.e., it is made detectable through reaction or binding with another entity that is detectable, e.g., a hapten is detectable by immunostaining after reaction with an appropriate antibody comprising a reporter such as a fluorophore).
  • the primary transcript can be cleaved by the Drosha ribonuclease III enzyme to produce an approximately 70-nucleotide stem-loop precursor miRNA (pre-miRNA), which can further be cleaved by the cytoplasmic Dicer ribonuclease to generate the mature miRNA and antisense miRNA star (miRNA*) products.
  • pre-miRNA a RNA-induced silencing complex
  • RISC RNA-induced silencing complex
  • Mutation and/or variant As used herein, the terms mutation and variant are used interchangeably to describe a nucleic acid or protein sequence change.
  • the term “mutant” as used herein refers to a mutated, or potentially non-functional form of a gene. The term includes any mutation that renders a gene not functional from a point mutation to large chromosomal rearrangements as is known in the art.
  • Polypeptide or protein refers to a polymer of amino acids, and not to a specific length. Thus, peptides, oligopeptides and proteins are included within the definition of polypeptide and/or protein. “Polypeptide” and “protein” are used interchangeably herein to describe protein molecules that may comprise either partial or full-length proteins. The term “peptide” is used to denote a less than full-length protein or a very short protein unless the context indicates otherwise.
  • proteins As is known in the art, “proteins”, “peptides,” “polypeptides” and “oligopeptides” are chains of amino acids (typically L-amino acids) whose alpha carbons are linked through peptide bonds formed by a condensation reaction between the carboxyl group of the alpha carbon of one amino acid and the amino group of the alpha carbon of another amino acid.
  • amino acids making up a protein are numbered in order, starting at the amino terminal residue and increasing in the direction toward the carboxy terminal residue of the protein.
  • Abbreviations for amino acid residues are the standard 3-letter and/or 1-letter codes used in the art to refer to one of the 20 common L-amino acids.
  • the term “specific,” when used in connection with an oligonucleotide primer, refers to an oligonucleotide or primer, which under appropriate hybridization or washing conditions, is capable of hybridizing to the target of interest and not substantially hybridizing to nucleic acids which are not of interest. Higher levels of sequence identity are preferred and include at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% sequence identity.
  • Solid support means a structure that provides a substrate onto which biomolecules may be bound.
  • a solid support may be an assay well (i.e., such as a microtiter plate), or the solid support may be a location on an array, or a mobile support, such as a bead.
  • the disclosure herein provides novel mutations identified in a disease and/or syndrome of interest gene that can be used for more accurate diagnosis of disorders relating to the gene and/or syndrome of interest.
  • the sample contains nucleic acid.
  • the testing step comprises nucleic acid sequencing.
  • the testing step comprises hybridization.
  • the hybridization is performed using one or more oligonucleotide probes specific for a region in the biomarker of interest.
  • hybridization is performed under conditions sufficiently stringent to disallow a single nucleotide mismatch.
  • the hybridization is performed with a microarray.
  • the testing step comprises restriction enzyme digestion.
  • the testing step comprises PCR amplification.
  • the PCR amplification is digital PCR amplification.
  • the testing step comprises primer extension.
  • the primer extension is single-base primer extension.
  • the testing step comprises performing a multiplex allele-specific primer extension (ASPE).
  • ASPE multiplex allele-specific primer extension
  • genomic DNA or mRNA that encodes for, or regulates expression of, the gene for at least one of anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRC5), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAM5), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNA5), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine
  • a method to detect biomarkers associated with chronic obstructive pulmaonary disease (COPD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid (e.g.
  • COPD chronic obstructive pulmaonary disease
  • ABO blood group transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase
  • ABO angiotensin I converting enzyme 2
  • ACE2 angiotensinogen
  • APLN apelin
  • APOA1 apolipoprotein A1
  • APOA2 apolipoprotein A2
  • APOB apolipoprotein B
  • APOE apolipoprotein E
  • caspase 1 caspase 1
  • CD36 molecule CD36
  • C-reactive protein C-reactive protein
  • CRP pentraxin-related
  • CRP pentraxin-related
  • ELL elongation factor for RNA polymerase II
  • F2 intercellular adhesion molecule 1
  • IAM1 interleukin 1 beta
  • the sample comprises blood, serum, plasma, cell-free nucleic acid (e.g., DNA or RNA), or a liquid or tissue biopsy.
  • cell-free nucleic acid e.g., DNA or RNA
  • the disclosure provides a method to detect the presence of, or susceptibility to a disease associated with exposure to an inhaled carcinogen in an individual.comprising:obtaining a sample from the individual; measuring the amount of at least one marker associated with at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD), in the sample; and comparing the expression of, and/or the presence of a mutation in a gene for, the at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD) in the sample with a control value for each of the markers.
  • LC lung cancer
  • COPD chronic obstructive pulmaonary disease
  • CVD cardiovascular disease
  • control value is determined from a healthy individual or individuals with no detected or detectable lung or cardiovascular pathology.
  • control is a disease control.
  • disease controls may include individuals with lung or heart disease that is not related to exposure to an inhaled carcinogen.
  • the method may comprise detecting biomarkers associated with chronic obstructive pulmaonary disease (COPD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation in a nucleic acid (e.g.
  • COPD chronic obstructive pulmaonary disease
  • the method may comprise detecting biomarkers associated with cardovascular disease (CVD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation in a nucleic acid (e.g.
  • CVD cardovascular disease
  • the composition may include reagents for the measurement of at least one normalization (e.g., housekeeping) gene.
  • the housekeeping gene may be glyceraldehyde 3-phosphate dehydrogenase.
  • other house keeping genes may be used.
  • measurement of various combinations of these markers may be performed.
  • the composition may comprise reagents for measuring the value of the markers of interest in a control.
  • the control value is determined from a healthy individual or individuals with no detected or detectable lung or cardiovascular pathology.
  • the control is a disease control.
  • Such disease controls may include individuals with lung or heart disease that is not related to exposure to an inhaled carcinogen.
  • kits or reagents comprise instructions or other information and/or computer-readable media comprising instructions and/or other information for performing the methods independent of a kit or reagents therein.
  • Antibodies against particular epitopes, polypeptides, and/or proteins can be generated using any of a variety of known methods in the art.
  • the epitope, polypeptide, or protein against which an antibody is desired can be produced and injected into an animal, typically a mammal (such as a donkey, mouse, rabbit, horse, chicken, etc.), and antibodies produced by the animal can be collected from the animal.
  • Monoclonal antibodies can also be produced by generating hybridomas that express an antibody of interest with an immortal cell line.
  • antibodies are labeled with a detectable moiety as described herein.
  • the biomarker may be allowed to complex with a first binding agent (e.g., primary antibody specific for the biomarker and labeled with detectable moiety) and a second binding agent (e.g., a secondary antibody that recognizes the primary antibody or a second primary antibody), where the second binding agent is complexed to a third binding agent (e.g., biotin) that can then interact with a capture support (e.g., magnetic bead) having a reagent (e.g., streptavidin) that recognizes the third binding agent linked to the capture support.
  • the complex (labeled primary antibody: biomarker: second primary antibody-biotin: streptavidin-bead may then be captured using a magnet (e.g., a magnetic probe) to measure the amount of the complex.
  • the variant may comprise a nucleic acid sequence that encompasses at least one of the following: (1) A-to-I editing sites; (2) splice sites; (3) conserved functional RNA structures; (4) validated transcription factor binding sites (TFBS); (5) microRNA (miRNA) binding sites; (6) polyadenylation sites; (7) known regulatory elements; (8) miRNA genes; (9) small nucleolar RNA genes encoded in the ROIs; and/or (10) ultra-conserved elements across placental mammals.
  • TFBS validated transcription factor binding sites
  • two complementary reactions are used.
  • One reaction employs a primer specific for the wild type allele (“wild-type-specific reaction”) and the other reaction employs a primer for the mutant allele (“mutant-specific reaction”).
  • the two reactions may employ a common second primer.
  • PCR primers specific for a particular allele e.g., the wild-type allele or mutant allele
  • the mismatch may be located at/near the 3′ end of the primer, leading to preferential amplification of the perfectly matched allele. Whether an amplification product can be detected from one or in both reactions indicates the absence or presence of the mutant allele.
  • primer extension can be combined with mass spectrometry for accurate and fast detection of the presence or absence of a mutation. See, U.S. Pat. No. 5,885,775 to Haff et al. (analysis of single nucleotide polymorphism analysis by mass spectrometry); U.S. Pat. No. 7,501,251 to Koster (DNA diagnosis based on mass spectrometry); the teachings of both of which are incorporated herein by reference.
  • oligonucleotide probes vary from 15 to 90, 15 to 80, 15 to 70, 15 to 60, 15 to 50, 15 to 40, 15 to 35, 15 to 30, 18 to 30, or 18 to 26 nucleotides in length.
  • the optimal length of an oligonucleotide probe may depend on the particular methods and/or conditions in which the oligonucleotide probe may be employed.
  • nucleic acid probes are useful as primers, e.g., for nucleic acid amplification and/or extension reactions.
  • the gene sequence being evaluated for a variant comprises the exon sequences.
  • the exon sequence and additional flanking sequence e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55 or more nucleotides of UTR and/or intron sequence
  • intron sequences or other non-coding regions may be evaluated for potentially deleterious mutations.
  • portions of these sequences may be used.
  • Such variant gene sequences may include sequences having at least one of the mutations as described herein.
  • the method may comprise obtaining a nucleic acid from a tissue or body fluid sample from a subject and sequencing at least a portion of a nucleic acid in order to obtain a sample nucleic acid sequence for at least one gene.
  • the method may comprise comparing the variant to known variants associated with a disease associated with exosure to inhaled carcinogens and determining whether the variant is a variant that has been previously identified as being associated with a disease associated with exosure to inhaled carcinogens.
  • the method may comprise identifying the variant as a new, previously uncharacterized variant.
  • next generation may be used.
  • Sanger sequencing may be used.
  • a combination of next-generation (massively-parallel sequencing) and Sanger sequencing may be used.
  • the sequencing comprises at least one of single-molecule sequencing-by-synthesis.
  • a plurality of DNA samples are analyzed in a pool to identify samples that show a variation.
  • a plurality of DNA samples are analyzed in a plurality of pools to identify an individual sample that shows the same variation in at least two pools.
  • sequencing of the nucleic acid is accomplished by massively parallel sequencing (also known as “next generation sequencing”) of single-molecules or groups of largely identical molecules derived from single molecules by amplification through a method such as PCR.
  • massively parallel sequencing is shown for example in Lapidus et al., U.S. Pat. No. 7,169,560, Quake et al. U.S. Pat. No. 6,818,395, Harris U.S. Pat. No. 7,282,337 and Braslaysky, et al., PNAS (USA), 100: 3960-3964 (2003), the contents of each of which are incorporated by reference herein.
  • next generation sequencing PCR or whole genome amplification can be performed on the nucleic acid in order to obtain a sufficient amount of nucleic acid for analysis.
  • no amplification is required because the method is capable of evaluating DNA sequences from unamplified DNA.
  • the sequence and/or genomic arrangement and/or genomic copy number of the nucleic acid from the test sample is compared to a standard reference derived from one or more individuals not known to suffer from a disease associated with exposure to inhaled carcinogens at the time their sample was taken. All differences between the sequence and/or genomic arrangement and/or genomic arrangement and/or copy number of the nucleic acid from the test sample and the standard reference are considered variants.
  • the DNA sequences derived from coding exons of genes included in the assay are enriched by bulk hybridization of randomly fragmented genomic DNA to specific RNA probes.
  • the same adapter sequences are attached to the ends of all fragments, allowing enrichment of all hybridization-captured fragments by PCR with one primer pair in one reaction. Regions that are less efficiently captured by hybridization are amplified by PCR with specific primers.
  • PCR with specific primers is may be used to amplify exons for which similar sequences (“pseudo exons”) exist elsewhere in the genome.
  • nucleotide A, G, T, or C
  • A, G, T, or C a fluorescently labeled dCTP
  • This nucleotide will only be incorporated into those growing complementary DNA strands that need a C as the next nucleotide.
  • an image of the flow cell is taken to determine which fragment was extended. DNA strands that have incorporated a C will emit light, while DNA strands that have not incorporated a C will appear dark. Chain termination is reversed to make the growing DNA strands extendible again, and the process is repeated for a total of 120 cycles.
  • the images are converted into strings of bases, commonly referred to as “reads,” which recapitulate the 3′ terminal 25 to 60 bases of each fragment.
  • the reads are then compared to the reference sequence for the DNA that was analyzed. Since any given string of 25 bases typically only occurs once in the human genome, most reads can be “aligned” to one specific place in the human genome. Finally, a consensus sequence of each genomic region may be built from the available reads and compared to the exact sequence of the reference at that position. Any differences between the consensus sequence and the reference are called as sequence variants.
  • certain molecules used in accordance with and/or provided by the invention comprise one or more detectable entities or moieties, i.e., such molecules are “labeled” with such entities or moieties.
  • Oregon Green Dyes e.g., Oregon Green 488, Oregon Green 500, Oregon Green 514., etc.
  • Texas Red Texas Red-X
  • SPECTRUM RED SPECTRUM GREEN
  • cyanine dyes e.g., CY-3, CY-5, CY-3.5, CY-5.5, etc.
  • ALEXA FLUOR dyes e.g., ALEXA FLUOR 350, ALEXA FLUOR 488, ALEXA FLUOR 532, ALEXA FLUOR 546, ALEXA FLUOR 568, ALEXA FLUOR 594, ALEXA FLUOR 633, ALEXA FLUOR 660, ALEXA FLUOR 680, etc.
  • BODIPY dyes e.g., BODIPY FL, BODIPY R6G, BODIPY R, BODIPY TR, BODIPY 530/550, BODIPY 558/568, BODIPY 5
  • a detectable moiety may include more than one chemical entity such as in fluorescent resonance energy transfer (FRET).
  • FRET fluorescent resonance energy transfer
  • the first fluorescent molecule absorbs light and transfers it through the resonance of excited electrons to the second fluorescent molecule (the “acceptor” fluor).
  • both the donor and acceptor dyes can be linked together and attached to the oligo primer. Methods to link donor and acceptor dyes to a nucleic acid have been described, for example, in U.S. Pat. No.
  • a detectable moiety is a radioactive isotope.
  • a molecule may be isotopically-labeled (i.e., may contain one or more atoms that have been replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature) or an isotope may be attached to the molecule.
  • Non-limiting examples of isotopes that can be incorporated into molecules include isotopes of hydrogen, carbon, fluorine, phosphorous, copper, gallium, yttrium, technetium, indium, iodine, rhenium, thallium, bismuth, astatine, samarium, and lutetium (i.e., 3H, 13C, 14C, 18F, 19F, 32P, 35S, 64Cu, 67Cu, 67Ga, 90Y, 99mTc, 111In, 125I, 123I, 129I, 131I, 135I, 186Re, 187Re, 201Tl, 212Bi, 213Bi, 211At, 153Sm, 177Lu).
  • kits for use in accordance with methods and compositions disclosed herein.
  • kits comprise one or more reagents detect the biomarker of interest.
  • Suitable reagents may include nucleic acid probes and/or antibodies or fragments thereof.
  • suitable reagents are provided in a form of an array such as a microarray or a mutation panel.
  • Kits may also contain instructions on how to determine if an individual has the disease and/or syndrome of interest, or is at risk of developing the disease and/or syndrome of interest.
  • the disclosure comprises methods to identify biomarkers for a syndrome or disease of interest (i.e., variants in nucleic acid sequence that are associated with a disease associated with exposure to inhaled carcinogens in a statistically significant manner).
  • the genes and/or genomic regions assayed for new markers may be selected based upon their importance in biochemical pathways that show genetic linkage and/or biological causation to the syndrome and/or disease of interest.
  • the genes and/or genomic regions assayed for markers may be selected based on genetic linkage to DNA regions that are genetically linked to the inheritance of a disease associated with exposure to an inhaled carcinogen in families.
  • the genes and/or genomic regions assayed for markers may be evaluated systematically to cover certain regions of chromosomes not yet evaluated.
  • a variant or a variant combination is reported or known to be transmitted exclusively or preferentially to individuals having the syndrome and/or disease of interest, it is considered to be at least potentially predisposing to the syndrome and/or disease of interest. Conversely, if a variant is found in both populations at a similar frequency, it is less likely to be associated with the development of the syndrome and/or disease of interest.
  • a variant or a variant combination is predicted to have an overall deleterious effect on a protein or gene expression (i.e., resulting in a nonsense mutation, a frameshift mutation, or a splice site mutation, or even a missense mutation), based on the predicted effect on the sequence and/or the structure of a protein or a nucleic acid, and if this variant or variant combination affects a gene or genes known to be associated with the syndrome and/or disease of interest, it is considered to be at least potentially predisposing to the syndrome and/or disease of interest.
  • the disclosure herein provides methods for diagnosing the presence or an increased risk of developing a disease associated with in a subject.
  • Such methods may include obtaining a nucleic acid from a sample of tissue or body fluid.
  • the method may further include sequencing the nucleic acid or determining the genomic arrangement or copy number of the nucleic acid to detect whether there is a variant or variants in the nucleic acid sequence or genomic arrangement or copy number.
  • the method may further include the steps of assessing the clinical significance of a variant or variants.
  • Such analysis may include an evaluation of the extent of association of the variant sequence in affected populations (i.e., subjects having the disease).
  • Such analysis may also include an analysis of the extent of the effect the mutation may have on gene expression and/or protein function.
  • the method may also include diagnosis the presence or an increased risk of developing a disease related to exposure to inhaled carcinogens based on the assessment.
  • EGFR expression levels are measured by fluorescent in situ hybridization (FISH) using 4-5 micron FFPE sections of tissue.
  • FISH fluorescent in situ hybridization
  • expresssion may be measured in serum, plasma, urine, bone marrow, blood, cerebrospinal fluid, FNA, or bone marrow.
  • Gene mutation analysis is performed using real-time PCR, single base extension and/or DNA sequencing. Gene mutation can be determined in, for examples, tissue, serum, plasma or urine.
  • Interleukin 8 (IL8 or chemokine (C-X-C motif) ligand 8, CXCL8) is a chemokine produced by macrophages and other cell types such as epithelial cells, airway smooth muscle cells and endothelial cells.
  • the interleukin-8 protein is encoded by the CXCL8 gene.
  • IL-8 is initially produced as a precursor peptide of 99 amino acids which then undergoes cleavage to create several active IL-8 isoforms.
  • a 72 amino acid peptide is the major form secreted by macrophages IL-8, also known as neutrophil chemotactic factor, has two primary functions.
  • Plasminogen activator inhibitor-1 also known as endothelial plasminogen activator inhibitor or serpin E1 is a protein that in humans is encoded by the SERPINE1 gene. Elevated PAI-1 is a risk factor for thrombosis and atherosclerosis. PAI-1 is a serine protease inhibitor (serpin) that functions as the principal inhibitor of tissue plasminogen activator (tPA) and urokinase (uPA), activators of plasminogen and fibrinolysis. It is a serine protease inhibitor (serpin) protein (SERPINE1).
  • the PAI-1 gene is SERPINE1, located on chromosome 7 (7q21.3-q22). There is a common polymorphism known as 4G/5G in the promoter region. The 5G allele is slightly less transcriptionally active than the 4G.
  • STAT3 Signal Transducer and Activator of Transcription 3
  • IL-6 's role as an anti-inflammatory cytokine is mediated through its inhibitory effects on TNF-alpha and IL-1, and activation of IL-1ra and IL-10.
  • IL-6 is an important mediator of fever and of the acute phase response. It is capable of crossing the blood-brain barrier, initiating synthesis of PGE2 in the hypothalamus, and changing the body's temperature setpoint. In muscle and fatty tissue, IL-6 stimulates energy mobilization that leads to increased body temperature.
  • IL-6 can be secreted by macrophages in response to specific microbial molecules, referred to as pathogen-associated molecular patterns (PAMPs), and is also considered a myokine (a cytokine produced from muscle), which is elevated in response to muscle contraction
  • PAMPs pathogen-associated molecular patterns
  • VEGF Vascular endothelial growth factor
  • VPF vascular permeability factor
  • VEGF has potent angiogenic, mitogenic, and vascular permeability-enhancing activities specific for endothelial cells.
  • VEGF is thought to play an important role in several physiologic processes, including wound healing, ovulation, menstruation, maintenance of blood pressure, and pregnancy.
  • VEGF has also been associated with a number of pathologic processes that involve angiogenesis, including arthritis, psoriasis, macular degeneration, and diabetic retinopathy.
  • tumor expression of proangiogenic factors, including VEGF has been associated with advanced tumor progression in a number of human cancers.
  • VEGF can be measured in serum using enzyme immunoassay (EIA).
  • CCL2 The chemokine (C-C motif) ligand 2 (CCL2) is also referred to as monocyte chemoattractant protein 1 (MCP1) and small inducible cytokine A2.
  • CCL2 is a small cytokine that belongs to the CC chemokine family. CCL2 recruits monocytes, memory T cells, and dendritic cells to the sites of inflammation produced by either tissue injury or infection. Administration of anti-CCL2 antibodies in a model of glomerulonephritis reduces infiltration of macrophages and T cells, reduces crescent formation, as well as scarring and renal impairment. Hypomethylation of CpG sites within the CCL2 promoter region is affected by high levels of blood glucose and TG, which increase CCL2 levels in the blood serum. The later plays an important role in the vascular complications of type 2 diabetes.
  • Phosphatidylinositol-4,5,-bisphosphate 3-kinase Catalytic Subunit Alpha PIK3CA

Abstract

Disclosed are compositions and methods to detect proteins associated with diseases associated with exposure to inhaled carcinogens. Such markers may be useful to allow individuals susceptible to diseases associated with exposure to inhaled carcinogens to manage their lifestyle and reduce further progression of disease.

Description

    RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Patent Application No. 62/505,536, filed May 12, 2017 and U.S. Provisional Patent Application 62/523,382, filed Jun. 22, 2017. The contents of U.S. Provisional Patent Application Nos. 62/505,536 and 62/523,382 are incorporated by reference in their entireties herein.
  • FIELD OF DISCLOSURE
  • The disclosure relates to methods and compositions for diagnosing lung cancer and related diseases.
  • BACKGROUND
  • The biological mechanisms behind diseases potentially associated with exposure to inhaled carcinogens. Such dieaseses may include lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and cardiovascular disease (CVD). Although highly prevalent diseases, there are few, if any, biomarkers that provide a reliable indication of these conditions. It would be helpful for individuals having exposure to risk factors to adjust their lifestyle so as to avoid triggering an onset of symptoms and/or promoting further progression of the disease. Thus, there is a need to develop and evaluate biomarkers for such diseases.
  • SUMMARY
  • The present disclosure may be embodied in a variety of ways.
  • In an embodiment disclosed are methods and compositions to detect biomarkers associated with at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD) and correlating the levels and/or changes in such biomarkers with exposure to an inhaled carcinogen.
  • In an embodiment, disclosed is a method to detect at least one biomarker associated with at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD) comprising:
  • obtaining a sample from the individual; and measuring the amount of the marker, and optionally, correlating the levels and/or changes in such biomarkers with exposure to an inhaled carcinogen.
  • In one embodiment, disclosed is a method to detect biomarkers associated with at least one of lung cancer (LC) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount, or a mutation in, a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRC5), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAM5), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNA5), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine receptor 4 (CXCR4), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), cytochrome P450 family 1 subfamily B member 1 (CYP1B1), epidermal growth factor receptor (EGFR), epoxide hydrolase 1 (EPHX1), erb-b2 receptor tyrosine kinase 2 (ERBB2), fructose-bisphosphatase 1 (FBP1), fascin actin-bundling protein 1 (FSCN1), glutathione S-transferase pi 1 (GSTP1), interleukin 10 (IL10), integrin subunit alpha 11 (ITGA11), KRAS proto-oncogene, GTPase (KRAS), keratin 19 (KRT19), leucine aminopeptidase 3 (LAP3), MDM2 proto-oncogene (MDM2), v-myc avian myelocytomatosis viral oncogene homolog (MYC), p21 (RAC1) activated kinase 1 (PAK1), poly(ADP-ribose) polymerase 1 (PARP1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), protein kinase N1 (PKN1), phosphatase and tensin homolog (PTEN), signal transducer and activator of transcription 3 (STAT3), serine/threonine kinase 11 (STK11), tumor protein p53 (TP53), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor C (VEGFC), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or X-ray repair cross complementing 1 (XRCC1).
  • In one embodiment, disclosed is a method to detect biomarkers associated with chronic obstructive pulmaonary disease (COPD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of adiponectin, ClQ and collagen domain containing (ADIPOQ), adrenoceptor beta 2 (ADRB2), advanced glycosylation end product-specific receptor (AGER), CD4 molecule (CD4), cystic fibrosis transmembrane conductance regulator (CFTR), cholinergic receptor nicotinic alpha 3 subunit (CHRNA3), cystatin C (CST3), C-X-C motif chemokine ligand 8 (CXCL8), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), D-box binding PAR bZIP transcription factor (DBP), epidermal growth factor receptor (EGFR), elastin (ELN), erythropoietin (EPO), glutathione S-transferase pi 1 (GSTP1), histone deacetylase 2 (HDAC2), high mobility group box 1 (HMGB1), 5-hydroxytryptamine receptor 4 (HTR4), immunoglobulin heavy constant epsilon (IGHE), interleukin 10 (IL10), interleukin 13 (IL13), interleukin 1 beta (IL1B), laminin subunit alpha 1 (LAMA1), leptin (LEP), membrane metallo-endopeptidase (MME), matrix metallopeptidase 12 (MMP12), matrix metallopeptidase 25 (MMP25), serpin family A member 1 (SERPINA1), sirtuin 1 (SIRT1), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or vascular endothelial growth factor A (VEGFA).
  • In one embodiment, disclosed is a method to detect biomarkers associated with cardovascular disease (CVD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of ABO blood group (transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase) (ABO), angiotensin I converting enzyme 2 (ACE2), angiotensinogen (AGT), albumin (ALB), apelin (APLN), apolipoprotein A1 (APOA1), apolipoprotein A2 (APOA2), apolipoprotein B (APOB), apolipoprotein E (APOE), caspase 1 (CASP1), CD36 molecule (CD36), C-reactive protein, pentraxin-related (CRP), elongation factor for RNA polymerase II (ELL), coagulation factor II, thrombin (F2), intercellular adhesion molecule 1 (ICAM1), interleukin 1 beta (IL1B), low density lipoprotein receptor (LDLR), leptin (LEP), myeloperoxidase (MPO), nitric oxide synthase 3 (NOS3), period circadian clock 1 (PERI1), prolactin (PRL), tumor necrosis factor (TNF), troponin C1, slow skeletal and cardiac type (TNNC1), troponin 13, cardiac type (TNNI3), troponin T2, cardiac type (TNNT2), vascular endothelial growth factor A (VEGFA), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or von Willebrand factor (VWF).
  • Additionally and/or alternatively, the method may include measurement of at least one normalization (e.g., housekeeping gene). Or, measurement of various combinations of these markers may be performed.
  • Other features, objects, and advantages of the disclosure herein are apparent in the detailed description, drawings and claims that follow. It should be understood, however, that the detailed description, the drawings, and the claims, while indicating embodiments of the disclosed methods, compositions and systems, are given by way of illustration only, not limitation. Various changes and modifications within the scope of the invention will become apparent to those skilled in the art.
  • FIGURES
  • The dislosure may be better understood in view of the following non-limiting figures.
  • FIG. 1 shows an example of a multi-node interaction network identifying markers associated with a disease (COPD) that may be associated with inhaled carcinogens.
  • FIG. 2 shows an example of a Venn diagram depicting markers associated with lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and cardiovascular disease (CVD).
  • DETAILED DESCRIPTION
  • Terms and Definitions
  • In order for the disclosure to be more readily understood, certain terms are first defined. Additional definitions for the following terms and other terms are set forth throughout the specification.
  • Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the disclosure are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements. Moreover, all ranges disclosed herein are to be understood to encompass any and all subranges subsumed therein. For example, a stated range of “1 to 10” should be considered to include any and all subranges between (and inclusive of) the minimum value of 1 and the maximum value of 10; that is, all subranges beginning with a minimum value of 1 or more, e.g. 1 to 6.1, and ending with a maximum value of 10 or less, e.g., 5.5 to 10. Additionally, any reference referred to as being “incorporated herein” is to be understood as being incorporated in its entirety.
  • It is further noted that, as used in this specification, the singular forms “a,” “an,” and “the” include plural referents unless expressly and unequivocally limited to one referent. The term “and/or” generally is used to refer to at least one or the other. In some case the term “and/or” is used interchangeably with the term “or.” The term “including” is used herein to mean, and is used interchangeably with, the phrase “including but not limited to.” The term “such as” is used herein to mean, and is used interchangeably with, the phrase “such as but not limited to.”
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. Practitioners are particularly directed to Current Protocols in Molecular Biology (Ausubel) for definitions and terms of the art.
  • Also as used herein, “at least one” contemplates any number from 1 to the entire group. For example, for a listing of four markers, the phrase at “least one” is understood to mean 1, 2, 3 or 4 markers.
  • Also, as used herein, “comprising” includes embodiments more particularly defined using the term “consisting of.”
  • Antibody: As used herein, the term “antibody” refers to a polypeptide consisting of one or more polypeptides substantially encoded by immunoglobulin genes or fragments of immunoglobulin genes. The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes. Light chains are typically classified as either kappa or lambda. Heavy chains are typically classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively. A typical immunoglobulin (antibody) structural unit is known to comprise a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms “variable light chain” (VL) and “variable heavy chain” (VH) refer to these light and heavy chains respectively. An antibody can be specific for a particular antigen. The antibody or its antigen can be either an analyte or a binding partner. Antibodies exist as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)′2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond. The F(ab)′2 may be reduced under mild conditions to break the disulfide linkage in the hinge region thereby converting the (Fab′)2 dimer into an Fab′ monomer. The Fab′ monomer is essentially an Fab with part of the hinge region (see, Fundamental Immunology, W. E. Paul, ed., Raven Press, N.Y. (1993), for a more detailed description of other antibody fragments). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of ordinary skill in the art will appreciate that such Fab′ fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term “antibody,” as used herein also includes antibody fragments either produced by the modification of whole antibodies or synthesized de novo using recombinant DNA methodologies. In some embodiments, antibodies are single chain antibodies, such as single chain Fv (scFv) antibodies in which a variable heavy and a variable light chain are joined together (directly or through a peptide linker) to form a continuous polypeptide. A single chain Fv (“scFv”) polypeptide is a covalently linked VH::VL heterodimer which may be expressed from a nucleic acid including VH- and VL-encoding sequences either joined directly or joined by a peptide-encoding linker. (See, e.g., Huston, et al. (1988) Proc. Nat. Acad. Sci. USA, 85:5879-5883, the entire contents of which are herein incorporated by reference.) A number of structures exist for converting the naturally aggregated, but chemically separated light and heavy polypeptide chains from an antibody V region into an scFv molecule which will fold into a three dimensional structure substantially similar to the structure of an antigen-binding site. See, e.g. U.S. Pat. Nos. 5,091,513 and 5,132,405 and 4,956,778.
  • The term “antibody” includes monoclonal antibodies, polyclonal antibodies, synthetic antibodies and chimeric antibodies, e.g., generated by combinatorial mutagenesis and phage display. The term “antibody” also includes mimetics or peptidomimetics of antibodies. Peptidomimetics are compounds based on, or derived from, peptides and proteins. The peptidomimetics of the present disclosure typically can be obtained by structural modification of a known peptide sequence using unnatural amino acids, conformational restraints, isosteric replacement, and the like.
  • Allele: As used herein, the term “allele” refers to different versions of a nucleotide sequence of a same genetic locus (e.g., a gene).
  • Allele specific primer extension (ASPE): As used herein, the term “allele specific primer extension (ASPE)” refers to a mutation detection method utilizing primers which hybridize to a corresponding DNA sequence and which are extended depending on the successful hybridization of the 3′ terminal nucleotide of such primer. Typically, extension primers that possess a 3′ terminal nucleotide which form a perfect match with the target sequence are extended to form extension products. Modified nucleotides can be incorporated into the extension product, such nucleotides effectively labeling the extension products for detection purposes. Alternatively, an extension primer may instead comprise a 3′ terminal nucleotide which forms a mismatch with the target sequence. In this instance, primer extension does not occur unless the polymerase used for extension inadvertently possesses exonuclease activity.
  • Amplification: As used herein, the term “amplification” refers to any methods known in the art for copying a target nucleic acid, thereby increasing the number of copies of a selected nucleic acid sequence. Amplification may be exponential or linear. A target nucleic acid may be either DNA or RNA. Typically, the sequences amplified in this manner form an “amplicon.” Amplification may be accomplished with various methods including, but not limited to, the polymerase chain reaction (“PCR”), transcription-based amplification, isothermal amplification, rolling circle amplification, etc. Amplification may be performed with relatively similar amount of each primer of a primer pair to generate a double stranded amplicon. However, asymmetric PCR may be used to amplify predominantly or exclusively a single stranded product as is well known in the art (e.g., Poddar, Molec. And Cell. Probes 14:25-32 (2000)). This can be achieved using each pair of primers by reducing the concentration of one primer significantly relative to the other primer of the pair (e.g., 100 fold difference). Amplification by asymmetric PCR is generally linear. A skilled artisan will understand that different amplification methods may be used together.
  • Animal: As used herein, the term “animal” refers to any member of the animal kingdom. In some embodiments, “animal” refers to humans, at any stage of development. In some embodiments, “animal” refers to non-human animals, at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, and/or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, insects, and/or worms. In some embodiments, an animal may be a transgenic animal, genetically-engineered animal, and/or a clone.
  • Approximately: As used herein, the term “approximately” or “about,” as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term “approximately” or “about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value). Also, throughout this application, the term “about” is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among samples.
  • Associated with a syndrome or disease of interest: As used herein, “associated with a syndrome or disease of interest” means that the variant is found with in patients with the syndrome or disease of interest more than in non-syndromic or non-disease controls. Generally, the statistical significance of such association can be determined by assaying a plurality of patients.
  • Biological Sample and Sample: As used herein, the term “biological sample” encompasses any sample obtained from a biological source. A biological sample can, by way of non-limiting example, include blood, plasma, serum, liquid or tissue biopsy, urine, feces, epidermal sample, skin sample, cheek swab, sperm, amniotic fluid, cultured cells, bone marrow sample and/or chorionic villi. Convenient biological samples may be obtained by, for example, scraping cells from the surface of the buccal cavity. The term biological sample encompasses samples which have been processed to release or otherwise make available a nucleic acid or protein for detection as described herein. Also included is cell-free nucleic acid (e.g., DNA or RNA) such as that found in plasma, amniotic fluid and the like. For example, a biological sample may include a cDNA that has been obtained by reverse transcription of RNA from cells in a biological sample. The biological sample may be obtained from a stage of life such as a fetus, young adult, adult, and the like. Fixed or frozen tissues also may be used.
  • Biomarker: As used herein, the term “biomarker” or “marker” refers to one or more nucleic acids, polypeptides and/or other biomolecules (e.g., cholesterol, lipids) that can be used to diagnose, or to aid in the diagnosis or prognosis of a disease or syndrome of interest, either alone or in combination with other biomarkers; monitor the progression of a disease or syndrome of interest; and/or monitor the effectiveness of a treatment for a syndrome or a disease of interest.
  • Binding agent: As used herein, the term “binding agent” refers to a molecule that can specifically and selectively bind to a second (i.e., different) molecule of interest. The interaction may be non-covalent, for example, as a result of hydrogen-bonding, van der Waals interactions, or electrostatic or hydrophobic interactions, or it may be covalent. The term “soluble binding agent” refers to a binding agent that is not associated with (i.e., covalently or non-covalently bound) to a solid support.
  • Carrier: The term “carrier” refers to a person who is symptom-free but carries a mutation that can be passed to his/her children. Typically, for an autosomal recessive disorder, a carrier has one allele that contains a disease causing mutation and a second allele that is normal or not disease-related.
  • Coding sequence vs. non-coding sequence: As used herein, the term “coding sequence” refers to a sequence of a nucleic acid or its complement, or a part thereof, that can be transcribed and/or translated to produce the mRNA for and/or the polypeptide or a fragment thereof. Coding sequences include exons in a genomic DNA or immature primary RNA transcripts, which are joined together by the cell's biochemical machinery to provide a mature mRNA. The anti-sense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced therefrom. As used herein, the term “non-coding sequence” refers to a sequence of a nucleic acid or its complement, or a part thereof, that is not transcribed into amino acid in vivo, or where tRNA does not interact to place or attempt to place an amino acid. Non-coding sequences include both intron sequences in genomic DNA or immature primary RNA transcripts, and gene-associated sequences such as promoters, enhancers, silencers, etc.
  • Complement: As used herein, the terms “complement,” “complementary” and “complementarity,” refer to the pairing of nucleotide sequences according to Watson/Crick pairing rules. For example, a sequence 5′-GCGGTCCCA-3′ has the complementary sequence of 5′-TGGGACCGC-3′. A complement sequence can also be a sequence of RNA complementary to the DNA sequence. Certain bases not commonly found in natural nucleic acids may be included in the complementary nucleic acids including, but not limited to, inosine, 7-deazaguanine, Locked Nucleic Acids (LNA), and Peptide Nucleic Acids (PNA). Complementary need not be perfect; stable duplexes may contain mismatched base pairs, degenerative, or unmatched bases. Those skilled in the art of nucleic acid technology can determine duplex stability empirically considering a number of variables including, for example, the length of the oligonucleotide, base composition and sequence of the oligonucleotide, ionic strength and incidence of mismatched base pairs.
  • Conserved: As used herein, the term “conserved residues” refers to amino acids that are the same among a plurality of proteins having the same structure and/or function. A region of conserved residues may be important for protein structure or function. Thus, contiguous conserved residues as identified in a three-dimensional protein may be important for protein structure or function. To find conserved residues, or conserved regions of 3-D structure, a comparison of sequences for the same or similar proteins from different species, or of individuals of the same species, may be made.
  • Control: As used herein, the term “control” has its art-understood meaning of being a standard against which results are compared. Typically, controls are used to augment integrity in experiments by isolating variables in order to make a conclusion about such variables. In some embodiments, a control is a reaction or assay that is performed simultaneously with a test reaction or assay to provide a comparator. In one experiment, the “test” (i.e., the variable being tested) is applied. In the second experiment, the “control,” the variable being tested is not applied. In some embodiments, a control is a historical control (i.e., of a test or assay performed previously, or an amount or result that is previously known). In some embodiments, a control is or comprises a printed or otherwise saved record. A control may be a positive control or a negative control.
  • A “control” or “predetermined standard” for a biomarker refers to the levels of expression of the biomarker in healthy subjects or the expression levels of said biomarker in non-diseased or non-syndromic tissue from the same subject. The control or predetermined standard expression levels or amounts of protein for a given biomarker can be established by prospective and/or retrospective statistical studies using only routine experimentation. Such predetermined standard expression levels and/or protein levels (amounts) can be determined by a person having ordinary skill in the art using well known methods.
  • Crude: As used herein, the term “crude,” when used in connection with a biological sample, refers to a sample which is in a substantially unrefined state. For example, a crude sample can be cell lysates or biopsy tissue sample. A crude sample may exist in solution or as a dry preparation.
  • Deletion: As used herein, the term “deletion” encompasses a mutation that removes one or more nucleotides from a naturally-occurring nucleic acid.
  • Disease or syndrome of interest: As used herein, a disease or syndrome of interest is a disease associated with exposure to inhaled carcinogens.
  • Detect: As used herein, the term “detect”, “detected” or “detecting” includes “measure,” “measured” or “measuring” and vice versa.
  • Detectable moiety: As used herein, the term “detectable moiety” or “detectable biomolecule” or “reporter” refers to a molecule that can be measured in a quantitative assay. For example, a detectable moiety may comprise an enzyme that may be used to convert a substrate to a product that can be measured (e.g., a visible product). Or, a detectable moiety may be a radioisotope that can be quantified. Or, a detectable moiety may be a fluorophore. Or, a detectable moiety may be a luminescent molecule. Or, other detectable molecules may be used.
  • Epigenetic: As used herein, an epigenetic element can change gene expression by a mechanism other than a change in the underlying DNA sequences. Such elements may include elements that regulate paramutation, imprinting, gene silencing, X chromosome inactivation, position effect, reprogramming, transvection, maternal effects, histone modification, and heterochromatin.
  • Epitope: As used herein, the term “epitope” refers to a fragment or portion of a molecule or a molecule compound (e.g., a polypeptide or a protein complex) that makes contact with a particular antibody or antibody like proteins.
  • Exon: As used herein an exon is a nucleic acid sequence that is found in mature or processed RNA after other portions of the RNA (e.g., intervening regions known as introns) have been removed by RNA splicing. As such, exon sequences generally encode for proteins or portions of proteins. An intron is the portion of the RNA that is removed from surrounding exon sequences by RNA splicing.
  • Expression and expressed RNA: As used herein expressed RNA is an RNA that encodes for a protein or polypeptide (“coding RNA”), and any other RNA that is transcribed but not translated (“non-coding RNA”). The term “expression” is used herein to mean the process by which a polypeptide is produced from DNA. The process involves the transcription of the gene into mRNA and the translation of this mRNA into a polypeptide. Depending on the context in which used, “expression” may refer to the production of RNA, protein or both.
  • The measurement of an amount of a protein and/or the expression of a biomarker of the disclosure may be assessed by any of a wide variety of well-known methods for detecting expression of a transcribed molecule or its corresponding protein. Non-limiting examples of such methods include immunological methods for detection of secreted proteins, protein purification methods, protein function or activity assays, nucleic acid hybridization methods, nucleic acid reverse transcription methods, and nucleic acid amplification methods. In certain embodiments, expression of a marker gene is assessed using an antibody (e.g. a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody), an antibody derivative (e.g. an antibody conjugated with a substrate or with the protein or ligand of a protein-ligand pair {e.g. biotin-streptavidin}), or an antibody fragment (e.g. a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically with a protein corresponding to the marker gene, such as the protein encoded by the open reading frame corresponding to the marker gene or such a protein which has undergone all or a portion of its normal post-translational modification. In certain embodiments, a reagent may be directly or indirectly labeled with a detectable substance. The detectable substance may be, for example, selected, e.g., from a group consisting of radioisotopes, fluorescent compounds, enzymes, and enzyme co-factor. Methods of labeling antibodies are well known in the art.
  • In another embodiment, expression of a marker gene is assessed by preparing mRNA/cDNA (i.e. a transcribed polynucleotide) from cells in a sample, and by hybridizing the mRNA/cDNA with a reference polynucleotide which is a complement of a polynucleotide comprising the marker gene, and fragments thereof. cDNA can, optionally, be amplified using any of a variety of polymerase chain reaction methods prior to hybridization with the reference polynucleotide; preferably, it is not amplified.
  • Familial history: As used herein, the term “familial history” typically refers to occurrence of events (e.g., disease related disorder or mutation carrier) relating to an individual's immediate family members including parents and siblings. Family history may also include grandparents and other relatives.
  • Flanking: As used herein, the term “flanking” is meant that a primer hybridizes to a target nucleic acid adjoining a region of interest sought to be amplified on the target. The skilled artisan will understand that preferred primers are pairs of primers that hybridize 3′ from a region of interest, one on each strand of a target double stranded DNA molecule, such that nucleotides may be add to the 3′ end of the primer by a suitable DNA polymerase. For example, primers that flank mutant sequences do not actually anneal to the mutant sequence but rather anneal to a sequence that adjoins the mutant sequence. In some cases, primers that flank an exon are generally designed not to anneal to the exon sequence but rather to anneal to sequence that adjoins the exon (e.g. intron sequence). However, in some cases, amplification primer may be designed to anneal to the exon sequence.
  • Gene: As used herein a gene is a unit of heredity. Generally, a gene is a portion of DNA that encodes a protein or a functional RNA. A gene is a locatable region of genomic sequence corresponding to a unit of inheritance. A gene may be associated with regulatory regions, transcribed regions, and or other functional sequence regions.
  • Genotype: As used herein, the term “genotype” refers to the genetic constitution of an organism. More specifically, the term refers to the identity of alleles present in an individual. “Genotyping” of an individual or a DNA sample refers to identifying the nature, in terms of nucleotide base, of the two alleles possessed by an individual at a known polymorphic site.
  • Gene regulatory element: As used herein a gene regulatory element or regulatory sequence is a segment of DNA where regulatory proteins, such as transcription factors, bind to regulate gene expression. Such regulatory regions are often upstream of the gene being regulated.
  • Healthy individual: As used herein, the term “healthy individual” or “control” refers to a subject has not been diagnosed with the syndrome and/or disease of interest.
  • Heterozygous: As used herein, the term “heterozygous” or “HET” refers to an individual possessing two different alleles of the same gene. As used herein, the term “heterozygous” encompasses “compound heterozygous” or “compound heterozygous mutant.” As used herein, the term “compound heterozygous” refers to an individual possessing two different alleles. As used herein, the term “compound heterozygous mutant” refers to an individual possessing two different copies of an allele, such alleles are characterized as mutant forms of a gene.
  • Homozygous: As used herein, the term “homozygous” refers to an individual possessing two copies of the same allele. As used herein, the term “homozygous mutant” refers to an individual possessing two copies of the same allele, such allele being characterized as the mutant form of a gene.
  • Hybridize: As used herein, the term “hybridize” or “hybridization” refers to a process where two complementary nucleic acid strands anneal to each other under appropriately stringent conditions. Oligonucleotides or probes suitable for hybridizations typically contain 10-100 nucleotides in length (e.g., 18-50, 12-70, 10-30, 10-24, 18-36 nucleotides in length). Nucleic acid hybridization techniques are well known in the art. Those skilled in the art understand how to estimate and adjust the stringency of hybridization conditions such that sequences having at least a desired level of complementary will stably hybridize, while those having lower complementary will not. For examples of hybridization conditions and parameters, see, e.g., Sambrook, et al., 1989, Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Press, Plainview, N.Y.; Ausubel, F. M. et al. 1994, Current Protocols in Molecular Biology. John Wiley & Sons, Secaucus, N.J.
  • Identity or percent identical: As used herein, the terms “identity” or “percent identical” refers to sequence identity between two amino acid sequences or between two nucleic acid sequences. Percent identity can be determined by aligning two sequences and refers to the number of identical residues (i.e., amino acid or nucleotide) at positions shared by the compared sequences. Sequence alignment and comparison may be conducted using the algorithms standard in the art (e.g. Smith and Waterman, 1981, Adv. Appl. Math. 2:482; Needleman and Wunsch, 1970, J. Mol. Biol. 48:443; Pearson and Lipman, 1988, Proc. Natl. Acad. Sci., USA, 85:2444) or by computerized versions of these algorithms (Wisconsin Genetics Software Package Release 7.0, Genetics Computer Group, 575 Science Drive, Madison, Wis.) publicly available as BLAST and FASTA. Also, ENTREZ, available through the National Institutes of Health, Bethesda Md., may be used for sequence comparison. In other cases, commercially available software, such as GenomeQuest, may be used to determine percent identity. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., BLASTN; available at the Internet site for the National Center for Biotechnology Information) may be used. In one embodiment, the percent identity of two sequences may be determined using GCG with a gap weight of 1, such that each amino acid gap is weighted as if it were a single amino acid mismatch between the two sequences. Or, the ALIGN program (version 2.0), which is part of the GCG (Accelrys, San Diego, Calif.) sequence alignment software package may be used.
  • As used herein, the term at least 90% identical thereto includes sequences that range from 90 to 100% identity to the indicated sequences and includes all ranges in between. Thus, the term at least 90% identical thereto includes sequences that are 91, 91.5, 92, 92.5, 93, 93.5, 94, 94.5, 95, 95.5, 96, 96.5, 97, 97.5, 98, 98.5, 99, 99.5 percent identical to the indicated sequence. Similarly, the term “at least 70% identical includes sequences that range from 70 to 100% identical, with all ranges in between. The determination of percent identity is determined using the algorithms described herein. Insertion or addition: As used herein, the term “insertion” or “addition” refers to a change in an amino acid or nucleotide sequence resulting in the addition of one or more amino acid residues or nucleotides, respectively, as compared to the naturally occurring molecule.
  • In vitro: As used herein, the term “in vitro” refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, etc., rather than within a multi-cellular organism.
  • In vivo: As used herein, the term “in vivo” refers to events that occur within a multi-cellular organism such as a human or a non-human animal.
  • Isolated: As used herein, the term “isolated” refers to a substance and/or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature and/or in an experimental setting), and/or (2) produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98%, about 99%, substantially 100%, or 100% of the other components with which they were initially associated. In some embodiments, isolated agents are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, substantially 100%, or 100% pure. As used herein, a substance is “pure” if it is substantially free of other components. As used herein, the term “isolated cell” refers to a cell not contained in a multi-cellular organism.
  • Labeled: The terms “labeled” and “labeled with a detectable agent or moiety” are used herein interchangeably to specify that an entity (e.g., a nucleic acid probe, antibody, etc.) can be measured by detection of the label (e.g., visualized, detection of radioactivity and the like) for example following binding to another entity (e.g., a nucleic acid, polypeptide, etc.). The detectable agent or moiety may be selected such that it generates a signal which can be measured and whose intensity is related to (e.g., proportional to) the amount of bound entity. A wide variety of systems for labeling and/or detecting proteins and peptides are known in the art. Labeled proteins and peptides can be prepared by incorporation of, or conjugation to, a label that is detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical, chemical or other means. A label or labeling moiety may be directly detectable (i.e., it does not require any further reaction or manipulation to be detectable, e.g., a fluorophore is directly detectable) or it may be indirectly detectable (i.e., it is made detectable through reaction or binding with another entity that is detectable, e.g., a hapten is detectable by immunostaining after reaction with an appropriate antibody comprising a reporter such as a fluorophore). Suitable detectable agents include, but are not limited to, radionucleotides, fluorophores, chemiluminescent agents, microparticles, enzymes, colorimetric labels, magnetic labels, haptens, molecular beacons, aptamer beacons, and the like.
  • Micro RNA: As used herein microRNAs (miRNAs) are short (20-24 nucleotide) non-coding RNAs that are involved in post-transcriptional regulation of gene expression. microRNA can affect both the stability and translation of mRNAs. For example, microRNAs can bind to complementary sequences in the 3′UTR of target mRNAs and cause gene silencing. miRNAs are transcribed by RNA polymerase II as part of capped and polyadenylated primary transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The primary transcript can be cleaved by the Drosha ribonuclease III enzyme to produce an approximately 70-nucleotide stem-loop precursor miRNA (pre-miRNA), which can further be cleaved by the cytoplasmic Dicer ribonuclease to generate the mature miRNA and antisense miRNA star (miRNA*) products. The mature miRNA can be incorporated into a RNA-induced silencing complex (RISC), which can recognize target mRNAs through imperfect base pairing with the miRNA and most commonly results in translational inhibition or destabilization of the target mRNA.
  • Multiplex PCR: As used herein, the term “multiplex PCR” refers to concurrent amplification of two or more regions which are each primed using a distinct primers pair.
  • Multiplex ASPE: As used herein, the term “multiplex ASPE” refers to an assay combining multiplex PCR and allele specific primer extension (ASPE) for detecting polymorphisms. Typically, multiplex PCR is used to first amplify regions of DNA that will serve as target sequences for ASPE primers. See the definition of allele specific primer extension.
  • Mutation and/or variant: As used herein, the terms mutation and variant are used interchangeably to describe a nucleic acid or protein sequence change. The term “mutant” as used herein refers to a mutated, or potentially non-functional form of a gene. The term includes any mutation that renders a gene not functional from a point mutation to large chromosomal rearrangements as is known in the art.
  • Nucleic acid: As used herein, a “nucleic acid” is a polynucleotide such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA). The term is used to include single-stranded nucleic acids, double-stranded nucleic acids, and RNA and DNA made from nucleotide or nucleoside analogues.
  • Polypeptide or protein: As used herein, the term “polypeptide” and/or “protein” refers to a polymer of amino acids, and not to a specific length. Thus, peptides, oligopeptides and proteins are included within the definition of polypeptide and/or protein. “Polypeptide” and “protein” are used interchangeably herein to describe protein molecules that may comprise either partial or full-length proteins. The term “peptide” is used to denote a less than full-length protein or a very short protein unless the context indicates otherwise.
  • As is known in the art, “proteins”, “peptides,” “polypeptides” and “oligopeptides” are chains of amino acids (typically L-amino acids) whose alpha carbons are linked through peptide bonds formed by a condensation reaction between the carboxyl group of the alpha carbon of one amino acid and the amino group of the alpha carbon of another amino acid. Typically, the amino acids making up a protein are numbered in order, starting at the amino terminal residue and increasing in the direction toward the carboxy terminal residue of the protein. Abbreviations for amino acid residues are the standard 3-letter and/or 1-letter codes used in the art to refer to one of the 20 common L-amino acids.
  • As used herein, a polypeptide or protein “domain” comprises a region along a polypeptide or protein that comprises an independent unit. Domains may be defined in terms of structure, sequence and/or biological activity. In one embodiment, a polypeptide domain may comprise a region of a protein that folds in a manner that is substantially independent from the rest of the protein. Domains may be identified using domain databases such as, but not limited to PFAM, PRODOM, PROSITE, BLOCKS, PRINTS, SBASE, ISREC PROFILES, SAMRT, and PROCLASS.
  • Primer: As used herein, the term “primer” refers to a short single-stranded oligonucleotide capable of hybridizing to a complementary sequence in a nucleic acid sample. Typically, a primer serves as an initiation point for template dependent DNA synthesis. Deoxyribonucleotides can be added to a primer by a DNA polymerase. In some embodiments, such deoxyribonucleotides addition to a primer is also known as primer extension. The term primer, as used herein, includes all forms of primers that may be synthesized including peptide nucleic acid primers, locked nucleic acid primers, phosphorothioate modified primers, labeled primers, and the like. A “primer pair” or “primer set” for a PCR reaction typically refers to a set of primers typically including a “forward primer” and a “reverse primer.” As used herein, a “forward primer” refers to a primer that anneals to the anti-sense strand of dsDNA. A “reverse primer” anneals to the sense-strand of dsDNA.
  • Polymorphism: As used herein, the term “polymorphism” refers to the coexistence of more than one form of a gene or portion thereof.
  • Portion and Fragment: As used herein, the terms “portion” and “fragment” are used interchangeably to refer to parts of a polypeptide, nucleic acid, or other molecular construct.
  • Sense strand vs. anti-sense strand: As used herein, the term “sense strand” refers to the strand of double-stranded DNA (dsDNA) that includes at least a portion of a coding sequence of a functional protein. As used herein, the term “anti-sense strand” refers to the strand of dsDNA that is the reverse complement of the sense strand.
  • Significant difference: As used herein, the term “significant difference” is well within the knowledge of a skilled artisan and will be determined empirically with reference to each particular biomarker. For example, a significant difference in the expression of a biomarker in a subject with the disease or syndrome of interest as compared to a healthy subject is any difference in protein amounts which is statistically significant.
  • Similar or homologue: As used herein, the term “similar” or “homologue” when referring to amino acid or nucleotide sequences means a polypeptide having a degree of homology or identity with the wild-type amino acid sequence. Homology comparisons can be conducted by eye, or more usually, with the aid of readily available sequence comparison programs. These commercially available computer programs can calculate percent homology between two or more sequences (e.g. Wilbur, W. J. and Lipman, D. J., 1983, Proc. Natl. Acad. Sci. USA, 80:726-730). For example, homologous sequences may be taken to include an amino acid sequences which in alternate embodiments are at least 70% identical, 75% identical, 80% identical, 85% identical, 90% identical, 95% identical, 97% identical, or 98% identical to each other.
  • Specific: As used herein, the term “specific,” when used in connection with an oligonucleotide primer, refers to an oligonucleotide or primer, which under appropriate hybridization or washing conditions, is capable of hybridizing to the target of interest and not substantially hybridizing to nucleic acids which are not of interest. Higher levels of sequence identity are preferred and include at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% sequence identity. In some embodiments, a specific oligonucleotide or primer contains at least 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 35, 40, 45, 50, 55, 60, 65, 70, or more bases of sequence identity with a portion of the nucleic acid to be hybridized or amplified when the oligonucleotide and the nucleic acid are aligned.
  • As is known in the art, conditions for hybridizing nucleic acid sequences to each other can be described as ranging from low to high stringency. Generally, highly stringent hybridization conditions refer to washing hybrids in low salt buffer at high temperatures. Hybridization may be to filter bound DNA using hybridization solutions standard in the art such as 0.5 M NaHPO4, 7% sodium dodecyl sulfate (SDS), at 65° C., and washing in 0.25 M NaHPO4, 3.5% SDS followed by washing 0.1×SSC/0.1% SDS at a temperature ranging from room temperature to 68° C. depending on the length of the probe (see e.g. Ausubel, F.M. et al., Short Protocols in Molecular Biology, 4th Ed., Chapter 2, John Wiley & Sons, N.Y). For example, a high stringency wash comprises washing in 6× SSC/0.05% sodium pyrophosphate at 37° C. for a 14 base oligonucleotide probe, or at 48° C. for a 17 base oligonucleotide probe, or at 55° C. for a 20 base oligonucleotide probe, or at 60° C. for a 25 base oligonucleotide probe, or at 65° C. for a nucleotide probe about 250 nucleotides in length. Nucleic acid probes may be labeled with radionucleotides by end-labeling with, for example, [γ-32P]ATP, or incorporation of radiolabeled nucleotides such as [α-32P]dCTP by random primer labeling. Alternatively, probes may be labeled by incorporation of biotinylated or fluorescein labeled nucleotides, and the probe detected using streptavidin or anti-fluorescein antibodies.
  • siRNA: As used herein, siRNA (small inhibitory RNA) is essentially a double-stranded RNA molecule composed of about 20 complementary nucleotides. siRNA is created by the breakdown of larger double-stranded (ds) RNA molecules. siRNA can suppress gene expression by inherently splitting its corresponding mRNA in two by way of the interaction of the siRNA with the mRNA, leading to degradation of the mRNA. siRNAs can also interact with DNA to facilitate chromatin silencing and the expansion of heterochromatin.
  • Subject: As used herein, the term “subject” refers to a human or any non-human animal. A subject can be a patient, which refers to a human presenting to a medical provider for diagnosis or treatment of a disease. A human includes pre and post-natal forms. Also, as used herein, the terms “individual,” “subject” or “patient” includes all warm-blooded animals. In one embodiment the subject is a human. In one embodiment, the individual is a subject who has a disease associated with exposure to inhaled carcinogens or has an enhanced risk of developing a disease associated with exposure to inhaled carcinogens.
  • Substantially: As used herein, the term “substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term “substantially” is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.
  • Substantially complementary: As used herein, the term “substantially complementary” refers to two sequences that can hybridize under stringent hybridization conditions. The skilled artisan will understand that substantially complementary sequences need not hybridize along their entire length. In some embodiments, “stringent hybridization conditions” refer to hybridization conditions at least as stringent as the following: hybridization in 50% formamide, 5×SSC, 50 mM NaH2PO4, pH 6.8, 0.5% SDS, 0.1 mg/mL sonicated salmon sperm DNA, and 5×Denhart's solution at 42° C. overnight; washing with 2×SSC, 0.1% SDS at 45° C.; and washing with 0.2×SSC, 0.1% SDS at 45° C. In some embodiments, stringent hybridization conditions should not allow for hybridization of two nucleic acids which differ over a stretch of 20 contiguous nucleotides by more than two bases.
  • Substitution: As used herein, the term “substitution” refers to the replacement of one or more amino acids or nucleotides by different amino acids or nucleotides, respectively, as compared to the naturally occurring molecule.
  • Suffering from: An individual who is “suffering from” a disease, disorder, and/or condition has been diagnosed with or displays one or more symptoms of the disease, disorder, and/or condition.
  • Susceptible to: An individual who is “susceptible to” a disease, disorder, and/or condition has not been diagnosed with the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may not exhibit symptoms of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.
  • Solid support: The term “solid support” or “support” means a structure that provides a substrate onto which biomolecules may be bound. For example, a solid support may be an assay well (i.e., such as a microtiter plate), or the solid support may be a location on an array, or a mobile support, such as a bead.
  • Upstream and downstream: As used herein, the term “upstream” refers to a residue that is N-terminal to a second residue where the molecule is a protein, or 5′ to a second residue where the molecule is a nucleic acid. Also as used herein, the term “downstream” refers to a residue that is C-terminal to a second residue where the molecule is a protein, or 3′ to a second residue where the molecule is a nucleic acid. Protein, polypeptide and peptide sequences disclosed herein are all listed from N-terminal amino acid to C-terminal acid and nucleic acid sequences disclosed herein are all listed from the 5′ end of the molecule to the 3′ end of the molecule.
  • Overview
  • The disclosure herein provides novel mutations identified in a disease and/or syndrome of interest gene that can be used for more accurate diagnosis of disorders relating to the gene and/or syndrome of interest.
  • In some embodiments, the sample contains protein. In some embodiments, the testing step comprises amino acid sequencing. In some embodiments, the testing step comprises performing an immunoassay using one or more antibodies that specifically recognize the biomarker of interest. In some embodiments, the testing step comprises protease digestion (e.g., trypsin digestion). In some embodiments, the testing step further comprises performing 2D-gel electrophoresis.
  • In some embodiments, the sample contains nucleic acid. In some embodiments, the testing step comprises nucleic acid sequencing. In some embodiments, the testing step comprises hybridization. In some embodiments, the hybridization is performed using one or more oligonucleotide probes specific for a region in the biomarker of interest. In some embodiments, for detection of mutations, hybridization is performed under conditions sufficiently stringent to disallow a single nucleotide mismatch. In some embodiments, the hybridization is performed with a microarray. In some embodiments, the testing step comprises restriction enzyme digestion. In some embodiments, the testing step comprises PCR amplification. In some embodiments, the PCR amplification is digital PCR amplification. In some embodiments, the testing step comprises primer extension. In some embodiments, the primer extension is single-base primer extension. In some embodiments, the testing step comprises performing a multiplex allele-specific primer extension (ASPE).
  • In some embodiments, the testing step comprises determining the presence of the one or more biomarkers using mass spectrometry. In some embodiments, the mass spectrometric format is selected from among Matrix-Assisted Laser Desorption/Ionization, Time-of-Flight (MALDI-TOF), Electrospray (ES), IR-MALDI, Ion Cyclotron Resonance (ICR), Fourier Transform, and combinations thereof.
  • In some embodiments, the sample is obtained from cells, tissue (e.g., tissue or liquid biopsy) whole blood, liquid or tissue biopsy, cell-free nucleic acid (e.g., DNA or RNA) mouthwash, plasma, serum, urine, stool, saliva, cord blood, chorionic villus sample, chorionic villus sample culture, amniotic fluid, amniotic fluid culture, transcervical lavage fluid, or combination thereof. In further embodiments, the sample is obtained from blood or blood products (e.g., plasma or serum) from a pregnant woman and/or fetal DNA. In certain embodiments, the sample is cell-free nucleic acid (e.g., DNA or RNA) from plasma, amniotic fluid and the like.
  • In some embodiments, the testing step comprises determining the identity of the nucleotide and/or amino acid at a pre-determined position in the biomarker. In some embodiments, the presence of the mutation is determined by comparing the identity of the nucleotide and/or amino acid at the pre-determined position to a control.
  • In embodiments, the method may comprise performing the assay (e.g., sequencing) in a plurality of individuals to determine the statistical significance of the association.
  • In another aspect, the disclosure provides reagents for detecting the biomarker of interest such as, but not limited to a nucleic acid probe that specifically binds to the biomarker (e.g., a mutation in a DNA sequence), or an array containing one or more probes that specifically bind to the biomarker. In some embodiments, the disclosure provides an antibody that specifically binds to the biomarker. In some embodiments, the disclosure provides a kit for comprising one or more of such reagents. In some embodiments, the one or more reagents are provided in a form of microarray. In some embodiments, the kit further comprises reagents for primer extension. In some embodiments, the kit further comprises a control indicative of a healthy individual. In some embodiments, the kit further comprises an instructions on how to determine if an individual has the syndrome or disease of interest based on the biomarker of interest.
  • In some cases, the amount of the one or more biomarkers may, in certain embodiments, be detected by: (a) detecting the amount of a polypeptide or protein which is regulated by said one or more biomarker; (b) detecting the amount of a polypeptide or protein which regulates said biomarker; or (c) detecting the amount of a metabolite of the biomarker.
  • In still another aspect, the disclosure herein provides a computer readable medium encoding information corresponding detection of the biomarker.
  • Methods and Compositions for Diagnosing Diseases Related to Exposure to Inhaled Carcinogens
  • Embodiments of the present disclosure comprise compositions and methods for diagnosing presence or increased risk of developing a disease or diseases associated with inhalation of a carcinogen and/or exposure to an inhaled carcinogen. The methods and compositions of the present disclosure may be used to obtain or provide genetic information from a subject in order to objectively diagnose the presence or increased risk for that subject, or other subjects to develop such diseases. The methods and compositions may be embodied in a variety of ways.
  • In one embodiment, disclosed is a method to detect biomarkers associated with at least one of lung cancer (LC) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount, or a mutation in, a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRC5), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAM5), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNA5), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine receptor 4 (CXCR4), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), cytochrome P450 family 1 subfamily B member 1 (CYP1B1), epidermal growth factor receptor (EGFR), epoxide hydrolase 1 (EPHX1), erb-b2 receptor tyrosine kinase 2 (ERBB2), fructose-bisphosphatase 1 (FBP1), fascin actin-bundling protein 1 (FSCN1), glutathione S-transferase pi 1 (GSTP1), interleukin 10 (IL10), integrin subunit alpha 11 (ITGA11), KRAS proto-oncogene, GTPase (KRAS), keratin 19 (KRT19), leucine aminopeptidase 3 (LAP3), MDM2 proto-oncogene (MDM2), v-myc avian myelocytomatosis viral oncogene homolog (MYC), p21 (RAC1) activated kinase 1 (PAK1), poly(ADP-ribose) polymerase 1 (PARP1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), protein kinase N1 (PKN1), phosphatase and tensin homolog (PTEN), signal transducer and activator of transcription 3 (STAT3), serine/threonine kinase 11 (STK11), tumor protein p53 (TP53), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor C (VEGFC), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or X-ray repair cross complementing 1 (XRCC1).
  • In one embodiment, disclosed is a method to detect biomarkers associated with chronic obstructive pulmaonary disease (COPD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of adiponectin, C1Q and collagen domain containing (ADIPOQ), adrenoceptor beta 2 (ADRB2), advanced glycosylation end product-specific receptor (AGER), CD4 molecule (CD4), cystic fibrosis transmembrane conductance regulator (CFTR), cholinergic receptor nicotinic alpha 3 subunit (CHRNA3), cystatin C (CST3), C-X-C motif chemokine ligand 8 (CXCL8), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), D-box binding PAR bZIP transcription factor (DBP), epidermal growth factor receptor (EGFR), elastin (ELN), erythropoietin (EPO), glutathione S-transferase pi 1 (GSTP1), histone deacetylase 2 (HDAC2), high mobility group box 1 (HMGB1), 5-hydroxytryptamine receptor 4 (HTR4), immunoglobulin heavy constant epsilon (IGHE), interleukin 10 (IL10), interleukin 13 (IL13), interleukin 1 beta (IL1B), laminin subunit alpha 1 (LAMA1), leptin (LEP), membrane metallo-endopeptidase (MME), matrix metallopeptidase 12 (MMP12), matrix metallopeptidase 25 (MMP25), serpin family A member 1 (SERPINA1), sirtuin 1 (SIRT1), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or vascular endothelial growth factor A (VEGFA).
  • In one embodiment, disclosed is a method to detect biomarkers associated with cardovascular disease (CVD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of ABO blood group (transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase) (ABO), angiotensin I converting enzyme 2 (ACE2), angiotensinogen (AGT), albumin (ALB), apelin (APLN), apolipoprotein A1 (APOA1), apolipoprotein A2 (APOA2), apolipoprotein B (APOB), apolipoprotein E (APOE), caspase 1 (CASP1), CD36 molecule (CD36), C-reactive protein, pentraxin-related (CRP), elongation factor for RNA polymerase II (ELL), coagulation factor II, thrombin (F2), intercellular adhesion molecule 1 (ICAM1), interleukin 1 beta (IL1B), low density lipoprotein receptor (LDLR), leptin (LEP), myeloperoxidase (MPO), nitric oxide synthase 3 (NOS3), period circadian clock 1 (PERI1), prolactin (PRL), tumor necrosis factor (TNF), troponin C1, slow skeletal and cardiac type (TNNC1), troponin 13, cardiac type (TNNI3), troponin T2, cardiac type (TNNT2), vascular endothelial growth factor A (VEGFA), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or von Willebrand factor (VWF).
  • Additionally and/or alternatively, the method may include measurement of at least one normalization (e.g., housekeeping) gene. In one non-limiting embodiment, the housekeeping gene may be glyceraldehyde 3-phosphate dehydrogenase. Or, other house keeping genes may be used. Or, measurement of various combinations of these markers may be performed.
  • Additionally and/or alternatively, other biomarkers may be measured.
  • As disclosed herein, a variety of methods may be used to measure the biomarkers of interest. In one embodiment, the measuring comprises measuring peptide or polypeptide biomarkers. For example, in one embodiment, the measuring comprises an immunoassay. Or, the measuring may comprise flow cytometry. Or, as discussed in detail herein, nucleic acid methods may be used.
  • A variety of sample types may be used. In certain embodiments, the sample comprises blood, serum, plasma, cell-free nucleic acid (e.g., DNA or RNA), or a liquid or tissue biopsy.
  • In certain embodiments, the disclosure provides a method of identifying a marker associated with exposure to an inhaled carcinogen in an individual. The method may comprise the steps of identifying at least one marker having increased or decreased expression in diseases associated with inhaled carcinogens such as, but not limited to lung cancer (LC), chronic obstructive pulmaonary disease (COPD) and/or cardiovascular disease (CVD) as compared to a control individual or population. In an embodiment, the control is a healthy individual with no detected or detectable lung or cardiovascular pathology. In some embodiments, the control is a disease control. Such disease controls may include individuals with lung or heart disease that is not related to exposure to an inhaled carcinogen.
  • In other embodiments, the disclosure provides a method to detect the presence of, or susceptibility to a disease associated with exposure to an inhaled carcinogen in an individual.comprising:obtaining a sample from the individual; measuring the amount of at least one marker associated with at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD), in the sample; and comparing the expression of, and/or the presence of a mutation in a gene for, the at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD) in the sample with a control value for each of the markers. In an embodiment, the control value is determined from a healthy individual or individuals with no detected or detectable lung or cardiovascular pathology. In some embodiments, the control is a disease control. Such disease controls may include individuals with lung or heart disease that is not related to exposure to an inhaled carcinogen.
  • The method may comprise detecting biomarkers associated with at least one of lung cancer (LC) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount, or a mutation in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for or regulates expression of the gene for at least one of anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRC5), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAM5), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNA5), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine receptor 4 (CXCR4), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), cytochrome P450 family 1 subfamily B member 1 (CYP1B1), epidermal growth factor receptor (EGFR), epoxide hydrolase 1 (EPHX1), erb-b2 receptor tyrosine kinase 2 (ERBB2), fructose-bisphosphatase 1 (FBP1), fascin actin-bundling protein 1 (FSCN1), glutathione S-transferase pi 1 (GSTP1), interleukin 10 (IL10), integrin subunit alpha 11 (ITGA11), KRAS proto-oncogene, GTPase (KRAS), keratin 19 (KRT19), leucine aminopeptidase 3 (LAP3), MDM2 proto-oncogene (MDM2), v-myc avian myelocytomatosis viral oncogene homolog (MYC), p21 (RAC1) activated kinase 1 (PAK1), poly(ADP-ribose) polymerase 1 (PARP1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), protein kinase N1 (PKN1), phosphatase and tensin homolog (PTEN), signal transducer and activator of transcription 3 (STAT3), serine/threonine kinase 11 (STK11), tumor protein p53 (TP53), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor C (VEGFC), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or X-ray repair cross complementing 1 (XRCC1).
  • Additionally and/or alternatively, the method may comprise detecting biomarkers associated with chronic obstructive pulmaonary disease (COPD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of adiponectin, C1Q and collagen domain containing (ADIPOQ), adrenoceptor beta 2 (ADRB2), advanced glycosylation end product-specific receptor (AGER), CD4 molecule (CD4), cystic fibrosis transmembrane conductance regulator (CFTR), cholinergic receptor nicotinic alpha 3 subunit (CHRNA3), cystatin C (CST3), C-X-C motif chemokine ligand 8 (CXCL8), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), D-box binding PAR bZIP transcription factor (DBP), epidermal growth factor receptor (EGFR), elastin (ELN), erythropoietin (EPO), glutathione S-transferase pi 1 (GSTP1), histone deacetylase 2 (HDAC2), high mobility group box 1 (HMGB1), 5-hydroxytryptamine receptor 4 (HTR4), immunoglobulin heavy constant epsilon (IGHE), interleukin 10 (IL10), interleukin 13 (IL13), interleukin 1 beta (IL1B), laminin subunit alpha 1 (LAMA1), leptin (LEP), membrane metallo-endopeptidase (MME), matrix metallopeptidase 12 (MMP12), matrix metallopeptidase 25 (MMP25), serpin family A member 1 (SERPINA1), sirtuin 1 (SIRT1), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or vascular endothelial growth factor A (VEGFA).
  • Additionally and/or alternatively the method may comprise detecting biomarkers associated with cardovascular disease (CVD) in an individual comprising the steps of: obtaining a sample from the individual; and measuring the amount of the marker, and/or the amount and/or a mutation in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of ABO blood group (transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase) (ABO), angiotensin I converting enzyme 2 (ACE2), angiotensinogen (AGT), albumin (ALB), apelin (APLN), apolipoprotein A1 (APOA1), apolipoprotein A2 (APOA2), apolipoprotein B (APOB), apolipoprotein E (APOE), caspase 1 (CASP1), CD36 molecule (CD36), C-reactive protein, pentraxin-related (CRP), elongation factor for RNA polymerase II (ELL), coagulation factor II, thrombin (F2), intercellular adhesion molecule 1 (ICAM1), interleukin 1 beta (IL1B), low density lipoprotein receptor (LDLR), leptin (LEP), myeloperoxidase (MPO), nitric oxide synthase 3 (NOS3), period circadian clock 1 (PER1), prolactin (PRL), tumor necrosis factor (TNF), troponin C1, slow skeletal and cardiac type (TNNC1), troponin I3, cardiac type (TNNI3), troponin T2, cardiac type (TNNT2), vascular endothelial growth factor A (VEGFA), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or von Willebrand factor (VWF).
  • Additionally and/or alternatively, other biomarkers may be measured.
  • As disclosed herein, a variety of methods may be used to measure the biomarkers of interest. In one embodiment, the measuring comprises measuring peptide or polypeptide biomarkers. For example, in one embodiment, the measuring comprises an immunoassay. Or, the measuring may comprise flow cytometry. Or, as discussed in detail herein, nucleic acid methods may be used.
  • A variety of sample types may be used. In certain embodiments, the sample comprises blood, serum, plasma, a liquid or tissue biopsy, or cell-free nucleic acid (e.g., DNA or RNA).
  • Yet other embodiments comprise a composition to detect biomarkers associated with a disease associated with exposure to inhaled carcinogins in an individual. In certain embodiments, the composition comprises reagents that quantify the amount of the marker, and/or the amount, or a mutation in, a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one marker associated with at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD), in the sample.
  • The composition may comprise reagents for detecting biomarkers associated with lung cancer (LC) in an individual by measuring the amount of the marker, and/or the amount, or a mutation in, a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRC5), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAM5), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNA5), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine receptor 4 (CXCR4), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), cytochrome P450 family 1 subfamily B member 1 (CYP1B1), epidermal growth factor receptor (EGFR), epoxide hydrolase 1 (EPHX1), erb-b2 receptor tyrosine kinase 2 (ERBB2), fructose-bisphosphatase 1 (FBP1), fascin actin-bundling protein 1 (FSCN1), glutathione S-transferase pi 1 (GSTP1), interleukin 10 (IL10), integrin subunit alpha 11 (ITGA11), KRAS proto-oncogene, GTPase (KRAS), keratin 19 (KRT19), leucine aminopeptidase 3 (LAP3), MDM2 proto-oncogene (MDM2), v-myc avian myelocytomatosis viral oncogene homolog (MYC), p21 (RAC1) activated kinase 1 (PAK1), poly(ADP-ribose) polymerase 1 (PARP1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), protein kinase N1 (PKN1), phosphatase and tensin homolog (PTEN), signal transducer and activator of transcription 3 (STAT3), serine/threonine kinase 11 (STK11), tumor protein p53 (TP53), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor C (VEGFC), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or X-ray repair cross complementing 1 (XRCC1).
  • Additionally and/or alternatively, the composition may comprise reagents for detecting biomarkers associated with chronic obstructive pulmaonary disease (COPD) in an individual by measuring the amount of the marker, and/or the amount, or a mutation in, a nucleic acid (e.g. genomic DNA or mRNA) that encodes for or regulates expression of the gene for at least one of adiponectin, C1Q and collagen domain containing (ADIPOQ), adrenoceptor beta 2 (ADRB2), advanced glycosylation end product-specific receptor (AGER), CD4 molecule (CD4), cystic fibrosis transmembrane conductance regulator (CFTR), cholinergic receptor nicotinic alpha 3 subunit (CHRNA3), cystatin C (CST3), C-X-C motif chemokine ligand 8 (CXCL8), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), D-box binding PAR bZIP transcription factor (DBP), epidermal growth factor receptor (EGFR), elastin (ELN), erythropoietin (EPO), glutathione S-transferase pi 1 (GSTP1), histone deacetylase 2 (HDAC2), high mobility group box 1 (HMGB1), 5-hydroxytryptamine receptor 4 (HTR4), immunoglobulin heavy constant epsilon (IGHE), interleukin 10 (IL10), interleukin 13 (IL13), interleukin 1 beta (IL1B), laminin subunit alpha 1 (LAMA1), leptin (LEP), membrane metallo-endopeptidase (MME), matrix metallopeptidase 12 (MMP12), matrix metallopeptidase 25 (MMP25), serpin family A member 1 (SERPINA1), sirtuin 1 (SIRT1), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or vascular endothelial growth factor A (VEGFA).
  • Additionally and/or alternatively, the composition may comprise reagents for detecting biomarkers associated with cardiovascular disease in an individual by measuring the amount of the marker, and/or the amount, or a mutation in, a nucleic acid (e.g. genomic DNA or mRNA) that encodes for or regulates expression of the gene for at least one of ABO blood group (transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase) (ABO), angiotensin I converting enzyme 2 (ACE2), angiotensinogen (AGT), albumin (ALB), apelin (APLN), apolipoprotein A1 (APOA1), apolipoprotein A2 (APOA2), apolipoprotein B (APOB), apolipoprotein E (APOE), caspase 1 (CASP1), CD36 molecule (CD36), C-reactive protein, pentraxin-related (CRP), elongation factor for RNA polymerase II (ELL), coagulation factor II, thrombin (F2), intercellular adhesion molecule 1 (ICAM1), interleukin 1 beta (IL1B), low density lipoprotein receptor (LDLR), leptin (LEP), myeloperoxidase (MPO), nitric oxide synthase 3 (NOS3), period circadian clock 1 (PERI1), prolactin (PRL), tumor necrosis factor (TNF), troponin C1, slow skeletal and cardiac type (TNNC1), troponin 13, cardiac type (TNNI3), troponin T2, cardiac type (TNNT2), vascular endothelial growth factor A (VEGFA), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or von Willebrand factor (VWF).
  • Additionally and/or alternatively, the composition may include reagents for the measurement of at least one normalization (e.g., housekeeping) gene. In one non-limiting embodiment, the housekeeping gene may be glyceraldehyde 3-phosphate dehydrogenase. Or, other house keeping genes may be used. Or, measurement of various combinations of these markers may be performed. In an embodiment, the composition may comprise reagents for measuring the value of the markers of interest in a control. The control value is determined from a healthy individual or individuals with no detected or detectable lung or cardiovascular pathology. In some embodiments, the control is a disease control. Such disease controls may include individuals with lung or heart disease that is not related to exposure to an inhaled carcinogen.
  • For example, as described in detail herein the composition may comprise reagents to measure peptide or polypeptide biomarkers. In one embodiment, the composition comprises reagents to perform an immunoassay. Or, the composition may comprise reagents to perform flow cytometry. Or, as discussed in detail herein, the composition may comprise reagents to determine the presence of a particular sequence and/or expression level of a nucleic acid.
  • Other embodiments include kits that contain at least some of the compositions disclosed herein and/or reagents for performing the methods disclosed herein. Such kits may include computer-readable media comprising instructions and/or other information for performing the methods. In an embodiment, the kit may comprise reagents for measuring the value of the marker of interest in a control, or a control value for the marker of interest. The control value may be determined from a healthy individual or individuals with no detected or detectable lung or cardiovascular pathology. In some embodiments, the control is a disease control. Such disease controls may include individuals with lung or heart disease that is not related to exposure to an inhaled carcinogen.
  • Other embodiments comprise instructions or other information and/or computer-readable media comprising instructions and/or other information for performing the methods independent of a kit or reagents therein.
  • Peptide, Polypeptide and Protein Assays
  • In certain embodiments, the biomarker of interest is detected at the protein (or peptide or polypeptide level), that is, a gene product is analyzed. For example, a protein or fragment thereof can be analyzed by amino acid sequencing methods, or immunoassays using one or more antibodies that specifically recognize one or more epitopes present on the biomarker of interest, or in some cases specific to a mutation of interest. Proteins can also be analyzed by protease digestion (e.g., trypsin digestion) and, in some embodiments, the digested protein products can be further analyzed by 2D-gel electrophoresis.
  • Antibody Detection
  • Specific antibodies that bind the biomarker of interest can be employed in any of a variety of methods known in the art. Antibodies against particular epitopes, polypeptides, and/or proteins can be generated using any of a variety of known methods in the art. For example, the epitope, polypeptide, or protein against which an antibody is desired can be produced and injected into an animal, typically a mammal (such as a donkey, mouse, rabbit, horse, chicken, etc.), and antibodies produced by the animal can be collected from the animal. Monoclonal antibodies can also be produced by generating hybridomas that express an antibody of interest with an immortal cell line.
  • In some embodiments, antibodies are labeled with a detectable moiety as described herein.
  • Antibody detection methods are well known in the art including, but are not limited to, enzyme-linked immunoadsorbent assays (ELISAs) and Western blots. Some such methods are amenable to being performed in an array format.
  • For example, in some embodiments, the biomarker of interest is detected using a first antibody (or antibody fragment) that specifically recognizes the biomarker. The antibody may be labeled with a detectable moiety (e.g., a chemiluminescent molecule), an enzyme, or a second binding agent (e.g., streptavidin). Or, the first antibody may be detected using a second antibody, as is known in the art.
  • In certain embodiments, the method may further comprise adding a capture support, the capture support comprising at least one capture support binding agent that recognizes and binds to the biomarker so as to immobilize the biomarker on the capture support. The method may, in certain embodiments, further comprise adding a second binding agent that can specifically recognize and bind to at least some of the plurality binding agent molecules on the capture support. In an embodiment, the binding agent that can specifically recognize and bind to at least some of the plurality binding agent molecules on the capture support is a soluble binding agent (e.g., a secondary antibody). The second binding agent may be labeled (e.g., with an enzyme) such that binding of the biomarker of interest is measured by adding a substrate for the enzyme and quantifying the amount of product formed.
  • In an embodiment, the capture solid support may be an assay well (i.e., such as a microtiter plate). Or, the capture solid support may be a location on an array, or a mobile support, such as a bead. Or the capture support may be a filter.
  • In some cases, the biomarker may be allowed to complex with a first binding agent (e.g., primary antibody specific for the biomarker and labeled with detectable moiety) and a second binding agent (e.g., a secondary antibody that recognizes the primary antibody or a second primary antibody), where the second binding agent is complexed to a third binding agent (e.g., biotin) that can then interact with a capture support (e.g., magnetic bead) having a reagent (e.g., streptavidin) that recognizes the third binding agent linked to the capture support. The complex (labeled primary antibody: biomarker: second primary antibody-biotin: streptavidin-bead may then be captured using a magnet (e.g., a magnetic probe) to measure the amount of the complex.
  • A variety of binding agents may be used in the methods of the disclosure. For example, the binding agent attached to the capture support, or the second antibody, may be either an antibody or an antibody fragment that recognizes the biomarker. Or, the binding agent may comprise a protein that binds a non-protein target (i.e., such as a protein that specifically binds to a small molecule biomarker, or a receptor that binds to a protein).
  • In certain embodiments, the solid supports may be treated with a passivating agent. For example, in certain embodiments the biomarker of interest may be captured on a passivated surface (i.e., a surface that has been treated to reduce non-specific binding). One such passivating agent is BSA. Additionally and/or alternatively, where the binding agent used is an antibody, the solid supports may be coated with protein A, protein G, protein A/G, protein L, or another agent that binds with high affinity to the binding agent (e.g., antibody). These proteins bind the Fc domain of antibodies and thus can orient the binding of antibodies that recognize the protein or proteins of interest.
  • Nucleic Acid Assays
  • In certain embodiments, the biomarkers disclosed herein are detected at the nucleic acid level. In one embodiment, the disclosure comprises methods for diagnosing the presence or an increased risk of developing the syndrome or disease of interest (e.g., diseases associated with exposure to inhaled carcinogens) in a subject. The method may comprise the steps of obtaining a nucleic acid from a tissue or body fluid sample from a subject and conducting an assay to identify whether there is a variant sequence (i.e., a mutation) in the subject's nucleic acid. In certain embodiments, the method may comprise comparing the variant to known variants associated with the syndrome or disease of interest and determining whether the variant is a variant that has been previously identified as being associated with the syndrome or disease of interest. Or, the method may comprise identifying the variant as a new, previously uncharacterized variant. If the variant is a new variant, the method may further comprise performing an analysis to determine whether the mutation is expected to be deleterious to expression of the gene and/or the function of the protein encoded by the gene. The method may further comprise using the variant profile (i.e., the compilation of mutations identified in the subject) to diagnose the presence of the syndrome or disease of interest or an increased risk of developing the syndrome or disease of interest.
  • Nucleic acid analyses can be performed on genomic DNA, cell-free nucleic acid (e.g. DNA or RNA), messenger RNAs, and/or cDNA. Also, in various embodiments, the nucleic acid comprises a gene, an RNA, an exon, an intron, a gene regulatory element, an expressed RNA, an siRNA, or an epigenetic element. Also, regulatory elements, including splice sites, transcription factor binding, A-I editing sites, microRNA binding sites, and functional RNA structure sites may be evaluated for mutations (i.e., variants). Thus, for each of the methods and compositions of the disclosure, the variant may comprise a nucleic acid sequence that encompasses at least one of the following: (1) A-to-I editing sites; (2) splice sites; (3) conserved functional RNA structures; (4) validated transcription factor binding sites (TFBS); (5) microRNA (miRNA) binding sites; (6) polyadenylation sites; (7) known regulatory elements; (8) miRNA genes; (9) small nucleolar RNA genes encoded in the ROIs; and/or (10) ultra-conserved elements across placental mammals.
  • In many embodiments, nucleic acids are extracted from a biological sample. Or, the nucleic acid may comprise cell-free nucleic acid (DNA or RNA). In some embodiments, nucleic acids are analyzed without having been amplified. In some embodiments, nucleic acids are amplified using techniques known in the art (such as polymerase chain reaction (PCR)) and amplified nucleic acids are used in subsequent analyses. Multiplex PCR, in which several amplicons (e.g., from different genomic regions) are amplified at once using multiple sets of primer pairs, may be employed. For example, nucleic acid can be analyzed by sequencing, hybridization, PCR amplification, restriction enzyme digestion, primer extension such as single-base primer extension or multiplex allele-specific primer extension (ASPE), or DNA sequencing. In some embodiments, nucleic acids are amplified in a manner such that the amplification product for a wild-type allele differs in size from that of a mutant allele. Thus, presence or absence of a particular mutant allele can be determined by detecting size differences in the amplification products, e.g., on an electrophoretic gel. For example, deletions or insertions of gene regions may be particularly amenable to using size-based approaches.
  • Certain exemplary nucleic acid analysis methods are described in detail below.
  • Allele-Specific Amplification
  • In some embodiments, for example, where the biomarker for the disease and/or syndrome of interest is a mutation, a biomarker is detected using an allele-specific amplification assay. This approach is variously referred to as PCR amplification of specific allele (PASA) (Sarkar, et al., 1990 Anal. Biochem. 186:64-68), allele-specific amplification (ASA) (Okayama, et al., 1989 J. Lab. Clin. Med. 114:105-113), allele-specific PCR (ASPCR) (Wu, et al. 1989 Proc. Natl. Acad. Sci. USA. 86:2757-2760), and amplification-refractory mutation system (ARMS) (Newton, et al., 1989 Nucleic Acids Res. 17:2503-2516). The entire contents of each of these references is incorporated herein. This method is applicable for single base substitutions as well as micro deletions/insertions.
  • For example, for PCR-based amplification methods, amplification primers may be designed such that they can distinguish between different alleles (e.g., between a wild-type allele and a mutant allele). Thus, the presence or absence of amplification product can be used to determine whether a gene mutation is present in a given nucleic acid sample. In some embodiments, allele specific primers can be designed such that the presence of amplification product is indicative of the gene mutation. In some embodiments, allele specific primers can be designed such that the absence of amplification product is indicative of the gene mutation.
  • In some embodiments, two complementary reactions are used. One reaction employs a primer specific for the wild type allele (“wild-type-specific reaction”) and the other reaction employs a primer for the mutant allele (“mutant-specific reaction”). The two reactions may employ a common second primer. PCR primers specific for a particular allele (e.g., the wild-type allele or mutant allele) generally perfectly match one allelic variant of the target, but are mismatched to other allelic variant (e.g., the mutant allele or wild-type allele). The mismatch may be located at/near the 3′ end of the primer, leading to preferential amplification of the perfectly matched allele. Whether an amplification product can be detected from one or in both reactions indicates the absence or presence of the mutant allele. Detection of an amplification product only from the wild-type-specific reaction indicates presence of the wild-type allele only (e.g., homozygosity of the wild-type allele). Detection of an amplification product in the mutant-specific reaction only indicates presence of the mutant allele only (e.g. homozygosity of the mutant allele). Detection of amplification products from both reactions indicate (e.g., a heterozygote). As used herein, this approach will be referred to as “allele specific amplification (ASA).”
  • Allele-specific amplification can also be used to detect duplications, insertions, or inversions by using a primer that hybridizes partially across the junction. The extent of junction overlap can be varied to allow specific amplification.
  • Amplification products can be examined by methods known in the art, including by visualizing (e.g., with one or more dyes) bands of nucleic acids that have been migrated (e.g., by electrophoresis) through a gel to separate nucleic acids by size.
  • Allele-Specific Primer Extension
  • In some embodiments, an allele-specific primer extension (ASPE) approach is used to detect a gene mutations. ASPE employs allele-specific primers that can distinguish between alleles (e.g., between a mutant allele and a wild-type allele) in an extension reaction such that an extension product is obtained only in the presence of a particular allele (e.g., mutant allele or wild-type allele). Extension products may be detectable or made detectable, e.g., by employing a labeled deoxynucleotide in the extension reaction. Any of a variety of labels are compatible for use in these methods, including, but not limited to, radioactive labels, fluorescent labels, chemiluminescent labels, enzymatic labels, etc. In some embodiments, a nucleotide is labeled with an entity that can then be bound (directly or indirectly) by a detectable label, e.g., a biotin molecule that can be bound by streptavidin-conjugated fluorescent dyes. In some embodiments, reactions are done in multiplex, e.g., using many allele-specific primers in the same extension reaction.
  • In some embodiments, extension products are hybridized to a solid or semi-solid support, such as beads, matrix, gel, among others. For example, the extension products may be tagged with a particular nucleic acid sequence (e.g., included as part of the allele-specific primer) and the solid support may be attached to an “anti-tag” (e.g., a nucleic acid sequence complementary to the tag in the extension product). Extension products can be captured and detected on the solid support. For example, beads may be sorted and detected. One such system that can be employed in this manner is the LUMINEX™ MAP system, which can be adapted for cystic fibrosis mutation detection by TM Bioscience and is sold commercially as a universal bead array (TAG-IT™)
  • Single Nucleotide Primer Extension
  • In some embodiments, a single nucleotide primer extension (SNuPE) assay is used, in which the primer is designed to be extended by only one nucleotide. In such methods, the identity of the nucleotide just downstream of the 3′ end of the primer is known and differs in the mutant allele as compared to the wild-type allele. SNuPE can be performed using an extension reaction in which the only one particular kind of deoxynucleotide is labeled (e.g., labeled dATP, labeled dCTP, labeled dGTP, or labeled dTTP). Thus, the presence of a detectable extension product can be used as an indication of the identity of the nucleotide at the position of interest (e.g., the position just downstream of the 3′ end of the primer), and thus as an indication of the presence or absence of a mutation at that position. SNuPE can be performed as described in U.S. Pat. Nos. 5,888,819; 5,846,710; 6,280,947; 6,482,595; 6,503,718; 6,919,174; Piggee, C. et al. Journal of Chromatography A 781 (1997), p. 367-375 (“Capillary Electrophoresis for the Detection of Known Point Mutations by Single-Nucleotide Primer Extension and Laser-Induced Fluorescence Detection”); Hoogendoorn, B. et al., Human Genetics (1999) 104:89-93, (“Genotyping Single Nucleotide Polymorphism by Primer Extension and High Performance Liquid Chromatography”), the entire contents of each of which are herein incorporated by reference.
  • In some embodiments, primer extension can be combined with mass spectrometry for accurate and fast detection of the presence or absence of a mutation. See, U.S. Pat. No. 5,885,775 to Haff et al. (analysis of single nucleotide polymorphism analysis by mass spectrometry); U.S. Pat. No. 7,501,251 to Koster (DNA diagnosis based on mass spectrometry); the teachings of both of which are incorporated herein by reference. Suitable mass spectrometric format includes, but is not limited to, Matrix-Assisted Laser Desorption/Ionization, Time-of-Flight (MALDI-TOF), Electrospray (ES), IR-MALDI, Ion Cyclotron Resonance (ICR), Fourier Transform, and combinations thereof.
  • Oligonucleotide Ligation Assay
  • In some embodiments, an oligonucleotide ligation assay (“OLA” or “OL”) is used. OLA employs two oligonucleotides that are designed to be capable of hybridizing to abutting sequences of a single strand of a target molecules. Typically, one of the oligonucleotides is biotinylated, and the other is detectably labeled, e.g., with a streptavidin-conjugated fluorescent moiety. If the precise complementary sequence is found in a target molecule, the oligonucleotides will hybridize such that their termini abut, and create a ligation substrate that can be captured and detected. See e.g., Nickerson et al. (1990) Proc. Natl. Acad. Sci. U.S.A. 87:8923-8927, Landegren, U. et al. (1988) Science 241:1077-1080 and U.S. Pat. No. 4,998,617, the entire contents of which are herein incorporated by reference in their entirety.
  • Hybridization Approach
  • In some embodiments, nucleic acids are analyzed by hybridization using one or more oligonucleotide probes specific for the biomarker of interest and under conditions sufficiently stringent to disallow a single nucleotide mismatch. In certain embodiments, suitable nucleic acid probes can distinguish between a normal gene and a mutant gene. Thus, for example, one of ordinary skill in the art could use probes of the invention to determine whether an individual is homozygous or heterozygous for a particular allele.
  • Nucleic acid hybridization techniques are well known in the art. Those skilled in the art understand how to estimate and adjust the stringency of hybridization conditions such that sequences having at least a desired level of complementary will stably hybridize, while those having lower complementary will not. For examples of hybridization conditions and parameters, see, e.g., Sambrook, et al., 1989, Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Press, Plainview, N.Y.; Ausubel, F. M. et al. 1994, Current Protocols in Molecular Biology. John Wiley & Sons, Secaucus, N.J.
  • In some embodiments, probe molecules that hybridize to the mutant or wildtype sequences can be used for detecting such sequences in the amplified product by solution phase or, more preferably, solid phase hybridization. Solid phase hybridization can be achieved, for example, by attaching probes to a microchip.
  • Nucleic acid probes may comprise ribonucleic acids and/or deoxyribonucleic acids. In some embodiments, provided nucleic acid probes are oligonucleotides (i.e., “oligonucleotide probes”). Generally, oligonucleotide probes are long enough to bind specifically to a homologous region of the gene of interest, but short enough such that a difference of one nucleotide between the probe and the nucleic acid sample being tested disrupts hybridization. Typically, the sizes of oligonucleotide probes vary from approximately 10 to 100 nucleotides. In some embodiments, oligonucleotide probes vary from 15 to 90, 15 to 80, 15 to 70, 15 to 60, 15 to 50, 15 to 40, 15 to 35, 15 to 30, 18 to 30, or 18 to 26 nucleotides in length. As appreciated by those of ordinary skill in the art, the optimal length of an oligonucleotide probe may depend on the particular methods and/or conditions in which the oligonucleotide probe may be employed.
  • In some embodiments, nucleic acid probes are useful as primers, e.g., for nucleic acid amplification and/or extension reactions. For example, in certain embodiments, the gene sequence being evaluated for a variant comprises the exon sequences. In certain embodiments, the exon sequence and additional flanking sequence (e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55 or more nucleotides of UTR and/or intron sequence) is analyzed in the assay. Or, intron sequences or other non-coding regions may be evaluated for potentially deleterious mutations. Or, portions of these sequences may be used. Such variant gene sequences may include sequences having at least one of the mutations as described herein.
  • Other embodiments of the disclosure provide isolated gene sequences containing mutations that relate to the syndrome and/or disease of interest. Such gene sequences may be used to objectively diagnose the presence or increased risk for a subject to develop diseases associated with exposure to inhaled carcinogens. In certain embodiments, the isolated nucleic acid may contain a non-variant sequence or a variant sequence of any one or combination thereof. For example, in certain embodiments, the gene sequence comprises the exon sequences. In certain embodiments, the exon sequence and additional flanking sequence (e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55 or more nucleotides of UTR and/or intron sequence) is analyzed in the assay. Or, intron sequences or other non-coding regions may be used. Or, portions of these sequences may be used. In certain embodiments, the gene sequence comprises an exon sequence from at least one of the biomarker genes disclosed herein.
  • In some embodiments, nucleic acid probes are labeled with a detectable moiety as described herein.
  • Arrays
  • A variety of the methods mentioned herein may be adapted for use with arrays that allow sets of biomarkers to be analyzed and/or detected in a single experiment. For example, multiple mutations that comprise biomarkers can be analyzed at the same time. In particular, methods that involve use of nucleic acid reagents (e.g., probes, primers, oligonucleotides, etc.) are particularly amenable for adaptation to an array-based platform (e.g., microarray). In some embodiments, an array containing one or more probes specific for detecting mutations in the biomarker of interest.
  • DNA Sequencing
  • In certain embodiments, diagnosis of the biomarker of interest is carried out by detecting variation in the sequence, genomic location or arrangement, and/or genomic copy number of a nucleic acid or a panel of nucleic acids by nucleic acid sequencing.
  • In some embodiments, the method may comprise obtaining a nucleic acid from a tissue or body fluid sample from a subject and sequencing at least a portion of a nucleic acid in order to obtain a sample nucleic acid sequence for at least one gene. In certain embodiments, the method may comprise comparing the variant to known variants associated with a disease associated with exosure to inhaled carcinogens and determining whether the variant is a variant that has been previously identified as being associated with a disease associated with exosure to inhaled carcinogens. Or, the method may comprise identifying the variant as a new, previously uncharacterized variant. If the variant is a new variant, or in some cases for previously characterized (i.e., identified) variants, the method may further comprise performing an analysis to determine whether the mutation is expected to be deleterious to expression of the gene and/or the function of the protein encoded by the gene. The method may further comprise using the variant profile (i.e., a compilation of variants identified in the subject) to diagnose the presence of a disease associated with exposure to inhaled carcinogens or an increased risk of developing such a disease.
  • For example, in certain embodiments, next generation (massively-parallel sequencing) may be used. Or, Sanger sequencing may be used. Or, a combination of next-generation (massively-parallel sequencing) and Sanger sequencing may be used. Additionally and/or alternatively, the sequencing comprises at least one of single-molecule sequencing-by-synthesis. Thus, in certain embodiments, a plurality of DNA samples are analyzed in a pool to identify samples that show a variation. Additionally or alternatively, in certain embodiments, a plurality of DNA samples are analyzed in a plurality of pools to identify an individual sample that shows the same variation in at least two pools.
  • One conventional method to perform sequencing is by chain termination and gel separation, as described by Sanger et al., 1977, Proc Natl Acad Sci U S A, 74:5463-67. Another conventional sequencing method involves chemical degradation of nucleic acid fragments. See, Maxam et al., 1977, Proc. Natl. Acad. Sci., 74:560-564. Also, methods have been developed based upon sequencing by hybridization. See, e.g., Harris et al., U.S. Patent Application Publication No. 20090156412. Each of these references are incorporated by reference in there entireties herein.
  • In other embodiments, sequencing of the nucleic acid is accomplished by massively parallel sequencing (also known as “next generation sequencing”) of single-molecules or groups of largely identical molecules derived from single molecules by amplification through a method such as PCR. Massively parallel sequencing is shown for example in Lapidus et al., U.S. Pat. No. 7,169,560, Quake et al. U.S. Pat. No. 6,818,395, Harris U.S. Pat. No. 7,282,337 and Braslaysky, et al., PNAS (USA), 100: 3960-3964 (2003), the contents of each of which are incorporated by reference herein.
  • In next generation sequencing, PCR or whole genome amplification can be performed on the nucleic acid in order to obtain a sufficient amount of nucleic acid for analysis. In some forms of next generation sequencing, no amplification is required because the method is capable of evaluating DNA sequences from unamplified DNA. Once determined, the sequence and/or genomic arrangement and/or genomic copy number of the nucleic acid from the test sample is compared to a standard reference derived from one or more individuals not known to suffer from a disease associated with exposure to inhaled carcinogens at the time their sample was taken. All differences between the sequence and/or genomic arrangement and/or genomic arrangement and/or copy number of the nucleic acid from the test sample and the standard reference are considered variants.
  • In next generation (massively parallel sequencing), all regions of interest are sequenced together, and the origin of each sequence read is determined by comparison (alignment) to a reference sequence. The regions of interest can be enriched together in one reaction, or they can be enriched separately and then combined before sequencing. In certain embodiments, and as described in more detail in the examples herein, the DNA sequences derived from coding exons of genes included in the assay are enriched by bulk hybridization of randomly fragmented genomic DNA to specific RNA probes. The same adapter sequences are attached to the ends of all fragments, allowing enrichment of all hybridization-captured fragments by PCR with one primer pair in one reaction. Regions that are less efficiently captured by hybridization are amplified by PCR with specific primers. In addition, PCR with specific primers is may be used to amplify exons for which similar sequences (“pseudo exons”) exist elsewhere in the genome.
  • In certain embodiments where massively parallel sequencing is used, PCR products are concatenated to form long stretches of DNA, which are sheared into short fragments (e.g., by acoustic energy). This step ensures that the fragment ends are distributed throughout the regions of interest. Subsequently, a stretch of dA nucleotides is added to the 3′ end of each fragment, which allows the fragments to bind to a planar surface coated with oligo(dT) primers (the “flow cell”). Each fragment may then be sequenced by extending the oligo(dT) primer with fluorescently-labeled nucleotides. During each sequencing cycle, only one type of nucleotide (A, G, T, or C) is added, and only one nucleotide is allowed to be incorporated through use of chain terminating nucleotides. For example, during the 1st sequencing cycle, a fluorescently labeled dCTP could be added. This nucleotide will only be incorporated into those growing complementary DNA strands that need a C as the next nucleotide. After each sequencing cycle, an image of the flow cell is taken to determine which fragment was extended. DNA strands that have incorporated a C will emit light, while DNA strands that have not incorporated a C will appear dark. Chain termination is reversed to make the growing DNA strands extendible again, and the process is repeated for a total of 120 cycles.
  • The images are converted into strings of bases, commonly referred to as “reads,” which recapitulate the 3′ terminal 25 to 60 bases of each fragment. The reads are then compared to the reference sequence for the DNA that was analyzed. Since any given string of 25 bases typically only occurs once in the human genome, most reads can be “aligned” to one specific place in the human genome. Finally, a consensus sequence of each genomic region may be built from the available reads and compared to the exact sequence of the reference at that position. Any differences between the consensus sequence and the reference are called as sequence variants.
  • Detectable Moieties
  • In certain embodiments, certain molecules (e.g., nucleic acid probes, antibodies, etc.) used in accordance with and/or provided by the invention comprise one or more detectable entities or moieties, i.e., such molecules are “labeled” with such entities or moieties.
  • Any of a wide variety of detectable agents can be used in the practice of the disclosure. Suitable detectable agents include, but are not limited to: various ligands, radionuclides; fluorescent dyes; chemiluminescent agents (such as, for example, acridinum esters, stabilized dioxetanes, and the like); bioluminescent agents; spectrally resolvable inorganic fluorescent semiconductors nanocrystals (i.e., quantum dots); microparticles; metal nanoparticles (e.g., gold, silver, copper, platinum, etc.); nanoclusters; paramagnetic metal ions; enzymes; colorimetric labels (such as, for example, dyes, colloidal gold, and the like); biotin; dioxigenin; haptens; and proteins for which antisera or monoclonal antibodies are available.
  • In some embodiments, the detectable moiety is biotin. Biotin can be bound to avidins (such as streptavidin), which are typically conjugated (directly or indirectly) to other moieties (e.g., fluorescent moieties) that are detectable themselves.
  • Below are described some non-limiting examples of some detectable moieties that may be used.
  • Fluorescent Dyes
  • In certain embodiments, a detectable moiety is a fluorescent dye. Numerous known fluorescent dyes of a wide variety of chemical structures and physical characteristics are suitable for use in the practice of the disclosure. A fluorescent detectable moiety can be stimulated by a laser with the emitted light captured by a detector. The detector can be a charge-coupled device (CCD) or a confocal microscope, which records its intensity.
  • Suitable fluorescent dyes include, but are not limited to, fluorescein and fluorescein dyes (e.g., fluorescein isothiocyanine or FITC, naphthofluorescein, 4′,5′-dichloro-2′,7′-dimethoxyfluorescein, 6-carboxyfluorescein or FAM, etc.), hexachloro-fluorescein (HEX), carbocyanine, merocyanine, styryl dyes, oxonol dyes, phycoerythrin, erythrosin, eosin, rhodamine dyes (e.g., carboxytetramethylrhodamine or TAMRA, carboxyrhodamine 6G, carboxy-X-rhodamine (ROX), lissamine rhodamine B, rhodamine 6G, rhodamine Green, rhodamine Red, tetramethylrhodamine (TMR), etc.), coumarin and coumarin dyes (e.g., methoxycoumarin, dialkylaminocoumarin, hydroxycoumarin, aminomethylcoumarin (AMCA), etc.), Q-DOTS. Oregon Green Dyes (e.g., Oregon Green 488, Oregon Green 500, Oregon Green 514., etc.), Texas Red, Texas Red-X, SPECTRUM RED, SPECTRUM GREEN, cyanine dyes (e.g., CY-3, CY-5, CY-3.5, CY-5.5, etc.), ALEXA FLUOR dyes (e.g., ALEXA FLUOR 350, ALEXA FLUOR 488, ALEXA FLUOR 532, ALEXA FLUOR 546, ALEXA FLUOR 568, ALEXA FLUOR 594, ALEXA FLUOR 633, ALEXA FLUOR 660, ALEXA FLUOR 680, etc.), BODIPY dyes (e.g., BODIPY FL, BODIPY R6G, BODIPY R, BODIPY TR, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/665, etc.), IRDyes (e.g., IRD40, IRD 700, IRD 800, etc.), and the like. For more examples of suitable fluorescent dyes and methods for coupling fluorescent dyes to other chemical entities such as proteins and peptides, see, for example, “The Handbook of Fluorescent Probes and Research Products”, 9th Ed., Molecular Probes, Inc., Eugene, Oreg. Favorable properties of fluorescent labeling agents include high molar absorption coefficient, high fluorescence quantum yield, and photostability. In some embodiments, labeling fluorophores exhibit absorption and emission wavelengths in the visible (i.e., between 400 and 750 nm) rather than in the ultraviolet range of the spectrum (i.e., lower than 400 nm).
  • A detectable moiety may include more than one chemical entity such as in fluorescent resonance energy transfer (FRET). Resonance transfer results an overall enhancement of the emission intensity. For instance, see Ju et. al. (1995) Proc. Nat'l Acad. Sci. (USA) 92:4347, the entire contents of which are herein incorporated by reference. To achieve resonance energy transfer, the first fluorescent molecule (the “donor” fluor) absorbs light and transfers it through the resonance of excited electrons to the second fluorescent molecule (the “acceptor” fluor). In one approach, both the donor and acceptor dyes can be linked together and attached to the oligo primer. Methods to link donor and acceptor dyes to a nucleic acid have been described, for example, in U.S. Pat. No. 5,945,526 to Lee et al., the entire contents of which are herein incorporated by reference. Donor/acceptor pairs of dyes that can be used include, for example, fluorescein/tetramethylrohdamine, IAEDANS/fluroescein, EDANS/DABCYL, fluorescein/fluorescein, BODIPY FL/BODIPY FL, and Fluorescein/ QSY 7 dye. See, e.g., U.S. Pat. No. 5,945,526 to Lee et al. Many of these dyes also are commercially available, for instance, from Molecular Probes Inc. (Eugene, Oreg.). Suitable donor fluorophores include 6-carboxyfluorescein (FAM), tetrachloro-6-carboxyfluorescein (TET), 2′-chloro-7′-phenyl-1,4-dichloro-6-carboxyfluorescein (VIC), and the like.
  • Enzymes
  • In certain embodiments, a detectable moiety is an enzyme. Examples of suitable enzymes include, but are not limited to, those used in an ELISA, e.g., horseradish peroxidase, beta-galactosidase, luciferase, alkaline phosphatase, etc. Other examples include beta-glucuronidase, beta-D-glucosidase, urease, glucose oxidase, etc. An enzyme may be conjugated to a molecule using a linker group such as a carbodiimide, a diisocyanate, a glutaraldehyde, and the like.
  • Radioactive Isotopes
  • In certain embodiments, a detectable moiety is a radioactive isotope. For example, a molecule may be isotopically-labeled (i.e., may contain one or more atoms that have been replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature) or an isotope may be attached to the molecule. Non-limiting examples of isotopes that can be incorporated into molecules include isotopes of hydrogen, carbon, fluorine, phosphorous, copper, gallium, yttrium, technetium, indium, iodine, rhenium, thallium, bismuth, astatine, samarium, and lutetium (i.e., 3H, 13C, 14C, 18F, 19F, 32P, 35S, 64Cu, 67Cu, 67Ga, 90Y, 99mTc, 111In, 125I, 123I, 129I, 131I, 135I, 186Re, 187Re, 201Tl, 212Bi, 213Bi, 211At, 153Sm, 177Lu).
  • In some embodiments, signal amplification is achieved using labeled dendrimers as the detectable moiety (see, e.g., Physiol Genomics 3:93-99, 2000), the entire contents of which are herein incorporated by reference in their entirety. Fluorescently labeled dendrimers are available from Genisphere (Montvale, N.J.). These may be chemically conjugated to the oligonucleotide primers by methods known in the art.
  • Kits
  • In certain embodiments, the disclosure provides kits for use in accordance with methods and compositions disclosed herein. Generally, kits comprise one or more reagents detect the biomarker of interest. Suitable reagents may include nucleic acid probes and/or antibodies or fragments thereof In some embodiments, suitable reagents are provided in a form of an array such as a microarray or a mutation panel.
  • In some embodiments, provided kits further comprise reagents for carried out various detection methods described herein (e.g., sequencing, hybridization, primer extension, multiplex ASPE, immunoassays, etc.). For example, kits may optionally contain buffers, enzymes, and/or reagents for use in methods described herein, e.g., for amplifying nucleic acids via primer-directed amplification, for performing ELISA experiments, etc.
  • In some embodiments, provided kits further comprise a control indicative of a healthy individual, e.g., a nucleic acid and/or protein sample from an individual who does not have the disease and/or syndrome of interest. In an embodiment, the control value is determined from a healthy individual or individuals with no detected or detectable lung or cardiovascular pathology. In some embodiments, the control is a disease control. Such disease controls may include individuals with lung or heart disease that is not related to exposure to an inhaled carcinogen.
  • Kits may also contain instructions on how to determine if an individual has the disease and/or syndrome of interest, or is at risk of developing the disease and/or syndrome of interest.
  • In some embodiments, provided is a computer readable medium encoding information corresponding to the biomarker of interest. Such computer readable medium may be included in a kit of the invention.
  • Methods to Identify Markers Associated With A Disease Associated With Exposure to Inhaled Carcinogens
  • Data Mining
  • In certain embodiments of the disclosure, biomarkers are identified using a data mining approach. For example, in some cases public databases (e.g., PubMed) may be searched for genes that have been shown to be linked to (directly or indirectly) to a certain disease. Such genes may then be evaluated as biomarkers. FIG. 1 shows an example of a multi-node interaction network identifying markers associated with diseases that may be associated with inhaled carcinogens. as described in detail in U.S. Provisional Patent Application No. 62/505,536, filed May 12, 2017 and U.S. Provisional Patent Application 62/523,382, filed Jun. 22, 2017, and incorporated by reference in their entireties herein. In the figure, the circled markers comprise the more relevant disease markers. FIG. 2 shows an example of a Venn diagram depicting markers associated with lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and cardiovascular disease (CVD). Many of the markers are those markers in FIG. 1 shown to be assocated with disease (circled).
  • Molecular
  • In certain embodiments, the disclosure comprises methods to identify biomarkers for a syndrome or disease of interest (i.e., variants in nucleic acid sequence that are associated with a disease associated with exposure to inhaled carcinogens in a statistically significant manner). The genes and/or genomic regions assayed for new markers may be selected based upon their importance in biochemical pathways that show genetic linkage and/or biological causation to the syndrome and/or disease of interest. Or, the genes and/or genomic regions assayed for markers may be selected based on genetic linkage to DNA regions that are genetically linked to the inheritance of a disease associated with exposure to an inhaled carcinogen in families. Or, the genes and/or genomic regions assayed for markers may be evaluated systematically to cover certain regions of chromosomes not yet evaluated.
  • In other embodiments, the genes or genomic regions evaluated for new markers may be part of a biochemical pathway that may be linked to the development of the syndrome and/or disease of interest (e.g., e.g., a disease associated with exposure to inhaled carcinogens). The variants and/or variant combinations may be assessed for their clinical significance based on one or more of the following methods. If a variant or a variant combination is reported or known to occur more often in nucleic acid from subjects with, than in subjects without, the syndrome and/or disease of interest it is considered to be at least potentially predisposing to the syndrome and/or disease of interest. If a variant or a variant combination is reported or known to be transmitted exclusively or preferentially to individuals having the syndrome and/or disease of interest, it is considered to be at least potentially predisposing to the syndrome and/or disease of interest. Conversely, if a variant is found in both populations at a similar frequency, it is less likely to be associated with the development of the syndrome and/or disease of interest.
  • If a variant or a variant combination is reported or known to have an overall deleterious effect on the function of a protein or a biological system in an experimental model system appropriate for measuring the function of this protein or this biological system, and if this variant or variant combination affects a gene or genes known to be associated with the syndrome and/or disease of interest, it is considered to be at least potentially predisposing to the syndrome and/or disease of interest. For example, if a variant or a variant combination is predicted to have an overall deleterious effect on a protein or gene expression (i.e., resulting in a nonsense mutation, a frameshift mutation, or a splice site mutation, or even a missense mutation), based on the predicted effect on the sequence and/or the structure of a protein or a nucleic acid, and if this variant or variant combination affects a gene or genes known to be associated with the syndrome and/or disease of interest, it is considered to be at least potentially predisposing to the syndrome and/or disease of interest.
  • Also, in certain embodiments, the overall number of variants may be important. If, in the test sample, a variant or several variants are detected that are, individually or in combination, assessed as at least probably associated with the syndrome and/or disease of interest, then the individual in whose genetic material this variant or these variants were detected can be diagnosed as being affected with or at high risk of developing the syndrome and/or disease of interest.
  • For example, the disclosure herein provides methods for diagnosing the presence or an increased risk of developing a disease associated with in a subject. Such methods may include obtaining a nucleic acid from a sample of tissue or body fluid. The method may further include sequencing the nucleic acid or determining the genomic arrangement or copy number of the nucleic acid to detect whether there is a variant or variants in the nucleic acid sequence or genomic arrangement or copy number. The method may further include the steps of assessing the clinical significance of a variant or variants. Such analysis may include an evaluation of the extent of association of the variant sequence in affected populations (i.e., subjects having the disease). Such analysis may also include an analysis of the extent of the effect the mutation may have on gene expression and/or protein function. The method may also include diagnosis the presence or an increased risk of developing a disease related to exposure to inhaled carcinogens based on the assessment.
  • The following examples serve to illustrate certain aspects of the disclosure. These examples are in no way intended to be limiting.
  • EXAMPLES
  • Biomarkers disclosed herein may be measured using any of the teachniques discuss above or in some cases by one of the non-limiting assays disclosed below.
  • EGFR
  • EGFR expression levels are measured by fluorescent in situ hybridization (FISH) using 4-5 micron FFPE sections of tissue. Alternatively, expresssion may be measured in serum, plasma, urine, bone marrow, blood, cerebrospinal fluid, FNA, or bone marrow. Gene mutation analysis is performed using real-time PCR, single base extension and/or DNA sequencing. Gene mutation can be determined in, for examples, tissue, serum, plasma or urine.
  • Interleukin 1 beta (IL1B)
  • Interleukin 1 beta (IL1β) is a cytokine protein that in humans is encoded by the IL1B gene. There are two genes for interleukin-1 (IL-1): IL-1 alpha and IL-1 beta. IL-1β precursor is cleaved by cytosolic caspase 1 (interleukin 1 beta convertase) to form mature IL-1β. IL-1β is a member of the interleukin 1 family of cytokines. This cytokine is produced by activated macrophages as a proprotein, which is proteolytically processed to its active form by caspase 1 (CASP1/ICE). This cytokine is an important mediator of the inflammatory response, and is involved in a variety of cellular activities, including cell proliferation, differentiation, and apoptosis. Increased production of IL-1β causes a number of different autoinflammatory syndromes.
  • CXC Motif Chemokine Ligand 8 (CXCL8)
  • Interleukin 8 (IL8 or chemokine (C-X-C motif) ligand 8, CXCL8) is a chemokine produced by macrophages and other cell types such as epithelial cells, airway smooth muscle cells and endothelial cells. In humans, the interleukin-8 protein is encoded by the CXCL8 gene. IL-8 is initially produced as a precursor peptide of 99 amino acids which then undergoes cleavage to create several active IL-8 isoforms. In culture, a 72 amino acid peptide is the major form secreted by macrophages IL-8, also known as neutrophil chemotactic factor, has two primary functions. It induces chemotaxis in target cells, primarily neutrophils but also other granulocytes, causing them to migrate toward the site of infection. IL-8 also induces phagocytosis once they have arrived. IL-8 is also known to be a potent promoter of angiogenesis. In target cells, IL-8 induces a series of physiological responses required for migration and phagocytosis, such as increases in intracellular Ca2+, exocytosis (e.g. histamine release), and the respiratory burst.
  • Tumor Necrosis Factor Alpha (TNF-α)
  • Tumor Necrosis Factor alpha (TNF-α), also known as cachectin and TNFSF1A, is the prototypic ligand of the TNF superfamily. TNF-α plays a central role in inflammation, immune system development, apoptosis, and lipid metabolism. TNF-α is also involved in a number of pathological conditions including asthma, Crohn's disease, rheumatoid arthritis, neuropathic pain, obesity, type 2 diabetes, septic shock, autoimmunity, and cancer.
  • TNF-α may be measured using the Quantikine assay. The Quantikine TNF-α immunoassay is a 4.0 hour solid phase ELISA designed to measure human TNF-α in serum and plasma. The assay contains E. coli-derived recombinant human TNF-α and antibodies raised against the recombinant factor, and employs the quantitative sandwich enzyme immunoassay technique. A monoclonal antibody specific for human TNF-α is pre-coated onto a microplate. Standards and samples are pipetted into the wells and TNF-α present is bound by the immobilized antibody. After washing away any unbound substances, a biotinylated polyclonal antibody specific for human TNF-α is added to the wells, the wells are washed to remove unbound antibody-biotin reagent, and an enzyme-linked streptavidin is added to the wells. After washing, a substrate solution (hydrogen peroxide and tetramethylbenzidine) is added to the wells and color develops in proportion to the amount of TNF-α bound in the initial step. The color development is stopped and the intensity of the color is measured at 450 nm, subtracting readings at 540 nm and 570 nm.
  • The samples may be serum or plasma. For serum, a serum separator tube (SST) is used and samples are allowed to clot for 30 minutes at room temperature before centrifugation for 15 minutes at 1000×g. The serum is removed and assayed. Samples are used immediately or aliquoted and stored at <−20° C. Plasma is collected using EDTA or heparin as an anticoagulant. The samples are centrifuged for 15 minutes at 1000×g within 30 minutes of collection, and samples assayed immediately or aliquoted and stored at <−20° C. For both serum and plasma samples, repeated freeze-thaw cycles should be avoided. Generally, grossly hemolyzed samples, high albumin samples, and citrate plasma should not be used.
  • Interferon Gamma (INFG)
  • Interferon gamma (IFNγ) is a dimerized soluble cytokine that is the only member of the type II class of interferons IFNγ, or type II interferon, is a cytokine that is critical for innate and adaptive immunity against viral, some bacterial and protozoal infections. IFNγ is an important activator of macrophages and inducer of Class II major histocompatibility complex (MHC) molecule expression. Aberrant IFNγ expression is associated with a number of autoinflammatory and autoimmune diseases. IFNγ is produced predominantly by natural killer (NK) and natural killer T (NKT) cells as part of the innate immune response, and by CD4 Th1 and CD8 cytotoxic T lymphocyte (CTL) effector T cells once antigen-specific immunity develops.
  • Cadherin (CDH1), SMAD Family Member 4 (SMAD4),
  • The entire coding region of a panel of genes related to hereditary cancer is examined by next generation sequencing analysis. Additionally, portions of the flanking noncoding regions are also examined. Comprehensive deletion/duplication testing is performed using microarray CGH for 20 genes, and by multiplex ligation-dependent probe amplification (MLPA) for the CHEK2 and PMS2 genes. Genes tested in this panel include APC, ATM, AXIN2, BLMM, BMPR1A, BRCA1, BRCA2, CDH1, CDKN2A, CHEK2, EPCAM, MLH1, MSH2, MSH6, MUTYH, PMS2, POLD1, POLE, PTEN, SMAD4, STK11 and TP53. Clinically significant findings are confirmed by Sanger sequencing or qPCR. Results are reported using ACMG guidelines and nomenclature recommended by the Human Genome Variation Society (HGVS).
  • Serpin Family E Member 1 (SERPINE1)
  • Plasminogen activator inhibitor-1 (PAI-1) also known as endothelial plasminogen activator inhibitor or serpin E1 is a protein that in humans is encoded by the SERPINE1 gene. Elevated PAI-1 is a risk factor for thrombosis and atherosclerosis. PAI-1 is a serine protease inhibitor (serpin) that functions as the principal inhibitor of tissue plasminogen activator (tPA) and urokinase (uPA), activators of plasminogen and fibrinolysis. It is a serine protease inhibitor (serpin) protein (SERPINE1).The PAI-1 gene is SERPINE1, located on chromosome 7 (7q21.3-q22). There is a common polymorphism known as 4G/5G in the promoter region. The 5G allele is slightly less transcriptionally active than the 4G.
  • Signal Transducer and Activator of Transcription 3 (STAT3)
  • Mutatations in STAT3 are detected by DNA sequencing of all coding nucleotides of gene STAT3, plus at least two and typically 20 flanking intronic nucleotides upstream and downstream of each coding exon, covering the conserved donor and acceptor splice sites, as well as typically 20 flanking nucleotides in the 5′ and 3′ UTR.
  • Intercellular Adhesion Molecule 1 (ICAM1)
  • ICAM-1 (Intercellular Adhesion Molecule 1) also known as CD54 (Cluster of Differentiation 54) is a protein that in humans is encoded by the ICAM1 gene. ICAM-1 is a member of the immunoglobulin superfamily. This cell surface glycoprotein is typically expressed on endothelial cells and cells of the immune system, and binds to integrins of type CD11a/CD18, or CD11b/CD18.
  • Interleukin 6 (IL6)
  • Interleukin 6 (IL-6) is an interleukin that acts as both a pro-inflammatory cytokine and an anti-inflammatory myokine. In humans, it is encoded by the IL6 gene. Interleukin 6 is secreted by T cells and macrophages to stimulate immune response, e.g. during infection and after tissue damage leading to inflammation. IL-6 also plays a role in fighting infection. In addition, osteoblasts secrete IL-6 to stimulate osteoclast formation. Smooth muscle cells in the tunica media of many blood vessels also produce IL-6 as a pro-inflammatory cytokine. IL-6's role as an anti-inflammatory cytokine is mediated through its inhibitory effects on TNF-alpha and IL-1, and activation of IL-1ra and IL-10. IL-6 is an important mediator of fever and of the acute phase response. It is capable of crossing the blood-brain barrier, initiating synthesis of PGE2 in the hypothalamus, and changing the body's temperature setpoint. In muscle and fatty tissue, IL-6 stimulates energy mobilization that leads to increased body temperature. IL-6 can be secreted by macrophages in response to specific microbial molecules, referred to as pathogen-associated molecular patterns (PAMPs), and is also considered a myokine (a cytokine produced from muscle), which is elevated in response to muscle contraction
  • Vascular Endothelial Growth Factor A (VEGFA)
  • Vascular endothelial growth factor (VEGF), also known as vascular permeability factor (VPF), is a homodimeric 34 to 45 kilodalton, heparin-binding glycoprotein. VEGF has potent angiogenic, mitogenic, and vascular permeability-enhancing activities specific for endothelial cells. VEGF is thought to play an important role in several physiologic processes, including wound healing, ovulation, menstruation, maintenance of blood pressure, and pregnancy. VEGF has also been associated with a number of pathologic processes that involve angiogenesis, including arthritis, psoriasis, macular degeneration, and diabetic retinopathy. Also, tumor expression of proangiogenic factors, including VEGF, has been associated with advanced tumor progression in a number of human cancers. VEGF can be measured in serum using enzyme immunoassay (EIA).
  • Interleukin 1 beta (IL1B)
  • Interleukin 1 beta (IL1β) is a cytokine protein that in humans is encoded by the IL1B gene. There are two genes for interleukin-1 (IL-1): IL-1 alpha and IL-1 beta. IL-1β precursor is cleaved by cytosolic caspase 1 (interleukin 1 beta convertase) to form mature IL-1β. IL-1β is a member of the interleukin 1 family of cytokines. This cytokine is produced by activated macrophages as a proprotein, which is proteolytically processed to its active form by caspase 1 (CASP1/ICE). This cytokine is an important mediator of the inflammatory response, and is involved in a variety of cellular activities, including cell proliferation, differentiation, and apoptosis. Increased production of IL-1β causes a number of different autoinflammatory syndromes.
  • C-C Motif Chemokine Ligand 2 (CCL2)
  • The chemokine (C-C motif) ligand 2 (CCL2) is also referred to as monocyte chemoattractant protein 1 (MCP1) and small inducible cytokine A2. CCL2 is a small cytokine that belongs to the CC chemokine family. CCL2 recruits monocytes, memory T cells, and dendritic cells to the sites of inflammation produced by either tissue injury or infection. Administration of anti-CCL2 antibodies in a model of glomerulonephritis reduces infiltration of macrophages and T cells, reduces crescent formation, as well as scarring and renal impairment. Hypomethylation of CpG sites within the CCL2 promoter region is affected by high levels of blood glucose and TG, which increase CCL2 levels in the blood serum. The later plays an important role in the vascular complications of type 2 diabetes.
  • Catenin Beta 1 (CTNNB1)
  • The Wnt signaling pathway is involved in both normal development and tumorigenesis. Activation of the pathway results in stabilization and nuclear translocation of beta-catenin protein. Nuclear localization of beta-catenin has been used to identify tumors in which mutations in APC or beta-catenin activate Wnt signaling.
  • Phosphatidylinositol-4,5,-bisphosphate 3-kinase Catalytic Subunit Alpha (PIK3CA)
  • Genomic DNA is isolated from the provided tumor specimen using the Cobas® DNA Sample Preparation Kit. Mutation detection is achieved through real-time PCR analysis on cobas® z480 analyzer using cobas® PIK3CA Mutation Test kit. Samples include formalin-fixed, paraffin-embedded tissue (FFPE) block or slides
  • CXC Motif Chemokine Ligand 8 (CXCL8)
  • Interleukin 8 (IL8 or chemokine (C-X-C motif) ligand 8, CXCL8) is a chemokine produced by macrophages and other cell types such as epithelial cells, airway smooth muscle cells and endothelial cells. In humans, the interleukin-8 protein is encoded by the CXCL8 gene. IL-8 is initially produced as a precursor peptide of 99 amino acids which then undergoes cleavage to create several active IL-8 isoforms. In culture, a 72 amino acid peptide is the major form secreted by macrophages IL-8, also known as neutrophil chemotactic factor, has two primary functions. It induces chemotaxis in target cells, primarily neutrophils but also other granulocytes, causing them to migrate toward the site of infection. IL-8 also induces phagocytosis once they have arrived. IL-8 is also known to be a potent promoter of angiogenesis. In target cells, IL-8 induces a series of physiological responses required for migration and phagocytosis, such as increases in intracellular Ca2+, exocytosis (e.g. histamine release), and the respiratory burst.
  • References and citations to other documents, such as patents, patent applications, patent publications, journals, books, papers, web contents, have been made throughout this disclosure. All such documents are hereby incorporated herein by reference in their entirety for all purposes. Various modifications and equivalents of those described herein, will become apparent to those skilled in the art from the full contents of this document, including references to the scientific and patent literature cited herein. The subject matter herein contains information, exemplification and guidance that can be adapted to the practice of this disclosure in its various embodiments and equivalents thereof.

Claims (20)

1. A method to detect at least one biomarker associated with exposure to an inhaled carcinogen comprising:
obtaining a sample from the individual; and
measuring the amount of at least one marker associated with at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD).
2. The method of claim 1, wherein detecting at least one biomarker associated with lung cancer (LC) in an individual comprises measuring the amount of the marker and/or the amount or a mutation in, a nucleic acid that encodes for or regulates expression of the gene for at least one of anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRC5), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAM5), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNA5), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine receptor 4 (CXCR4), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), cytochrome P450 family 1 subfamily B member 1 (CYP1B1), epidermal growth factor receptor (EGFR), epoxide hydrolase 1 (EPHX1), erb-b2 receptor tyrosine kinase 2 (ERBB2), fructose-bisphosphatase 1 (FBP1), fascin actin-bundling protein 1 (FSCN1), glutathione S-transferase pi 1 (GSTP1), interleukin 10 (IL10), integrin subunit alpha 11 (ITGA11), KRAS proto-oncogene, GTPase (KRAS), keratin 19 (KRT19), leucine aminopeptidase 3 (LAP3), MDM2 proto-oncogene (MDM2), v-myc avian myelocytomatosis viral oncogene homolog (MYC), p21 (RAC1) activated kinase 1 (PAK1), poly(ADP-ribose) polymerase 1 (PARP1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), protein kinase N1 (PKN1), phosphatase and tensin homolog (PTEN), signal transducer and activator of transcription 3 (STAT3), serine/threonine kinase 11 (STK11), tumor protein p53 (TP53), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor C (VEGFC), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or X-ray repair cross complementing 1 (XRCC1).
3. The method of claim 1, wherein detecting at least one biomarker associated with chronic obstructive pulmaonary disease (COPD) in an individual comprises measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid that encodes for, or regulates expression of, the gene for at least one of adiponectin, C1Q and collagen domain containing (ADIPOQ), adrenoceptor beta 2 (ADRB2), advanced glycosylation end product-specific receptor (AGER), CD4 molecule (CD4), cystic fibrosis transmembrane conductance regulator (CFTR), cholinergic receptor nicotinic alpha 3 subunit (CHRNA3), cystatin C (CST3), C-X-C motif chemokine ligand 8 (CXCL8), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), D-box binding PAR bZIP transcription factor (DBP), epidermal growth factor receptor (EGFR), elastin (ELN), erythropoietin (EPO), glutathione S-transferase pi 1 (GSTP1), histone deacetylase 2 (HDAC2), high mobility group box 1 (HMGB1), 5-hydroxytryptamine receptor 4 (HTR4), immunoglobulin heavy constant epsilon (IGHE), interleukin 10 (IL10), interleukin 13 (IL13), interleukin 1 beta (IL1B), laminin subunit alpha 1 (LAMA1), leptin (LEP), membrane metallo-endopeptidase (MME), matrix metallopeptidase 12 (MMP12), matrix metallopeptidase 25 (MMP25), serpin family A member 1 (SERPINA1), sirtuin 1 (SIRT1), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or vascular endothelial growth factor A (VEGFA).
4. The method of claim 1, wherein detecting at least one biomarker associated with cardovascular disease (CVD) in an individual comprises measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid that encodes for, or regulates expression of, the gene for at least one of ABO blood group (transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase) (ABO), angiotensin I converting enzyme 2 (ACE2), angiotensinogen (AGT), albumin (ALB), apelin (APLN), apolipoprotein A1 (APOA1), apolipoprotein A2 (APOA2), apolipoprotein B (APOB), apolipoprotein E (APOE), caspase 1 (CASP1), CD36 molecule (CD36), C-reactive protein, pentraxin-related (CRP), elongation factor for RNA polymerase II (ELL), coagulation factor II, thrombin (F2), intercellular adhesion molecule 1 (ICAM1), interleukin 1 beta (IL1B), low density lipoprotein receptor (LDLR), leptin (LEP), myeloperoxidase (MPO), nitric oxide synthase 3 (NOS3), period circadian clock 1 (PER1), prolactin (PRL), tumor necrosis factor (TNF), troponin C1, slow skeletal and cardiac type (TNNC1), troponin 13, cardiac type (TNNI3), troponin T2, cardiac type (TNNT2), vascular endothelial growth factor A (VEGFA), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or von Willebrand factor (VWF).
5. The method of claim 1, wherein the measuring comprises measurement of protein.
6. The method of claim 1, wherein the measuring comprises analysis of nucleic acid sequence or expression.
7. The method of claim 1, wherein the sample comprises a liquid or tissue biopsy, cell-free nucleic acid, blood, urine, serum or plasma.
8. A method of identifying a marker associated with a disease related to exposure to an inhaled carcinogen in an individual comprising: identifying at least one marker having increased or decreased expression in, or a mutation, in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for or regulates expression of the gene for lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD) as compared to a control value.
9. A method to detect the presence of, or susceptibility to, a disease associated with exposure to an inhaled carcinogen in an individual comprising:
obtaining a sample from the individual;
measuring the amount of at least one marker associated with at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD), in the sample; and
comparing the expression of, and/or the presence of a mutation in a gene for, the at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD) in the sample with a control value for each of the markers.
10. The method of claim 9, wherein detecting at least one biomarker associated with lung cancer (LC) in an individual comprises measuring the amount of the marker, and/or the amount, or a mutation in, a nucleic acid that encodes for or regulates expression of the gene for at least one of anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRC5), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAM5), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNA5), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine receptor 4 (CXCR4), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), cytochrome P450 family 1 subfamily B member 1 (CYP1B1), epidermal growth factor receptor (EGFR), epoxide hydrolase 1 (EPHX1), erb-b2 receptor tyrosine kinase 2 (ERBB2), fructose-bisphosphatase 1 (FBP1), fascin actin-bundling protein 1 (FSCN1), glutathione S-transferase pi 1 (GSTP1), interleukin 10 (IL10), integrin subunit alpha 11 (ITGA11), KRAS proto-oncogene, GTPase (KRAS), keratin 19 (KRT19), leucine aminopeptidase 3 (LAP3), MDM2 proto-oncogene (MDM2), v-myc avian myelocytomatosis viral oncogene homolog (MYC), p21 (RAC1) activated kinase 1 (PAK1), poly(ADP-ribose) polymerase 1 (PARP1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), protein kinase N1 (PKN1), phosphatase and tensin homolog (PTEN), signal transducer and activator of transcription 3 (STAT3), serine/threonine kinase 11 (STK11), tumor protein p53 (TP53), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor C (VEGFC), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or X-ray repair cross complementing 1 (XRCC1).
11. The method of claim 9, wherein detecting at least one biomarker associated with chronic obstructive pulmaonary disease (COPD) in an individual comprises measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid that encodes for, or regulates expression of, the gene for at least one of adiponectin, C1Q and collagen domain containing (ADIPOQ), adrenoceptor beta 2 (ADRB2), advanced glycosylation end product-specific receptor (AGER), CD4 molecule (CD4), cystic fibrosis transmembrane conductance regulator (CFTR), cholinergic receptor nicotinic alpha 3 subunit (CHRNA3), cystatin C (CST3), C-X-C motif chemokine ligand 8 (CXCL8), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), D-box binding PAR bZIP transcription factor (DBP), epidermal growth factor receptor (EGFR), elastin (ELN), erythropoietin (EPO), glutathione S-transferase pi 1 (GSTP1), histone deacetylase 2 (HDAC2), high mobility group box 1 (HMGB1), 5-hydroxytryptamine receptor 4 (HTR4), immunoglobulin heavy constant epsilon (IGHE), interleukin 10 (IL10), interleukin 13 (IL13), interleukin 1 beta (IL1B), laminin subunit alpha 1 (LAMA1), leptin (LEP), membrane metallo-endopeptidase (MME), matrix metallopeptidase 12 (MMP12), matrix metallopeptidase 25 (MMP25), serpin family A member 1 (SERPINA1), sirtuin 1 (SIRT1), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or vascular endothelial growth factor A (VEGFA).
12. The method of claim 9, wherein detecting at least one biomarker associated with cardovascular disease (CVD) in an individual comprises measuring the amount of the marker, and/or the amount and/or a mutation, in a nucleic acid (e.g. genomic DNA or mRNA) that encodes for, or regulates expression of, the gene for at least one of ABO blood group (transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase) (ABO), angiotensin I converting enzyme 2 (ACE2), angiotensinogen (AGT), albumin (ALB), apelin (APLN), apolipoprotein A1 (APOA1), apolipoprotein A2 (APOA2), apolipoprotein B (APOB), apolipoprotein E (APOE), caspase 1 (CASP1), CD36 molecule (CD36), C-reactive protein, pentraxin-related (CRP), elongation factor for RNA polymerase II (ELL), coagulation factor II, thrombin (F2), intercellular adhesion molecule 1 (ICAM1), interleukin 1 beta (IL1B), low density lipoprotein receptor (LDLR), leptin (LEP), myeloperoxidase (MPO), nitric oxide synthase 3 (NOS3), period circadian clock 1 (PERI1), prolactin (PRL), tumor necrosis factor (TNF), troponin C1, slow skeletal and cardiac type (TNNC1), troponin 13, cardiac type (TNNI3), troponin T2, cardiac type (TNNT2), vascular endothelial growth factor A (VEGFA), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or von Willebrand factor (VWF).
13. The method of claim, wherein the measuring comprises measurement of protein.
14. The method of claim 9, wherein the measuring comprises analysis of nucleic acid sequence or expression.
15. The method of claim 9, wherein the sample comprises a tissue or liquid biopsy, cell-free nucleic acid, blood, urine, serum or plasma.
16. A composition to detect a biomarker associated with a disease associated with exposure to an inhaled carcinogen in an individual comprising reagents that quantify the levels of and/or detect the presence of a mutation, in a nucleic acid that encodes for or regulates expression of the gene for at least one of lung cancer (LC), chronic obstructive pulmaonary disease (COPD), and/or cardiovascular disease (CVD).
17. The composition of claim 16, wherein the at least one biomarker associated with lung cancer (LC) comprises anaplastic lymphoma receptor tyrosine kinase (ALK), B-cell CLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (BIRCS), B-Raf proto-oncogene, serine/threonine kinase (BRAF), CD274 molecule (CD274), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), carcinoembryonic antigen related cell adhesion molecule 5 (CEACAMS), chitinase 3 like 1 (CHI3L1), cholinergic receptor nicotinic alpha 5 subunit (CHRNAS), CLPTM1-like (CLPTM1L), catechol-O-methyltransferase (COMT), catenin beta 1 (CTNNB1), C-X-C motif chemokine receptor 4 (CXCR4), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), cytochrome P450 family 1 subfamily B member 1 (CYP1B1), epidermal growth factor receptor (EGFR), epoxide hydrolase 1 (EPHX1), erb-b2 receptor tyrosine kinase 2 (ERBB2), fructose-bisphosphatase 1 (FBP1), fascin actin-bundling protein 1 (FSCN1), glutathione S-transferase pi 1 (GSTP1), interleukin 10 (IL10), integrin subunit alpha 11 (ITGA11), KRAS proto-oncogene, GTPase (KRAS), keratin 19 (KRT19), leucine aminopeptidase 3 (LAP3), MDM2 proto-oncogene (MDM2), v-myc avian myelocytomatosis viral oncogene homolog (MYC), p21 (RAC1) activated kinase 1 (PAK1), poly(ADP-ribose) polymerase 1 (PARP1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), protein kinase N1 (PKN1), phosphatase and tensin homolog (PTEN), signal transducer and activator of transcription 3 (STAT3), serine/threonine kinase 11 (STK11), tumor protein p53 (TP53), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor C (VEGFC), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or X-ray repair cross complementing 1 (XRCC1).
18. The composition of claim 16, wherein the at least one biomarker associated with chronic obstructive pulmaonary disease (COPD) comprises adiponectin, C1Q and collagen domain containing (ADIPOQ), adrenoceptor beta 2 (ADRB2), advanced glycosylation end product-specific receptor (AGER), CD4 molecule (CD4), cystic fibrosis transmembrane conductance regulator (CFTR), cholinergic receptor nicotinic alpha 3 subunit (CHRNA3), cystatin C (CST3), C-X-C motif chemokine ligand 8 (CXCL8), cytochrome P450 family 1 subfamily A member 1 (CYP1A1), D-box binding PAR bZIP transcription factor (DBP), epidermal growth factor receptor (EGFR), elastin (ELN), erythropoietin (EPO), glutathione S-transferase pi 1 (GSTP1), histone deacetylase 2 (HDAC2), high mobility group box 1 (HMGB1), 5-hydroxytryptamine receptor 4 (HTR4), immunoglobulin heavy constant epsilon (IGHE), interleukin 10 (IL10), interleukin 13 (IL13), interleukin 1 beta (IL1B), laminin subunit alpha 1 (LAMA1), leptin (LEP), membrane metallo-endopeptidase (MME), matrix metallopeptidase 12 (MMP12), matrix metallopeptidase 25 (MMP25), serpin family A member 1 (SERPINA1), sirtuin 1 (SIRT1), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or vascular endothelial growth factor A (VEGFA).
19. The composition of claim 16, wherein the at least one biomarker associated with cardovascular disease (CVD) comprises ABO blood group (transferase A, alpha 1-3-N-acetylgalactosaminyltransferase; transferase B, alpha 1-3-galactosyltransferase) (ABO), angiotensin I converting enzyme 2 (ACE2), angiotensinogen (AGT), albumin (ALB), apelin (APLN), apolipoprotein A1 (APOA1), apolipoprotein A2 (APOA2), apolipoprotein B (APOB), apolipoprotein E (APOE), caspase 1 (CASP1), CD36 molecule (CD36), C-reactive protein, pentraxin-related (CRP), elongation factor for RNA polymerase II (ELL), coagulation factor II, thrombin (F2), intercellular adhesion molecule 1 (ICAM1), interleukin 1 beta (IL1B), low density lipoprotein receptor (LDLR), leptin (LEP), myeloperoxidase (MPO), nitric oxide synthase 3 (NOS3), period circadian clock 1 (PERI), prolactin (PRL), tumor necrosis factor (TNF), troponin C1, slow skeletal and cardiac type (TNNC1), troponin 13, cardiac type (TNNI3), troponin T2, cardiac type (TNNT2), vascular endothelial growth factor A (VEGFA), interferon gamma (INFG), interleukin 2 (IL2), tumor necrosis factor (TNF), interleukin 4 (IL4), or von Willebrand factor (VWF).
20. A kit that comprises the composition of claim 16.
US16/611,628 2017-05-12 2018-05-11 Compositions and methods for detection of diseases related to exposure to inhaled carcinogens Abandoned US20200332364A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/611,628 US20200332364A1 (en) 2017-05-12 2018-05-11 Compositions and methods for detection of diseases related to exposure to inhaled carcinogens

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762505536P 2017-05-12 2017-05-12
US201762523382P 2017-06-22 2017-06-22
PCT/US2018/032293 WO2018209218A1 (en) 2017-05-12 2018-05-11 Compositions and methods for detection of diseases related to exposure to inhaled carcinogens
US16/611,628 US20200332364A1 (en) 2017-05-12 2018-05-11 Compositions and methods for detection of diseases related to exposure to inhaled carcinogens

Publications (1)

Publication Number Publication Date
US20200332364A1 true US20200332364A1 (en) 2020-10-22

Family

ID=62685096

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/611,628 Abandoned US20200332364A1 (en) 2017-05-12 2018-05-11 Compositions and methods for detection of diseases related to exposure to inhaled carcinogens
US15/977,000 Active 2038-08-14 US10861583B2 (en) 2017-05-12 2018-05-11 Systems and methods for biomarker identification

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/977,000 Active 2038-08-14 US10861583B2 (en) 2017-05-12 2018-05-11 Systems and methods for biomarker identification

Country Status (6)

Country Link
US (2) US20200332364A1 (en)
EP (1) EP3622424A1 (en)
JP (1) JP7179766B2 (en)
CN (2) CN110603597B (en)
CA (2) CA3057420C (en)
WO (2) WO2018209218A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110079614B (en) * 2019-05-23 2021-03-16 华中农业大学 Molecular marker related to pig muscle fiber area and intramuscular fat content, detection method and application thereof
CN110444248B (en) * 2019-07-22 2021-09-24 山东大学 Cancer biomolecule marker screening method and system based on network topology parameters
CN112786141B (en) * 2021-01-21 2023-07-14 北京晶泰科技有限公司 Functional peptide recommendation method and device and computing equipment
US20220261668A1 (en) * 2021-02-12 2022-08-18 Tempus Labs, Inc. Artificial intelligence engine for directed hypothesis generation and ranking
CN114121166B (en) * 2021-11-19 2024-04-19 北京交通大学 Cognitive behavior and molecular network mechanism correlation method based on complex network

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4998617A (en) 1986-09-15 1991-03-12 Laura Lupton Inc Facial cosmetic liquid make up kit
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
JPS6412935A (en) 1987-07-02 1989-01-17 Mitsubishi Electric Corp Constant-speed travel device for vehicle
US5846710A (en) 1990-11-02 1998-12-08 St. Louis University Method for the detection of genetic diseases and gene sequence variations by single nucleotide primer extension
US5888819A (en) 1991-03-05 1999-03-30 Molecular Tool, Inc. Method for determining nucleotide identity through primer extension
US5945526A (en) 1996-05-03 1999-08-31 Perkin-Elmer Corporation Energy transfer dyes with enhanced fluorescence
US5885775A (en) 1996-10-04 1999-03-23 Perseptive Biosystems, Inc. Methods for determining sequences information in polynucleotides using mass spectrometry
EP1164203B1 (en) 1996-11-06 2007-10-10 Sequenom, Inc. DNA Diagnostics based on mass spectrometry
US6280947B1 (en) 1999-08-11 2001-08-28 Exact Sciences Corporation Methods for detecting nucleotide insertion or deletion using primer extension
US6503718B2 (en) 1999-01-10 2003-01-07 Exact Sciences Corporation Methods for detecting mutations using primer extension for detecting disease
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
US6919174B1 (en) 1999-12-07 2005-07-19 Exact Sciences Corporation Methods for disease detection
AU2003278137A1 (en) 2002-06-20 2004-01-06 Bristol-Myers Squibb Company Identification and regulation of a g-protein coupled receptor, rai-3
CN1701343A (en) * 2002-09-20 2005-11-23 德克萨斯大学董事会 Computer program products, systems and methods for information discovery and relational analyses
WO2005017652A2 (en) * 2003-08-05 2005-02-24 Rosetta Inpharmatics, Llc Computer systems and methods for inferring causality from cellular constituent abundance data
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
JP2008537120A (en) 2005-04-14 2008-09-11 ザ トラスティーズ オブ ボストン ユニバーシティ Diagnosis of lung injury using classification prediction
US20070072226A1 (en) * 2005-09-27 2007-03-29 Indiana University Research & Technology Corporation Mining protein interaction networks
US7282337B1 (en) 2006-04-14 2007-10-16 Helicos Biosciences Corporation Methods for increasing accuracy of nucleic acid sequencing
US8600718B1 (en) * 2006-11-17 2013-12-03 Microsoft Corporation Computer systems and methods for identifying conserved cellular constituent clusters across datasets
WO2009062199A1 (en) * 2007-11-09 2009-05-14 Fox Chase Cancer Center EGFR/NEDD9/TGF-β LNTERACTOME AND METHODS OF USE THEREOF FOR THE IDENTIFICATION OF AGENTS HAVING EFFICACY IN THE TREATMENT OF HYPERPROLIFERATIVE DISORDERS
US20090156412A1 (en) 2007-12-17 2009-06-18 Helicos Biosciences Corporation Surface-capture of target nucleic acids
GB2464677A (en) * 2008-10-20 2010-04-28 Univ Nottingham Trent A method of analysing data by using an artificial neural network to identify relationships between the data and one or more conditions.
MX358408B (en) * 2011-03-02 2018-08-20 Berg Llc Interrogatory cell-based assays and uses thereof.
EP2608122A1 (en) * 2011-12-22 2013-06-26 Philip Morris Products S.A. Systems and methods for quantifying the impact of biological perturbations
JP6320999B2 (en) * 2012-06-21 2018-05-09 フィリップ モリス プロダクツ エス アー Systems and methods related to network-based biomarker signatures
SG11201501691VA (en) * 2012-09-05 2015-04-29 Element Inc Biometric authentication in connection with camera-equipped devices
MX2018001831A (en) * 2015-08-14 2018-09-06 Univ Columbia Biomarkers for treatment of alopecia areata.
CA3035315A1 (en) * 2015-08-28 2017-03-09 University Of Maryland, College Park Computer system and methods for harnessing synthetic rescues and applications thereof
WO2017075294A1 (en) * 2015-10-28 2017-05-04 The Board Institute Inc. Assays for massively combinatorial perturbation profiling and cellular circuit reconstruction
WO2017161357A1 (en) * 2016-03-18 2017-09-21 Caris Science, Inc. Oligonucleotide probes and uses thereof

Also Published As

Publication number Publication date
CN110603597A (en) 2019-12-20
CA3057420A1 (en) 2018-11-15
JP2020522045A (en) 2020-07-27
CN110603597B (en) 2024-02-23
JP7179766B2 (en) 2022-11-29
WO2018209165A1 (en) 2018-11-15
EP3622424A1 (en) 2020-03-18
US10861583B2 (en) 2020-12-08
CA3060364A1 (en) 2018-11-15
WO2018209218A1 (en) 2018-11-15
CA3057420C (en) 2023-08-01
US20190065665A1 (en) 2019-02-28
CN110892080A (en) 2020-03-17

Similar Documents

Publication Publication Date Title
US20200332364A1 (en) Compositions and methods for detection of diseases related to exposure to inhaled carcinogens
US9631238B2 (en) Mutations associated with cystic fibrosis
JP7441346B2 (en) Compositions and methods for detecting head and neck cancer
JP2011509096A (en) Mutations in contact-related protein 2 (CNTNAP2) associated with increased risk of idiopathic autism
US20220074952A1 (en) Compositions and Methods to Detect Non-Coeliac Gluten Sensitivity
US20200087729A1 (en) Compositions and Methods to Detect Kidney Fibrosis

Legal Events

Date Code Title Description
AS Assignment

Owner name: LABORATORY CORPORATION OF AMERICA HOLDINGS, NORTH CAROLINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:JESSEN, WALTER JOSEPH;REEL/FRAME:050943/0925

Effective date: 20191024

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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