US20170219586A1 - Salivary Transcriptomics and Proteomic Biomarkers for Breast Cancer Detection - Google Patents

Salivary Transcriptomics and Proteomic Biomarkers for Breast Cancer Detection Download PDF

Info

Publication number
US20170219586A1
US20170219586A1 US15/488,832 US201715488832A US2017219586A1 US 20170219586 A1 US20170219586 A1 US 20170219586A1 US 201715488832 A US201715488832 A US 201715488832A US 2017219586 A1 US2017219586 A1 US 2017219586A1
Authority
US
United States
Prior art keywords
biomarkers
seq
subject
biomarker
disease
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
US15/488,832
Inventor
David T. Wong
Lei Zhang
Hua Xiao
Hui Zhou
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.)
University of California
Original Assignee
University of California
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 University of California filed Critical University of California
Priority to US15/488,832 priority Critical patent/US20170219586A1/en
Publication of US20170219586A1 publication Critical patent/US20170219586A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF CALIFORNIA LOS ANGELES
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • biomarker discoveries for breast cancer detection have focused on blood/or tissue, using proteomic, transcriptomic, and genomic approaches.
  • detection biomarkers In comparison to prognostic biomarkers, the development of detection biomarkers has been limited, mainly due to a lack of sensitivity and specificity for this clinical context.
  • tissue biomarkers for early detection will be limited to patients at very high risk because they rely on invasive procedures.
  • a method of determining the likelihood of the presence or occurrence of breast cancer in a test subject includes analyzing a saliva sample from the subject with an assay that specifically detects at least two biomarkers in the saliva sample.
  • the biomarkers are selected from the group of: S100A8 (S100 calcium binding protein A8) (SEQ ID NO: 1), CSTA (cystatin A) (SEQ ID NO:2), GRM1 (glutamate receptor, metabotropic 1) (SEQ ID NO: 3), TPT1 (tumor protein, translationally-controlled 1) (SEQ ID NO:4), GRIK1 (glutamate receptor, ionotropic, kainate 1) (SEQ ID NO: 5), H6PD (hexose-6-phosphate dehydrogenase) (SEQ ID NO: 6), IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) (SEQ ID NO: 7), MDM4 (3T3 cell double minute 4) (SEQ ID NO: 8), and CA6 (carbonic anhydrase VI) (SEQ ID NO:8).
  • S100A8 S100 calcium binding protein A8
  • CSTA cystatin A
  • GRM1 glutmate receptor, metabotropic 1
  • one of the biomarkers of the at least two biomarkers is cystatin A (CSTA).
  • two of the at least two biomarkers is CSTA and transformed 3T3 cell double minute 4 (MDM4). The relative occurrence of these biomarkers or these biomarkers and others in these instances is determined and compared to a control, for example, thereby allowing the breast cancer status of the test subject to be determined.
  • the method of determining the likelihood of the presence or occurrence of breast cancer entails measuring at least three biomarkers.
  • two of the at least three biomarkers are CSTA and MDM4. The relative occurrence of these biomarkers or these biomarkers and others in these instances is determined and compared to a control, for example, thereby allowing the breast cancer status of the test subject to be determined.
  • one of the biomarkers of the at least two biomarkers is anhydrase VI (CA6) polypeptide.
  • the method of determining the likelihood of the presence or occurrence of breast cancer in a test subject includes an assay in which a nucleic acid encoding at least one biomarker is detected.
  • the nucleic acid can be detected by, for example, mass spectroscopy, polymerase chain reaction (PCR), microarray hybridization, thermal sequencing, capillary array sequencing, or solid phase sequencing.
  • the method of determining the likelihood of the presence or occurrence of breast cancer in a test subject includes an assay in which a polypeptide encoding at least one biomarker is detected.
  • the polypeptide can be detected by, for example, enzyme-linked immunosorbent assay (ELISA), Western blot, flow cytometry, immunofluorescence, immunohistochemistry, or mass spectroscopy.
  • ELISA enzyme-linked immunosorbent assay
  • a method for assessing the efficacy of a therapy includes analyzing a first saliva sample from the subject with an assay that specifically detects at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and CA6. This first analysis provides a first expression profile.
  • a therapy is applied to a subject.
  • An analysis of a second saliva sample from the subject is undertaken with an assay that specifically detects at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and CA6 thereby providing a second expression profile.
  • the first and second expression profiles are compared in order to assess the efficacy of a therapy.
  • a solid support in another embodiment, includes a capture binding probe selective for at least two biomarkers selected from the group of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4.
  • a first and a second solid support are provided, wherein the first solid support includes a capture binding probe selective for at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and wherein the second solid support includes a capture binding ligand for CA6.
  • the capture binding ligand of the kit is an antibody.
  • the kit provides one or more primers for the selective amplification of at least two biomarkers, wherein at least two of the biomarkers are selected from the group of: S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4.
  • one or more of the primers possess a detectable label.
  • a method of determining the likelihood of the presence or occurrence of breast cancer in a test subject includes analyzing a saliva sample from the subject with an assay that specifically detects at least nine biomarkers in the saliva sample.
  • the biomarkers are selected from the group of: S100A8 (S100 calcium binding protein A8) (SEQ ID NO: 1), CSTA (cystatin A) (SEQ ID NO:2), GRM1 (glutamate receptor, metabotropic 1) (SEQ ID NO: 3), TPT1 (tumor protein, translationally-controlled 1) (SEQ ID NO:4), GRIK1 (glutamate receptor, ionotropic, kainate 1) (SEQ ID NO: 5), H6PD (hexose-6-phosphate dehydrogenase) (SEQ ID NO: 6), IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) (SEQ ID NO: 7), MDM4 (3T3 cell double minute 4) (SEQ ID NO: 8), and CA6 (carbonic anhydrase VI) (SEQ ID NO:8).
  • S100A8 S100 calcium binding protein A8
  • CSTA cystatin A
  • GRM1 glutmate receptor, metabotropic 1
  • the number of biomarkers used can be 2, 3, 4, 5, 6, 7, 8, 9, or more.
  • FIG. 1 is a schematic of the study designed to identify and validate biomarkers associated with breast cancer.
  • FIG. 2 is a schematic representation of the protocol for saliva collection.
  • FIG. 3 represents the demographic information of all the subjects used.
  • the Mann-Whitney rank sum test was used to determine marker validation.
  • Possible confounding factors including age, ethnicity, smoking status, menopausal status, and HRT treatment, were evaluated for the biomarkers by logistic regression model. Linear regression model was constructed for each marker and used the factors cancer/normal and one of the confounders. cv.err:cross validation error rate.
  • FIG. 5 demonstrates the sensitivity achieved using a combination of the identified biomarkers.
  • A The shading of the contigency table boxes reflects the fraction of each samples type in each quandrant. “Cancer” and “Non’ headings indicate subjects with and withour cancer, respectively. SB+ and SB ⁇ , salivary biomarker test positive or negative; NPV, negative predictive value; PPV, positive predictive value; Sen, sensitivity; Spec, specificity.
  • B Score plot of principle component analysis (PCA). Combining the nine biomarkers, the control subjects (light shaded) separate from breast cancer patients (dark shading) with cumulative proportions of 66.9% for PC1 and 21.6% for PC2.
  • FIG. 6 represents cross-disease comparisons of the salivary mRNA biomarkers.
  • the identified mRNA biomarkers for breast cancer detection were checked against other microarray datasets. t-test p-values were calculated for the identified breast cancer genes to other microarray datasets to check for significant variation (*after Boneferonni correction, P ⁇ 0.0006) between patients and controls in those diseases.
  • Sample sizes were 10 versus 10 for oral cancer, 10 versus 10 for lung cancer, 12 versus 12 for pancreatic cancer, 11 versus 11 for ovarian cancer, 13 versus 13 for diabetes, 8 versus 10 for primary Sjögren's Syndrome, and 10 versus 10 for breast cancer.
  • saliva is a source of easily accessible bodily fluids, there has been little effort to study its value in cancer diagnosis. Protein, as well as RNA, can be detected in saliva.
  • the present invention discloses the diagnostic/prognostic significance of nine salivary biomarkers S100A8 (SEQ ID NO: 1) (S100 calcium binding protein A8, also referred to as myloid-related protein 8 (MRP8) or S100A9 (MRP14)), CSTA (SEQ ID NO: 2)(cystatin A), GRM1 (SEQ ID NO: 3)(glutamate receptor, metabotropic 1), TPT1 (SEQ ID NO: 4)(tumor protein, translationally-controlled 1), GRIK1 (SEQ ID NO: 5) (glutamate receptor, ionotropic, kainate 1), H6PD (SEQ ID NO: 6)(hexose-6-phosphate dehydrogenase or glucose 1-dehydrogenase), IGF2BP1 (SEQ ID NO: 7)(insulin-like growth factor 2 mRNA binding protein 1), MDM4 (SEQ ID NO: 8)(Mdm4, transformed 3T3 cell double minute 4; HDMX; MDMX;
  • Methods for detecting salivary biomarkers include techniques such as ELISA, PCR, for example, RT-PCR or mass spectroscopy, alone or in combination with other markers. Any specific probe can be used for detection, such as an antibody, a receptor, a ligand, RT-PCR etc. Mass spectroscopy can also be used for protein detection.
  • the present invention can be used alone or as a complement to traditional antigen analysis to enhance the diagnosis of breast and other cancers.
  • nucleic acids e.g., gene, pre-mRNA, mRNA, and polypeptides, polymorphic variants, alleles, mutants, and interspecies homologs that have an amino acid sequence that has greater than about 60% amino acid sequence identity, 65%, 70%, 75%, 80%, 85%, 90%, preferably 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or greater amino acid sequence identity, preferably over a region of over a region of at least about 25, 50, 100, 200, 500, 1000, or more amino acids, to a polypeptide encoded by a referenced nucleic acid or an amino acid sequence described herein.
  • the nucleic acids and proteins of the invention include both naturally occurring or recombinant molecules.
  • the nucleic acid or protein sequence is provided
  • “Cancer” refers to human cancers and carcinomas, sarcomas, adenocarcinomas, lymphomas, leukemias, etc., including solid and lymphoid cancers, kidney, breast, lung, kidney, bladder, colon, ovarian, prostate, pancreas, stomach, brain, head and neck, skin, uterine, testicular, esophagus, and liver cancer, including hepatocarcinoma, lymphoma, including non-Hodgkin's lymphomas (e.g., Burkitt's, Small Cell, and Large Cell lymphomas) and Hodgkin's lymphoma, leukemia, and multiple myeloma.
  • non-Hodgkin's lymphomas e.g., Burkitt's, Small Cell, and Large Cell lymphomas
  • Hodgkin's lymphoma e.g., leukemia, and multiple myeloma.
  • “Therapeutic treatment” and “cancer therapies” refers to chemotherapy, hormonal therapy, radiotherapy, and immunotherapy.
  • overexpress refers to a protein that is transcribed or translated at a detectably greater level, usually in a cancer cell, in comparison to a normal cell.
  • the term includes overexpression due to transcription, post transcriptional processing, translation, post-translational processing, cellular localization (e.g, organelle, cytoplasm, nucleus, cell surface), and RNA and protein stability, as compared to a normal cell.
  • Overexpression can be detected using conventional techniques for detecting mRNA (i.e., RT-PCR, PCR, hybridization) or proteins (i.e., ELISA, immunohistochemical techniques, mass spectroscopy).
  • Overexpression can be 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a normal cell. In certain instances, overexpression is 1-fold, 2-fold, 3-fold, 4-fold or more higher levels of transcription or translation in comparison to a normal cell.
  • cancer-associated antigen or “tumor-specific marker” or “tumor marker” interchangeably refers to a molecule (typically protein or nucleic acid such as RNA) that is expressed in the cell, expressed on the surface of a cancer cell or secreted by a cancer cell in comparison to a normal cell, and which is useful for the diagnosis of cancer, for providing a prognosis, and for preferential targeting of a pharmacological agent to the cancer cell.
  • a cancer-associated antigen is overexpressed in a cancer cell in comparison to a normal cell, for instance, about 1.2-fold over expression, about 2-fold overexpression, about 3-fold overexpression or more in comparison to a normal cell.
  • a cancer-associated antigen is a cell surface molecule that is inappropriately synthesized in the cancer cell, for instance, a molecule that contains deletions, additions or mutations in comparison to the molecule expressed on a normal cell. Oftentimes, a cancer-associated antigen will be expressed exclusively on the cell surface of a cancer cell and not synthesized or expressed on the surface of a normal cell.
  • Exemplified cell surface tumor markers include the proteins c-erbB-2 and human epidermal growth factor receptor (HER) for breast cancer, PSMA for prostate cancer, and carbohydrate mucins in numerous cancers, including breast, ovarian and colorectal.
  • markers may be used singly or in combination with other markers for any of the uses, e.g., diagnosis or prognosis of breast cancer, disclosed herein.
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e., about 60% identity, preferably 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or higher identity over a specified region, when compared and aligned for maximum correspondence over a comparison window or designated region) as measured using a BLAST or BLAST 2.0 sequence comparison algorithms with default parameters described below, or by manual alignment and visual inspection (see, e.g., NCBI web site hypertext transfer protocol://www.ncbi.nlm.nih.gov/BLAST/ or the like).
  • sequences are then said to be “substantially identical.”
  • This definition also refers to, or may be applied to, the compliment of a test sequence.
  • the definition also includes sequences that have deletions and/or additions, as well as those that have substitutions.
  • the preferred algorithms can account for gaps and the like.
  • identity exists over a region that is at least about 25 amino acids or nucleotides in length, or more preferably over a region that is 50-100 amino acids or nucleotides in length.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • sequence algorithm program parameters Preferably, default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • a “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Methods of alignment of sequences for comparison are well-known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol.
  • BLAST and BLAST 2.0 are used, with the parameters described herein, to determine percent sequence identity for the nucleic acids and proteins of the invention.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (hypertext transfer protocol://www.ncbi.nlm.nih gov/).
  • This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence.
  • T is referred to as the neighborhood word score threshold (Altschul et al., supra).
  • a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • Nucleic acid refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double-stranded form, and complements thereof.
  • nucleic acid is used interchangeably with gene, cDNA, mRNA, oligonucleotide, and polynucleotide.
  • a particular nucleic acid sequence also implicitly encompasses “splice variants” and nucleic acid sequences encoding truncated forms of cancer antigens.
  • a particular protein encoded by a nucleic acid implicitly encompasses any protein encoded by a splice variant or truncated form of that nucleic acid.
  • “Splice variants,” as the name suggests, are products of alternative splicing of a gene. After transcription, an initial nucleic acid transcript may be spliced such that different (alternate) nucleic acid splice products encode different polypeptides. Mechanisms for the production of splice variants vary, but include alternate splicing of exons.
  • Alternate polypeptides derived from the same nucleic acid by read-through transcription are also encompassed by this definition. Any products of a splicing reaction, including recombinant forms of the splice products, are included in this definition. Nucleic acids can be truncated at the 5′ end or at the 3′ end. Polypeptides can be truncated at the N-terminal end or the C-terminal end. Truncated versions of nucleic acid or polypeptide sequences can be naturally occurring or recombinantly created.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, .gamma.-carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an .alpha.
  • amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • “Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide.
  • nucleic acid variations are “silent variations,” which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan
  • TGG which is ordinarily the only codon for tryptophan
  • amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
  • the following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
  • a “label” or a “detectable moiety” is a composition detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means.
  • useful labels include fluorescent dyes, electron-dense reagents, enzymes (for example, as commonly used in an ELISA), biotin, digoxigenin, or haptens and proteins which can be made detectable, e.g., by incorporating a radiolabel into the peptide or used to detect antibodies specifically reactive with the peptide.
  • recombinant when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified.
  • recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
  • stringent hybridization conditions refers to conditions under which a probe will hybridize to its target subsequence, typically in a complex mixture of nucleic acids, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Probes, “Overview of principles of hybridization and the strategy of nucleic acid assays” (1993). Generally, stringent conditions are selected to be about 5-10° C. lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength pH.
  • Tm thermal melting point
  • the T m is the temperature (under defined ionic strength, pH, and nucleic concentration) at which 50% of the probes complementary to the target hybridize to the target sequence at equilibrium (as the target sequences are present in excess, at T m , 50% of the probes are occupied at equilibrium).
  • Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide.
  • a positive signal is at least two times background, preferably 10 times background hybridization.
  • Exemplary stringent hybridization conditions can be as following: 50% formamide, 5 ⁇ SSC, and 1% SDS, incubating at 42° C., or, 5 ⁇ SSC, 1% SDS, incubating at 65° C., with wash in 0.2 ⁇ SSC, and 0.1% SDS at 65° C.
  • Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides which they encode are substantially identical. This occurs, for example, when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code. In such cases, the nucleic acids typically hybridize under moderately stringent hybridization conditions.
  • Exemplary “moderately stringent hybridization conditions” include a hybridization in a buffer of 40% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 1 ⁇ SSC at 45° C. A positive hybridization is at least twice background.
  • Those of ordinary skill will readily recognize that alternative hybridization and wash conditions can be utilized to provide conditions of similar stringency. Additional guidelines for determining hybridization parameters are provided in numerous reference, e.g., and Current Protocols in Molecular Biology, ed. Ausubel, et al., supra.
  • a temperature of about 36° C. is typical for low stringency amplification, although annealing temperatures may vary between about 32° C. and 48° C. depending on primer length.
  • a temperature of about 62° C. is typical, although high stringency annealing temperatures can range from about 50° C. to about 65° C., depending on the primer length and specificity.
  • Typical cycle conditions for both high and low stringency amplifications include a denaturation phase of 90° C. ⁇ 95° C. for 30 sec-2 min., an annealing phase lasting 30 sec.-2 min., and an extension phase of about 72° C. for 1-2 min. Protocols and guidelines for low and high stringency amplification reactions are provided, e.g., in Innis et al. (1990) PCR Protocols, A Guide to Methods and Applications, Academic Press, Inc. N.Y.).
  • Antibody means a protein comprising one or more polypeptides substantially encoded by all or part of the recognized immunoglobulin genes.
  • the recognized immunoglobulin genes include the kappa ( ⁇ ), lambda ( ⁇ ) and heavy chain genetic loci, which together compose the myriad variable region genes, and the constant region genes mu ( ⁇ ), delta ( ⁇ ), gamma ( ⁇ ), epsilon ( ⁇ ) and alpha ( ⁇ ), which encode the IgM, IgD, IgG, IgE, and IgA isotypes respectively.
  • Antibody herein is meant to include full length antibodies and antibody fragments, and may refer to a natural antibody from any organism, an engineered antibody or an antibody generated recombinantly for experimental, therapeutic or other purposes as further defined below.
  • Antibody fragments include Fab, Fab′, F(ab′) 2 , Fv, scFv or other antigen-binding subsequences of antibodies and can include those produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA technologies.
  • the term “antibody” refers to both monoclonal and polyclonal antibodies. Antibodies can be antagonists, agonists, neutralizing, inhibitory or stimulatory.
  • Biomarkers may originate from epidemiological studies, animal studies, pathophysiological considerations and end-organ experiments. Ideally, a biomarker will have a high predictive value for a meaningful outcome measure, can be or is validated in appropriately designed prospective trials, reflects therapeutic success by corresponding changes in the surrogate marker results, and should be easy to assess in clinical practice.
  • Biomarkers can be used in conjunction with other diagnostic tools or used alone.
  • biomarker refers to a molecule whose measurement provides information as to the state of a subject.
  • the biomarker is used to assess a pathological state. Measurements of the biomarker may be used alone or combined with other data obtained regarding a subject in order to determine the state of the subject.
  • the biomarker is “differentially present” in a sample taken from a subject of one phenotypic status (e.g., having a disease) as compared with another phenotypic status (e.g., not having the disease).
  • the biomarker is “differentially present” in a sample taken from a subject undergoing no therapy or one type of therapy as compared with another type of therapy.
  • the biomarker may be “differentially present” even if there is no phenotypic difference, e.g. the biomarkers may allow the detection of asymptomatic risk.
  • a biomarker may be over-expressed (over-abundant) or under-expressed (under abundant) relative to a control.
  • the biomarker can be an allelic variant, truncated or mutated form of a wild-type nucleic acid or protein.
  • the biomarker can be a splice variant.
  • a biomarker may be determined to be “differentially present” in a variety of ways, for example, between different phenotypic statuses if the mean or median level (particularly the expression level of the associated mRNAs as described below) of the biomarker in the different groups is calculated to be statistically significant. Common tests for statistical significance include, among others, t-test, ANOVA, Kruskal-Wallis, Wilcoxon, Mann-Whitney and odds ratio.
  • a biomarker may be, for example, a small molecule, an analyte or target analyte, a nucleic acid, a protein, a metabolite or any derivative thereof or any and all combinations of these molecules, with proteins and nucleic acids finding particular use in the invention.
  • a large number of analytes may be detected using the present methods; basically, any biomarker for which a binding ligand, described below, may be made may be detected using the methods of the invention.
  • the biomarkers used in the panels of the invention can be detected either as proteins or as nucleic acids (e.g. mRNA or cDNA transcripts) in any combination.
  • the protein form of a biomarker is measured.
  • protein assays may be done using standard techniques such as ELISA assays.
  • the nucleic acid form of a biomarker e.g., the corresponding mRNA
  • one or more biomarkers from a particular panel are measured using a protein assay and one or more biomarkers from the same panel are measured using a nucleic acid assay.
  • protein refers to at least two or more peptides or amino acids joined by one or more peptide bonds.
  • a protein or an amino acid may be naturally or nonnaturally occurring and may be also be an analog, a derivative or a peptidomimetic structure.
  • protein refers to wild-type sequences, variants of wild-type sequences and either of these containing analogs or derivatized amino acids. In various embodiments, variants of the sequences described herein, including proteins and nucleic acids based on e.g.
  • splice variants variants comprising a deletion, addition, substitution, fragments, preproprotein, processed preproprotein (e.g. without a signaling peptide), processed proprotein (e.g. resulting in an active form), nonhuman sequences and variant nonhuman sequences may be used as biomarkers.
  • the biomarker is a nucleic acid.
  • nucleic acid or “oligonucleotide” or grammatical equivalents herein means at least two nucleotides covalently linked together.
  • a nucleic acid of the present invention will generally contain phosphodiester bonds, although in some cases, as outlined below, for example in the use of binding ligand probes, nucleic acid analogs are included that may have alternate backbones.
  • Biomarkers can also be bacterial nucleic acids or proteins. Over 700 species of bacteria have been identified to exist within the mouth. The presence, absence, or level of 16S rRNA from bacteria in a sample may correlate with a disease or condition. “Bacteria” refers to small prokaryotic organisms (linear dimensions of around 1 ⁇ m) with non-compartmentalized circular DNA and ribosomes of about 70 S. “16S RNA” refers to a nucleic acid component of the 30S subunit of prokaryotic ribosomes; the gene that encodes the 16S rRNA or the 16S rRNA itself. Bacterial strains of species or phylotypes have less than about a 2% difference in 16S rRNA. Closely related species or phylotypes generally have between about a 2% and about a 4% difference in 16S rRNA, whereas a genus often has between about a 5% and about a 10% difference in 16S rRNA.
  • probes on a microarray can be designed, for example, to take advantage of conserved features of the 16S rRNA gene.
  • probes complementary to the more conserved features regions identify species in a large phylogenetic group, each group corresponding to a higher taxon (for example, domain, phylum, class, order, or family).
  • Probes complementary to more variable regions distinguish genera and species.
  • Biomarkers can also include micro RNAs.
  • MicroRNAs refers to a class of small naturally occurring non-coding RNAs (18-24 nucleotides) that regulate gene expression. Many microRNAs are well conserved across species and they are present in a broad range of species: plants, nematodes, fruit flies and humans. MicroRNAs have partially or perfect complementary sequence to one or more messenger RNA molecules (mRNAs) and their main function is to negatively regulate the expression of genes. In particular, microRNAs bind to the 3′ untranslated regions of mRNAs (3-UTR) thus leading to down regulation of mRNAs in a variety of ways such as mRNA cleavage, translational repression and deadenylation.
  • mRNAs messenger RNA molecules
  • One of the first techniques used for detection and profiling of microRNAs was Northern Blotting, where hybridization is done with a complementary 32P, digoxigenin-labeled oligo or modified Locked-nucleic-acid (LNA) oligonucleotides after gel separation.
  • LNA Locked-nucleic-acid
  • RNA-binding assay a synthetic oligonucleotide, the probe, which is in an appropriate overlap-flap structure is enzymatically cleavage by a structure-specific 5* nuclease
  • in situ hybridization using fluorescent-labeled complementary probes containing chemically modified nucleotides e.g. LNAs.
  • oligonucleotide micro-array based detection platforms either with DNA capture probes or using modified Locked-nucleic-acid (LNA) oligonucleotides in which the ribose moiety is modified with an extra bridge that connects the 2′-0 and 4′-C atoms.
  • LNA Locked-nucleic-acid
  • Biomarkers can also include metabolites.
  • “Metabolite” or “small molecule” refers to organic and inorganic molecules which are present in a sample. The term does not include large macromolecules, such as large proteins (e.g., proteins with molecular weights over 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000), large nucleic acids (e.g., nucleic acids with molecular weights of over 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000), or large polysaccharides (e.g., polysaccharides with a molecular weights of over 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000).
  • large proteins e.g., proteins with molecular weights over 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000
  • large nucleic acids e.
  • a “metabolic profile”, or “small molecule profile”, means a complete or partial inventory of small molecules within a targeted cell, tissue, organ, organism, or fraction thereof (e.g., cellular compartment).
  • the inventory may include the quantity and/or type of small molecules present.
  • the “small molecule profile” may be determined using a single technique or multiple different techniques.
  • a metabolic profile can be developed by analyzing a sample using for example, techniques such as GC-MS (gas chromatography-mass spectrometry) and LC-MS (liquid chromatography-mass spectrometry).
  • techniques such as GC-MS (gas chromatography-mass spectrometry) and LC-MS (liquid chromatography-mass spectrometry).
  • biomarker panel Any combination of the biomarkers described herein is used to assemble a biomarker panel, which is detected or measured as described herein. As is generally understood in the art, a combination may refer to an entire set or any subset or subcombination thereof.
  • biomarker panel “biomarker profile,” or “biomarker fingerprint” refers to a set of biomarkers. As used herein, these terms can also refer to any form of the biomarker that is measured. Thus, if cystatin A is part of a biomarker panel, then either cystatin A mRNA, for example, or protein could be considered to be part of the panel. While individual biomarkers are useful as diagnostics, combination of biomarkers can sometimes provide greater value in determining a particular status than single biomarkers alone.
  • a biomarker panel may include 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more types of biomarkers.
  • the biomarker panel consists of a minimum number of biomarkers to generate a maximum amount of information.
  • the biomarker panel consists of 2, 3, 4, 5, 6, 7, 8, 9 or more types of biomarkers.
  • a biomarker panel “consists of” a set of biomarkers no biomarkers other than those of the set are present.
  • the biomarker panel consists of 2 biomarkers disclosed herein.
  • the biomarker panel consists of 3 biomarkers disclosed herein. In various embodiments, the biomarker panel consists of 4 biomarkers disclosed herein. In various embodiments, the biomarker paenl consists of 5 biomarkers disclosed herein.
  • the biomarker panel comprises cystatin A. In various exemplary embodiments, the biomarker panel comprises carbonic anhydrase VI.
  • the biomarker panel comprises or consists of two or more of the biomarkers selected from the group of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and CA6.
  • two or more of the biomarkers selected from the group of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and CA6 can be combined with 1, 2, 3, 4 or more additional biomarkers.
  • the biomarker panel can include any combination of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4 and the remainder of these markers.
  • a biomarker can also be a clinical parameter.
  • clinical parameter refers to all non-sample or non-analyte biomarkers of subject health status or other characteristics, such as, without limitation, age, ethnicity, gender, family history, height, and weight.
  • biomarkers of the invention show a statistically significant difference in breast cancer diagnosis.
  • diagnostic tests that use these biomarkers alone or in combination show a sensitivity and specificity of at least about 85%, at least about 90%, at least about 95%, at least about 98% and about 100%.
  • Biomarkers generally can be measured and detected through a variety of assays, methods and detection systems known to one of skill in the art.
  • the term “measuring,” “detecting,” or “taking a measurement” refers to a quantitative or qualitative determination of a property of an entity, for example, quantifying the amount or concentration of a molecule or the activity level of a molecule.
  • concentration or “level” can refer to an absolute or relative quantity. Measuring a molecule may also include determining the absence or presence of the molecule.
  • RI refractive index spectroscopy
  • UV ultra-violet spectroscopy
  • fluorescence analysis electrochemical analysis
  • radiochemical analysis radiochemical analysis
  • radiochemical analysis radiochemical analysis
  • radiochemical analysis near-infrared spectroscopy
  • near-IR near-infrared
  • IR infrared
  • NMR nuclear magnetic resonance spectroscopy
  • LS light scattering analysis
  • mass spectrometry pyrolysis mass spectrometry
  • nephelometry dispersive Raman spectroscopy
  • gas chromatography liquid chromatography
  • gas chromatography combined with mass spectrometry liquid chromatography combined with mass spectrometry
  • MALDI-TOF matrix-assisted laser desorption ionization-time of flight
  • MALDI-TOF matrix-assisted laser desorption ionization-time of flight
  • ion spray spectroscopy combined with mass spectrometry
  • capillary electrophoresis colorimetry and surface plasmon resonance
  • biomarkers can be measured using the above-mentioned detection methods, or other methods known to the skilled artisan.
  • Other biomarkers can be similarly detected using reagents that are specifically designed or tailored to detect them.
  • biomarkers can be combined in the compositions and methods of the present invention.
  • the protein form of the biomarkers is measured.
  • the nucleic acid form of the biomarkers is measured.
  • the nucleic acid form is mRNA.
  • measurements of protein biomarkers are used in conjunction with measurements of nucleic acid biomarkers.
  • sequence information provided by the database entries for the biomarker sequences
  • expression of the biomarker sequences can be detected (if present) and measured using techniques well known to one of ordinary skill in the art.
  • sequences in sequence database entries or sequences disclosed herein can be used to construct probes for detecting biomarker RNA sequences in, e.g., Northern blot hybridization analyses or methods which specifically, and, preferably, quantitatively amplify specific nucleic acid sequences.
  • sequences can be used to construct primers for specifically amplifying the biomarker sequences in, e.g., amplification-based detection methods such as reverse-transcription based polymerase chain reaction (RT-PCR).
  • amplification-based detection methods such as reverse-transcription based polymerase chain reaction (RT-PCR).
  • RT-PCR reverse-transcription based polymerase chain reaction
  • sequence comparisons in test and reference populations can be made by comparing relative amounts of the examined DNA sequences in the test and reference cell populations.
  • RNA can also be measured using, for example, other target amplification methods (e.g., TMA, SDA, NASBA), signal amplification methods (e.g., bDNA), nuclease protection assays, in situ hybridization and the like.
  • biochip assays are biochip assays.
  • biochip or “chip” herein is meant a composition generally comprising a solid support or substrate to which a capture binding ligand (also called an adsorbent, affinity reagent or binding ligand, or when nucleic acid is measured, a capture probe) is attached and can bind either proteins, nucleic acids or both.
  • a capture binding ligand also called an adsorbent, affinity reagent or binding ligand, or when nucleic acid is measured, a capture probe
  • the protein biomarkers are measured on a chip separate from that used to measure the nucleic acid biomarkers.
  • biomarkers are measured on the same platform, such as on one chip.
  • biomarkers are measured using different platforms and/or different experimental runs.
  • binding ligand By “binding ligand,” “capture binding ligand,” “capture binding species,” “capture probe” or grammatical equivalents herein is meant a compound that is used to detect the presence of or to quantify, relatively or absolutely, a target analyte, target species or target sequence (all used interchangeably) and that will bind to the target analyte, target species or target sequence.
  • the capture binding ligand or capture probe allows the attachment of a target species or target sequence to a solid support for the purposes of detection as further described herein. Attachment of the target species to the capture binding ligand may be direct or indirect.
  • the target species is a biomarker.
  • the composition of the binding ligand will depend on the composition of the biomarker. Binding ligands for a wide variety of biomarkers are known or can be readily found using known techniques. For example, when the biomarker is a protein, the binding ligands include proteins (particularly including antibodies or fragments thereof (F ab s, etc.) as discussed further below) or small molecules. The binding ligand may also have cross-reactivity with proteins of other species. Antigen-antibody pairs, receptor-ligands, and carbohydrates and their binding partners are also suitable analyte-binding ligand pairs. In various embodiments, the binding ligand may be nucleic acid.
  • Nucleic acid binding ligands find particular use when proteins are the targets; alternatively, as is generally described in U.S. Pat. Nos. 5,270,163; 5,475,096; 5,567,588; 5,595,877; 5,637,459; 5,683,867; 5,705,337 and related patents, hereby incorporated by reference, nucleic acid “aptamers” can be developed for binding to virtually any biomarker. Nucleic acid binding ligands also find particular use when nucleic acids are binding targets. There is a wide body of literature relating to the development of binding partners based on combinatorial chemistry methods. In these embodiments, when the binding ligand is a nucleic acid, preferred compositions and techniques are outlined in PCT Publication WO/1998/020162, hereby incorporated by reference.
  • the capture binding ligand is an antibody. These embodiments are particularly useful for the detection of the protein form of a biomarker.
  • Detecting or measuring the level (e.g. the transcription level) of a biomarker involves binding of the biomarker to a capture binding ligand, generally referred to herein as a “capture probe” when the mRNA of the biomarker is to be detected on a solid support.
  • the biomarker is a target sequence.
  • target sequence or “target nucleic acid” or grammatical equivalents herein means a nucleic acid sequence that may be a portion of a gene, a regulatory sequence, genomic DNA, cDNA, RNA including mRNA and rRNA, or others.
  • the target sequence may be a target sequence from a sample, or a secondary target such as a product of an amplification reaction such as PCR etc.
  • measuring a nucleic acid can thus refer to measuring the complement of the nucleic acid. It may be any length, with the understanding that longer sequences are more specific.
  • the target sequence may also comprise different target domains; for example, a first target domain of the sample target sequence may hybridize to a first capture probe, a second target domain may hybridize to a label probe (e.g. a “sandwich assay” format), etc.
  • the target domains may be adjacent or separated as indicated.
  • first and second are not meant to confer an orientation of the sequences with respect to the 5′-3′ orientation of the target sequence. For example, assuming a 5′-3′ orientation of the target sequence, the first target domain may be located either 5′ to the second domain, or 3′ to the second domain.
  • the assays of the invention can take on a number of embodiments.
  • the assays are done in solution format, using any number of solution based formats.
  • end-point or real time PCR formats are used, as are well known in the art. These assays can be done either as a panel, in individual tubes or wells, or as multiplex assays, using sets of primers and different labels within a single tube or well.
  • other formats can be utilized, including, but not limited to for example ligation based assays utilizing FRET dye pairs. In this embodiment, only upon ligation of two (or more) probes hybridized to the target sequence is a signal generated.
  • the assays are done on a solid support, utilizing a capture probe associated with the surface.
  • the capture probes (or capture binding ligands, as they are sometimes referred to) can be covalently attached to the surface, for example using capture probes terminally modified with functional groups, for example amino groups, that are attached to modified surfaces such as silanized glass.
  • non-covalent attachment such as electrostatic, hydrophobic/hydrophilic adhesion can be utilized.
  • a large number of attachments are possible on a wide variety of surfaces.
  • the assays can take on a number of formats.
  • the target sequence comprises a detectable label, as described herein.
  • the label is generally added to the target sequence during amplification of the target in one of two ways: either labeled primers are utilized during the amplification step or labeled dNTPs are used, both of which are well known in the art.
  • the label can either be a primary or secondary label as discussed herein.
  • the label on the primer and/or a dNTP is a primary label such as a fluorophore.
  • the label may be a secondary label such as biotin or an enzyme; for example, in one embodiment, the primers or dNTPs are labeled with biotin, and then a streptavidin/label complex is added.
  • the streptavidin/label complex contains a label such as a fluorophore.
  • the streptavidin/label complex comprises an enzymatic label.
  • the complex can comprise horseradish peroxidase, and upon addition of TMB, the action of the horseradish peroxidase causes the TMB to precipitate, causing an optically detectable event. This has a particular benefit in that the optics for detection does not require the use of a fluorimeter.
  • the solid phase assay relies on the use of a labeled soluble capture ligand, sometimes referred to as a “label probe” or “signaling probe” when the target analyte is a nucleic acid.
  • the assay is a “sandwich” type assay, where the capture probe binds to a first domain of the target sequence and the label probe binds to a second domain.
  • the label probe can also be either a primary (e.g. a fluorophore) or a secondary (biotin or enzyme) label.
  • the label probe comprises biotin, and a streptavidin/enzyme complex is used, as discussed herein.
  • the complex can comprise horseradish peroxidase, and upon addition of TMB, the action of the horseradish peroxidase causes the TMB to precipitate, causing an optically detectable event.
  • Detection of a target species requires a “label” or “detectable marker” (as described below) that can be incorporated in a variety of ways.
  • the composition comprises a “label” or a “detectable marker.”
  • the target species or target analyte or target sequence
  • binding of the target species thus provides the label at the surface of the solid support.
  • a sandwich format in which target species are unlabeled.
  • a “capture” or “anchor” binding ligand is attached to the detection surface as described herein, and a soluble binding ligand (frequently referred to herein as a “signaling probe,” “label probe” or “soluble capture ligand”) binds independently to the target species and either directly or indirectly comprises at least one label or detectable marker.
  • label or “labeled” herein is meant that a compound has at least one molecule, element, isotope or chemical compound attached to enable the detection of the compound.
  • labels fall into four classes: a) isotopic labels, which may be radioactive or heavy isotopes; b) magnetic, electrical, thermal; c) colored or luminescent dyes; and d) enzymes; although labels include particles such as magnetic particles as well.
  • the dyes may be chromophores or phosphors but are preferably fluorescent dyes, which due to their strong signals provide a good signal-to-noise ratio for decoding.
  • Suitable dyes for use in the invention include, but are not limited to, fluorescent lanthanide complexes, including those of Europium and Terbium, fluorescein, rhodamine, tetramethylrhodamine, eosin, erythrosin, coumarin, methyl-coumarins, pyrene, Malacite green, stilbene, Lucifer Yellow, Cascade Blue, Texas Red, Alexa dyes and others described in the 6th Edition of the Molecular Probes Handbook by Richard P. Haugland, hereby expressly incorporated by reference. Additional labels include nanocrystals or Q-dots as described in U.S. Pat. No. 6,544,732 incorporated by reference.
  • a secondary detectable label is used.
  • a secondary label is one that is indirectly detected; for example, a secondary label can bind or react with a primary label for detection, can act on an additional product to generate a primary label (e.g. enzymes), or may allow the separation of the compound comprising the secondary label from unlabeled materials, etc.
  • Secondary labels include, but are not limited to, one of a binding partner pair; chemically modifiable moieties; nuclease inhibitors, enzymes such as horseradish peroxidase, alkaline phosphatases, lucifierases, etc. Secondary labels can also include additional labels.
  • the secondary label is a binding partner pair.
  • the label may be a hapten or antigen, which will bind its binding partner.
  • suitable binding partner pairs include, but are not limited to: antigens (such as proteins (including peptides)) and antibodies (including fragments thereof (F ab s, etc.)); proteins and small molecules, including biotin/streptavidin; enzymes and substrates or inhibitors; other protein-protein interacting pairs; receptor-ligands; and carbohydrates and their binding partners. Nucleic acid-nucleic acid binding proteins pairs are also useful. In general, the smaller of the pair is attached to the NTP for incorporation into the primer.
  • Preferred binding partner pairs include, but are not limited to, biotin (or imino-biotin) and streptavidin, digeoxinin and Abs, and ProlinxTM reagents.
  • an enzyme serves as the secondary label, bound to the soluble capture ligand.
  • the soluble capture ligand comprises biotin, which is then bound to a enzyme-streptavidin complex and forms a colored precipitate with the addition of TMB.
  • the label or detectable marker is a conjugated enzyme (for example, horseradish peroxidase).
  • the system relies on detecting the precipitation of a reaction product or on a change in, for example, electronic properties for detection.
  • none of the compounds comprises a label.
  • fluorescent signal generating moiety or “fluorophore” refers to a molecule or part of a molecule that absorbs energy at one wavelength and re-emits energy at another wavelength. Fluorescent properties that can be measured include fluorescence intensity, fluorescence lifetime, emission spectrum characteristics, energy transfer, and the like.
  • Signals from single molecules can be generated and detected by a number of detection systems, including, but not limited to, scanning electron microscopy, near field scanning optical microscopy (NSOM), total internal reflection fluorescence microscopy (TIRFM), and the like.
  • NOM near field scanning optical microscopy
  • TRFM total internal reflection fluorescence microscopy
  • a detection system for fluorophores includes any device that can be used to measure fluorescent properties as discussed above.
  • the detection system comprises an excitation source, a fluorophore, a wavelength filter to isolate emission photons from excitation photons and a detector that registers emission photons and produces a recordable output, in some embodiments as an electrical signal or a photographic image.
  • detection devices include without limitation spectrofluorometers and microplate readers, fluorescence microscopes, fluorescence scanners (including e.g. microarray readers) and flow cytometers.
  • the binding of the biomarker to the binding ligand is specific or selective, and the binding ligand is part of a binding pair.
  • binding ligand is part of a binding pair.
  • solid support refers to any material that can be modified to contain discrete individual sites appropriate for the attachment or association of a capture binding ligand.
  • Suitable substrates include metal surfaces such as gold, electrodes, glass and modified or functionalized glass, plastics (including acrylics, polystyrene and copolymers of styrene and other materials, polypropylene, polyethylene, polybutylene, polycarbonate, polyurethanes, Teflon, derivatives thereof, etc.), polysaccharides, nylon or nitrocellulose, resins, mica, silica or silica-based materials including silicon and modified silicon, carbon, metals, inorganic glasses, fiberglass, ceramics, GETEK (a blend of polypropylene oxide and fiberglass) and a variety of other polymers.
  • ClonDiag materials described below.
  • the surface of a biochip comprises a plurality of addressable locations, each of which comprises a capture binding ligand.
  • An “array location,” “addressable location,” “pad” or “site” herein means a location on the substrate that comprises a covalently attached capture binding ligand.
  • An “array” herein means a plurality of capture binding ligands in a regular, ordered format, such as a matrix. The size of the array will depend on the composition and end use of the array. Arrays containing from about two or more different capture binding ligands to many thousands can be made. Generally, the array will comprise 3, 4, 5, 6, 7 or more types of capture binding ligands depending on the end use of the array.
  • the array can include controls, replicates of the markers and the like. Exemplary ranges are from about 3 to about 50.
  • the compositions of the invention may not be in array format; that is, for some embodiments, compositions comprising a single capture ligand may be made as well.
  • multiple substrates may be used, either of different or identical compositions. Thus for example, large arrays may comprise a plurality of smaller substrates.
  • the invention provides a composition comprising a solid support comprising a capture binding ligand for each biomarker of a biomarker panel.
  • the capture ligand is a nucleic acid.
  • the capture binding ligand is an antibody.
  • the composition further comprises a soluble binding ligand for each biomarker of a biomarker panel.
  • compositions and methods of the present invention can be implemented with array platforms such as GeneChip® (Affymetrix), CodeLinkTM Bioarray (Amersham), Expression Array System (Applied Biosystems), SurePrint microarrays (Agilent), Sentrix® LD BeadChip or Sentrix® Array Matrix (Illumina) and Verigene (Nanosphere).
  • array platforms such as GeneChip® (Affymetrix), CodeLinkTM Bioarray (Amersham), Expression Array System (Applied Biosystems), SurePrint microarrays (Agilent), Sentrix® LD BeadChip or Sentrix® Array Matrix (Illumina) and Verigene (Nanosphere).
  • detection and measurement of biomarkers utilizes colorimetric methods and systems in order to provide an indication of binding of a target analyte or target species.
  • colorimetric methods the presence of a bound target species such as a biomarker will result in a change in the absorbance or transmission of light by a sample or substrate at one or more wavelengths. Detection of the absorbance or transmission of light at such wavelengths thus provides an indication of the presence of the target species.
  • a detection system for colorimetric methods includes any device that can be used to measure colorimetric properties as discussed above.
  • the device is a spectrophotometer, a colorimeter or any device that measures absorbance or transmission of light at one or more wavelengths.
  • the detection system comprises a light source; a wavelength filter or monochromator; a sample container such as a cuvette or a reaction vial; a detector, such as a photoresistor, that registers transmitted light; and a display or imaging element.
  • a ClonDiag chip platform is used for the colorimetric detection of biomarkers.
  • a ClonDiag ArrayTube (AT) is used.
  • One unique feature of the ArrayTube is the combination of a micro probe array (the biochip) and micro reaction vial.
  • detection of the target sequence is done by amplifying and biotinylating the target sequence contained in a sample and optionally digesting the amplification products. The amplification product is then allowed to hybridize with probes contained on the ClonDiag chip.
  • a solution of a streptavidin-enzyme conjugate such as Poly horseradish peroxidase (HRP) conjugate solution
  • HRP horseradish peroxidase
  • a dye solution such as o-dianisidine substrate solution is contacted with the chip. Oxidation of the dye results in precipitation that can be detected colorimetrically.
  • TMB 3,3′,5,5′-tetramethylbenzidine
  • the ArrayTube biochip comprises capture binding ligands such as antibodies.
  • a sample is contacted with the biochip, and any target species present in the sample is allowed to bind to the capture binding ligand antibodies.
  • a soluble capture binding ligand or a detection compound such as a horseradish peroxidase conjugated antibody is allowed to bind to the target species.
  • a dye, such as TMB, is then added and allowed to react with the horseradish peroxidase, causing precipitation and a color change that is detected by a suitable detection device.
  • Suitable reader instruments and detection devices include the ArrayTube Workstation ATS and the ATR 03.
  • the ClonDiag ArrayStrip (AS) can be used.
  • the ArrayStrip provides a 96-well format for high volume testing.
  • Each ArrayStrip consists of a standard 8-well strip with a microarray integrated into the bottom of each well. Up to 12 ArrayStrips can be inserted into one microplate frame enabling the parallel multiparameter testing of up to 96 samples.
  • the ArrayStrip can be processed using the ArrayStrip Processor ASP, which performs all liquid handling, incubation, and detection steps required in array based analysis.
  • a method of using the ArrayStrip to detect the protein comprises conditioning the AS array with buffer or blocking solution; loading of up to 96 sample solutions in the AS wells to allow for binding of the protein; 3 ⁇ washing; conjugating with a secondary antibody linked to HRP; 3 ⁇ washing; precipitation staining with TMB; and AS array imaging and optional data storage.
  • immunoassays carried out in accordance with the present invention may be homogeneous assays or heterogeneous assays.
  • the immunological reaction usually involves the specific antibody (e.g., anti-biomarker protein antibody), a labeled analyte, and the sample of interest.
  • the signal arising from the label is modified, directly or indirectly, upon the binding of the antibody to the labeled analyte.
  • Both the immunological reaction and detection of the extent thereof can be carried out in a homogeneous solution.
  • Immunochemical labels which may be employed include free radicals, radioisotopes, fluorescent dyes, enzymes, bacteriophages, or coenzymes.
  • the reagents are usually the sample, the antibody, and means for producing a detectable signal.
  • Samples as described above may be used.
  • the antibody can be immobilized on a support, such as a bead (such as protein A and protein G agarose beads), plate or slide, and contacted with the specimen suspected of containing the antigen in a liquid phase.
  • the support is then separated from the liquid phase and either the support phase or the liquid phase is examined for a detectable signal employing means for producing such signal.
  • the signal is related to the presence of the analyte in the sample.
  • Means for producing a detectable signal include the use of radioactive labels, fluorescent labels, or enzyme labels.
  • an antibody which binds to that site can be conjugated to a detectable group and added to the liquid phase reaction solution before the separation step.
  • the presence of the detectable group on the solid support indicates the presence of the antigen in the test sample.
  • suitable immunoassays include immunoblotting, immunofluorescence methods, immunoprecipitation, chemiluminescence methods, electrochemiluminescence (ECL) or enzyme-linked immunoassays.
  • Antibodies can be conjugated to a solid support suitable for a diagnostic assay (e.g., beads such as protein A or protein G agarose, microspheres, plates, slides or wells formed from materials such as latex or polystyrene) in accordance with known techniques, such as passive binding.
  • Antibodies as described herein may likewise be conjugated to detectable labels or groups such as radiolabels (e.g., 35 S, 125 I, 131 I), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescent labels (e.g., fluorescein, Alexa, green fluorescent protein, rhodamine) in accordance with known techniques.
  • a diagnostic assay e.g., beads such as protein A or protein G agarose, microspheres, plates, slides or wells formed from materials such as latex or polystyrene
  • Antibodies as described herein may likewise be conjugated to detectable labels or groups such as radiolabel
  • the invention provides a method of assaying a sample from a patient to determine concentrations of a biomarker panel in the sample.
  • the method comprises contacting the sample with a composition comprising a solid support comprising a capture binding ligand or capture probe for each biomarker of a biomarker panel.
  • kits for use in determining breast health or breast cancer status for a number of medical (including diagnostic and therapeutic), industrial, forensic and research applications.
  • Kits may comprise a carrier, such as a box, carton, tube or the like, having in close confinement therein one or more containers, such as vials, tubes, ampoules, bottles, pouches, envelopes and the like.
  • the kits comprise one or more components selected from one or more media or media ingredients and reagents for the measurement of the various biomarkers and biomarker panels disclosed herein.
  • kits of the invention may also comprise, in the same or different containers, one or more DNA polymerases, one or more primers, one or more suitable buffers, one or more nucleotides (such as deoxynucleoside triphosphates (dNTPs) and preferably fluorescently labeled dNTPs) and labeling components.
  • the one or more components may be contained within the same container, or may be in separate containers to be admixed prior to use.
  • the kits of the present invention may also comprise one or more instructions or protocols for carrying out the methods of the present invention.
  • the kits may also comprise a computer or a component of a computer, such as a computer-readable storage medium or device.
  • Examples of storage media include, without limitation, optical disks such as CD, DVD and Blu-ray Discs (BD); magneto-optical disks; magnetic media such as magnetic tape and internal hard disks and removable disks; semi-conductor memory devices such as EPROM, EEPROM and flash memory; and RAM.
  • the computer-readable storage medium may comprise software encoding references to the various therapies and treatment regimens disclosed herein.
  • the software may be interpreted by a computer to provide the practitioner with treatments according to various measured concentrations of biomarkers as provided herein.
  • the kit comprises a biomarker assay involving a lateral-flow-based point-of-care rapid test with detection of risk thresholds, or a biochip with quantitative assays for the constituent biomarkers.
  • compositions and methods of the present invention can be used in the prognosis, diagnosis and treatment of disease in a subject.
  • the invention provides compositions and methods for laboratory and point-of-care tests for measuring biomarkers in a sample from a subject.
  • the invention can be generally applied for a number of different diseases.
  • the disease is breast cancer.
  • biomarkers and biomarker panels disclosed herein can be used in methods to diagnose, identify or screen subjects that have, do not have or are at risk for having disease; to monitor subjects that are undergoing therapies for disease; to determine or suggest a new therapy or a change in therapy; to differentially diagnose disease states associated with the disease from other diseases or within sub-classifications of disease; to evaluate the severity or changes in severity of disease in a patient; to stage a subject with the disease and to select or modify therapies or interventions for use in treating subjects with the disease.
  • the methods of the present invention are used to identify and/or diagnose subjects who are asymptomatic or presymptomatic for a disease.
  • “asymptomatic” or presymptomatic” means not exhibiting the traditional symptoms or enough abnormality for disease.
  • a method of determining a prognosis of a disease in a subject, diagnosing a disease in a subject, or treating a disease in a subject comprises taking a measurement of a biomarker panel in a sample from the subject.
  • the biomarker panel consists of two or more of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and/or CA6.
  • disease status includes any distinguishable manifestation of the disease, including non-disease.
  • disease status includes, without limitation, the presence or absence of disease, the risk of developing disease, the stage of the disease, the progression of disease (e.g., progress of disease or remission of disease over time), the severity of disease and the effectiveness or response to treatment of disease.
  • a “subject” in the context of the present invention is an animal, preferably a mammal.
  • the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples.
  • a subject is human and may be referred to as a patient.
  • Mammals other than humans can be advantageously used as subjects that represent animal models of a disease or for veterinarian applications.
  • a subject can be one who has been previously diagnosed or identified as having a disease, and optionally has already undergone, or is undergoing, a therapeutic intervention for a disease. Alternatively, a subject can also be one who has not been previously diagnosed as having a disease.
  • a subject can be one who exhibits one or more risk factors for a disease, or one who does not exhibit a disease risk factor, or one who is asymptomatic for a disease.
  • a subject can also be one who is suffering from or at risk of developing a disease.
  • the subject can be already undergoing therapy or can be a candidate for therapy.
  • biomarkers may be measured in using several techniques designed to achieve more predictable subject and analytical variability.
  • sample refers to a specimen or culture obtained from a subject and includes fluids, gases and solids including for example tissue.
  • the sample comprises saliva.
  • wash steps and/or fragmentation may be applied to a sample.
  • a biomarker panel is measured directly in a subject without the need to obtain a separate sample from the patient.
  • the invention provides a method of diagnosing a subject for a disease comprising taking a measurement of a biomarker panel; and correlating the measurement with the disease.
  • correlating generally refers to determining a relationship between one type of data with another or with a state.
  • correlating the measurement with disease comprises comparing the measurement with a reference biomarker profile or some other reference value.
  • correlating the measurement with disease comprises determining whether the subject is currently in a state of disease.
  • the quantity or activity measurements of a biomarker panel can be compared to a reference value. Differences in the measurements of biomarkers in the subject sample compared to the reference value are then identified.
  • the reference value is given by a risk category as described further below.
  • the reference value is a baseline value.
  • a baseline value is a composite sample of an effective amount of biomarkers from one or more subjects who do not have a disease, who are asymptomatic for a disease or who have a certain level of a disease.
  • a baseline value can also comprise the amounts of biomarkers in a sample derived from a subject who has shown an improvement in risk factors of a disease as a result of treatments or therapies. In these embodiments, to make comparisons to the subject-derived sample, the amounts of biomarkers are similarly calculated.
  • a reference value can also comprise the amounts of biomarkers derived from subjects who have a disease confirmed by an invasive or non-invasive technique, or are at high risk for developing a disease.
  • subjects identified as having a disease, or being at increased risk of developing a disease are chosen to receive a therapeutic regimen to slow the progression of a disease, or decrease or prevent the risk of developing a disease.
  • a disease is considered to be progressive (or, alternatively, the treatment does not prevent progression) if the amount of biomarker changes over time relative to the reference value, whereas a disease is not progressive if the amount of biomarkers remains constant over time (relative to the reference population, or “constant” as used herein).
  • the term “constant” as used in the context of the present invention is construed to include changes over time with respect to the reference value.
  • the biomarkers of the present invention can be used to generate a “reference biomarker profile” of those subjects who do not have a disease according to a certain threshold, are not at risk of having a disease or would not be expected to develop a disease.
  • the biomarkers disclosed herein can also be used to generate a “subject biomarker profile” taken from subjects who have a disease or are at risk for having a disease.
  • the subject biomarker profiles can be compared to a reference biomarker profile to diagnose or identify subjects at risk for developing a disease, to monitor the progression of disease, as well as the rate of progression of disease, and to monitor the effectiveness of disease treatment modalities.
  • the reference and subject biomarker profiles of the present invention can be contained in a machine-readable medium, such as but not limited to, analog tapes like those readable by a VCR; optical media such as CD-ROM, DVD-ROM and the like; and solid state memory, among others.
  • a machine-readable medium such as but not limited to, analog tapes like those readable by a VCR; optical media such as CD-ROM, DVD-ROM and the like; and solid state memory, among others.
  • the invention provides methods of treating a disease in a subject comprising taking a measurement of a biomarker panel in a sample from the subject, and affecting a therapy with respect to the subject.
  • the terms “therapy” and “treatment” may be used interchangeably.
  • the therapy can be selected from, without limitation, initiating therapy, continuing therapy, modifying therapy or ending therapy.
  • a therapy also includes any prophylactic measures that may be taken to prevent disease.
  • treatment comprises administering a disease-modulating drug to a subject.
  • the drug can be a therapeutic or prophylactic used in subjects diagnosed or identified with a disease or at risk of having the disease.
  • modifying therapy refers to altering the duration, frequency or intensity of therapy, for example, altering dosage levels.
  • effecting a therapy comprises causing a subject to or communicating to a subject the need to make a change in lifestyle, for example, increasing exercise, changing diet, reducing or eliminating smoking and so on.
  • the therapy can also include surgery, for example, mastectomy.
  • Measurement of biomarker levels allow for the course of treatment of a disease to be monitored.
  • the effectiveness of a treatment regimen for a disease can be monitored by detecting one or more biomarkers in an effective amount from samples obtained from a subject over time and comparing the amount of biomarkers detected. For example, a first sample can be obtained prior to the subject receiving treatment and one or more subsequent samples are taken after or during treatment of the subject. Changes in biomarker levels across the samples may provide an indication as to the effectiveness of the therapy.
  • a test sample from the subject can also be exposed to a therapeutic agent or a drug, and the level of one or more biomarkers can be determined. Biomarker levels can be compared to a sample derived from the subject before and after treatment or exposure to a therapeutic agent or a drug, or can be compared to samples derived from one or more subjects who have shown improvements relative to a disease as a result of such treatment or exposure.
  • the invention provides a method of assessing the efficacy of a therapy with respect to a subject comprising taking a first measurement of a biomarker panel in a first sample from the subject; effecting the therapy with respect to the subject; taking a second measurement of the biomarker panel in a second sample from the subject and comparing the first and second measurements to assess the efficacy of the therapy.
  • therapeutic or prophylactic agents suitable for administration to a particular subject can be identified by detecting a biomarker (which may be two or more) in an effective amount from a sample obtained from a subject and exposing the subject-derived sample to a test compound that determines the amount of the biomarker(s) in the subject-derived sample.
  • treatments or therapeutic regimens for use in subjects having a disease or subjects at risk for developing a disease can be selected based on the amounts of biomarkers in samples obtained from the subjects and compared to a reference value. Two or more treatments or therapeutic regimens can be evaluated in parallel to determine which treatment or therapeutic regimen would be the most efficacious for use in a subject to delay onset, or slow progression of a disease.
  • a recommendation is made on whether to initiate or continue treatment of a disease.
  • effecting a therapy comprises administering a disease-modulating drug to the subject.
  • the subject may be treated with one or more disease-modulating drugs until altered levels of the measured biomarkers return to a baseline value measured in a population not suffering from the disease, experiencing a less severe stage or form of a disease or showing improvements in disease biomarkers as a result of treatment with a disease-modulating drug.
  • improvements related to a changed level of a biomarker or clinical parameter may be the result of treatment with a disease-modulating drug.
  • a number of compounds such as a disease-modulating drug may be used to treat a subject and to monitor progress using the methods of the invention.
  • the disease-modulating drug comprises
  • Any drug or combination of drugs disclosed herein may be administered to a subject to treat a disease.
  • the drugs herein can be formulated in any number of ways, often according to various known formulations in the art or as disclosed or referenced herein.
  • any drug or combination of drugs disclosed herein is not administered to a subject to treat a disease.
  • the practitioner may refrain from administering the drug or combination of drugs, may recommend that the subject not be administered the drug or combination of drugs or may prevent the subject from being administered the drug or combination of drugs.
  • one or more additional drugs may be optionally administered in addition to those that are recommended or have been administered.
  • An additional drug will typically not be any drug that is not recommended or that should be avoided.
  • one or more additional drugs comprise one or more glucose lowering drugs.
  • the therapy chosen by a practitioner can depend on the concentrations of biomarkers determined in a sample. In various exemplary embodiments, the therapy depends on which category from a range of categories particular to each biomarker the measured concentration of each biomarker falls in. In various exemplary embodiments, the therapy depends on the combination of risk levels for different symptoms or diseases that are indicated by a biomarker panel.
  • the term “category” refers to a subset of a partition of the possible concentrations that a biomarker may have. Each category may be associated with a label or classification chosen by the practitioner.
  • the labels may be refer to, for example, the risk level of an individual for having or being subject to a disease state. The categories and labels may be derived from the current literature or according to the findings of the practitioner.
  • Each biomarker of a biomarker panel can thus be associated with a discrete set of categories, for example, risk categories. Combining one category from each biomarker forms a “decision point.”
  • the complete set of decision points comprises all possible n-tuples of categories, wherein n is the number of biomarkers in the biomarker panel. This complete set will have m 1 ⁇ m 2 ⁇ . . . m n possible decision points, wherein in is the number of categories for biomarker i.
  • Every decision point can be associated with a condition or a disease state, which is not necessarily unique. That is, one or more decision points can be associated with the same disease state.
  • the association of every possible decision point with a condition or disease state can be referred to as a “disease classification matrix” or a “disease classification tree.”
  • Every decision point can also be associated with a particular therapy, which is not necessarily unique. That is, one or more decision points can be associated with the same therapy.
  • the association of every possible decision point with one or more therapies can be referred to as a “therapy decision matrix” or “therapy decision tree.”
  • Each decision point can be associated with more than one type of information. For example, both disease state and therapy can be indicated by a decision point.
  • Unstimulated whole saliva samples were collected with previously established protocols. Subjects were asked to refrain from eating, drinking, smoking, or oral hygiene procedures for at least 30 minutes before the collection. Lipstick was wiped off, and the subject rinsed her mouth once with plain water. Typically, patients donated approximately 5-10 ml of saliva. Samples were then centrifuged at 2,600 g for 15 minutes at 4° C. The supernatant was then stored at ⁇ 80° C. until use. Of note, protease inhibitors cocktail, containing 1 ⁇ l aprotinin, 10 ⁇ l PMSF (phenylmethanesulfonyl fluoride) and 3 ⁇ l sodium orthovanadate (all from Sigma, St. Louis, Mo.) were added to each 1 ml saliva sample.
  • PMSF phenylmethanesulfonyl fluoride
  • 3 ⁇ l sodium orthovanadate all from Sigma, St. Louis, Mo.
  • cDNA was transcribed and biotinylated using GeneChip® Expression 3′-Amplification Reagents for in vitro transcription labeling (Affymetrix, Santa Clara, Calif.). Approximately 20 ⁇ g of labeled RNA were subsequently submitted for GeneChip® analysis using an Affymetrix Human Genome U133 Plus 2.0 Array. Chip hybridization and scanning were performed using the MIAME (Minimum Information About a Microarray Experiment) criteria. All Affymetrix Human Genome U133 Plus 2.0 Array data generated in this study were uploaded to the GEO database, accession number GSE20266.
  • the CEL files from all databases were imported into the statistical R 2.7.0 (hypertext transfer protocol://www.r-project.org) with same and ROC packages.
  • the Probe Logarithmic Intensity Error Estimation (PLIER) expression measures were computed after background correction and quantile normalization for each microarray dataset. Probeset-level quantile normalization was performed across all samples to make the effect sizes similar among all datasets. Finally, for every probeset, significance analysis of microarray (SAM) was applied to identify differential expression between the cancer and healthy control samples. The probesets were then ranked by the false discovery rate (FDR) corrected p-values.
  • PIER Probe Logarithmic Intensity Error Estimation
  • the biomarker candidates generated by microarray profiling were subjected to further screening by real-time quantitative RT-PCR (qPCR) on the same set of samples used for the microarray analysis.
  • total RNA was reverse-transcribed using reverse transcriptase and gene-specific primers using the following thermal cycling conditions: 1 min at 60° C., 30 min at 50° C., 2 min at 95° C., followed by 15 cycles of 15s at 95° C., 30s at 50° C., 10 s at 72° C. These steps were followed with a final extension of 5 min. at 72° C. and then cooling to 4° C.
  • the preamplified product was cleaned using ExoSAP-IT (USB Corporation) and diluted 1/40 in water. 2 ⁇ l of the cDNA was used for qPCR.
  • qPCR was carried out in a 96-well plate in a reaction volume of 10 ⁇ l using power SYBR®-Green Master Mix (Applied Biosystems, Foster City, Calif.) for 15 min at 95° C. for initial denaturing, followed by 40 cycles of 95° C. for 30 s and 60° C. for 30 s in the ABI 7500HT Fast Real Time PCR system (Applied Biosystems, Foster City, Calif.). All qPCRS were performed in duplicate for all candidate mRNA. The specificity of the PCR was confirmed according to the melting curve of each gene, and the average threshold cycle (Ct) was examined.
  • Ct average threshold cycle
  • RT-qPCR primers were designed using Primer Express 3.0 software (Applied Biosystems, Foster City, Calif.). All primers were synthesized by Sigma-Genosys (Woodlands, Tex.), and the amplicons were intron spanning whenever possible.
  • Raw data were normalized by subtracting GAPDH Ct values from the biomarker Ct values to generate ⁇ Ct.
  • the Mann-Whitney rank sum test was used for between-group biomarker comparisons.
  • the data analysis for qPCR was performed using the 2 ⁇ Ct method, where GAPDH is used as the reference gene.
  • the qPCR based gene expression values between two groups were compared using the non-parametric Wilcoxon test.
  • To normalize for RNA input qPCR was also performed for GAPDH.
  • Raw data were normalized by subtracting GAPDH Ct values from the marker Ct values to provide ⁇ Ct and then analyze with the stats, utilities packages from R 2.7.0 (world wide web.r-project.org) and the ROC package from Bioconductor 2.2 (world wide web.bioconductor.org).
  • Statistical comparisons were made with the use of the Mann-Whitney U test with consideration of two different distributions for control and pancreatic cancer groups.
  • Biomarkers that differentiated between groups of subjects were identified and compared by Area Under Curve (AUC) value.
  • the AUC is based on constructing a receiver operating characteristic (ROC) curve which plots the sensitivity versus one minus the specificity.
  • the AUC value is computed by numerical integration of the ROC curve.
  • the range of this value can be 0.5 to 1.0. A value of 0.5 indicates that the biomarker is no better that a coin toss, while 1.0 indicates the relatively best diagnostic accuracy.
  • Saliva from 13 healthy control subjects and 13 breast cancer subjects were centrifuged at 2600 g at 4° C. for 15 minutes.
  • Saliva supernatant from the 13 health control subjects and 13 breast cancer subjects were pooled to form a control sample and a cancer sample for proteomic profiling.
  • 250 ⁇ g of proteins in the pooled saliva samples were precipitated by methanol and then resuspended in 2-D cell lysis buffer (30 mM Tris-HCl, pH 8.8, containing 7M urea, 2M thiourea and 4% CHAPS detergent).
  • the total proteins of each pooled sample, breast cancer and control were labeled with the cyanine dyes Cy2 and Cy5 respectively.
  • the two labeled sample sets were then combined and subjected to two-dimensional difference gel electrophoresis.
  • isoelectric focusing (IEF) (pH3-10) was run following the protocol provided by Amersham BioSciences.
  • the IPG strips were rinses in the SDS-gel running buffer before transferring to 13.5% SDS-gels.
  • the SDS-gels were run at 15° C. until the dye front ran out of the gels.
  • Gel images were scanned immediately following the SDS-PAGE using Typhoon TRIOTM (Amersham BioSciences). The fold change of the protein expression levels was obtained from in-gel DeCyderTM analysis.
  • the desalted peptides were mixed with CHCA matrix ( ⁇ -cyano-4-hydroxycinnamic acid) and spotted into wells of a MALDI plate for MALDI-TOF MS (ABI4800) identification.
  • Protein identification was based on peptide fingerprint mass mapping (using MS spectra) and peptide fragmentation mapping (using MS/MS spectra).
  • Combined MS and MS/MS spectra were submitted for database search using GPS Explorer software equipped with the MASCOT search engine to identify proteins from primary sequence databases.
  • the membrane was then washed in wash buffer containing 10 mM Tris-HCl, pH 7.6, 150 mM NaCl, and 0.1% (v/v) Tween®-20 (Sigma-Aldrich) before blocking for one hour in wash buffer containing 5% non-fat dry milk. After further washes in wash buffer, the membrane was incubated with primary antibody (mouse anti-human carbonic anhydrase VI (Lifespan Biotech) at 1 ⁇ g/ml, mouse anti-psoriasin (Abeam) at 1 ⁇ g/ml, mouse anti-actin (Sigma-Aldrich) at 1 ⁇ g/ml according to manufacturers instructions in blocking buffer at room temperature for 2 h.
  • primary antibody mouse anti-human carbonic anhydrase VI (Lifespan Biotech) at 1 ⁇ g/ml
  • mouse anti-psoriasin Abeam
  • mouse anti-actin Sigma-Aldrich
  • the membrane was then washed before applying the secondary antibody (anti-mouse IgG peroxidase-linked species specific whole antibody from sheep, GE Healthcare) according to manufacturer's instructions for one hour at room temperature. Finally, the membrane was washed and visualized using ECL PlusTM detection kit (GE Healthcare). The signal intensity of the bands was measured using Image J software (NIH, Bethesda, Md., USA). The intensity of a band representing the protein of interest was divided by the intensity of it corresponding (3-actin expression on the same blot for normalization.
  • the protein expression pattern of carbonic anhydrase VI and psoriasin was further tested by Western blot with a new subject sample set including 31 cancer subject samples and 62 control subject samples. All the samples were coded with a random number from 1 to 93 and used for blind testing by Western blot.
  • a patient undergoing routine dental care is screened during the visit. For example, a 62 year old female patient, and former smoker, prior to oral exam is asked to provide a saliva sample.
  • a saliva sample is collected and analyzed either at the point of care or is submitted for analysis by a reference laboratory. The saliva sample is tested for the biomarkers of the instant invention and optionally other biomarkers. Results from the analysis are provided to the dental professional and the patient is informed as to whether she has breast cancer.

Abstract

Presented herein are biomarkers related to breast cancer. The presently identified salivary biomarkers create the basis for a breast cancer detection bioassay with sensitivity and specificity. Means and methods for evaluating the data generated using multiple biomarkers in order to validate findings and further use of the multiplexed breast cancer assay in clinical, diagnostic and therapeutic uses is also included.

Description

    CROSS-REFERENCES TO RELATED APPLICATIONS
  • The present application claims priority to provisional application U.S. Ser. No. 61/303,200, filed Feb. 10, 2010, herein incorporated by reference in its entirety.
  • STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • This invention was made with Government support under Grant No. DE016275, awarded by the National Institutes of Health. The Government has certain rights in this invention.
  • BACKGROUND
  • Breast cancer is the most frequent neoplasm and the leading cause of cancer mortality in women worldwide. According to estimates, approximately 41,000 women in the United States and 130,000 women in the European Union die from breast cancer yearly.
  • Detection of breast cancer at the earliest stages results in a much greater favorable outcome, with 10-year disease-free survival rate as high as 98% in patients in which the tumor stage is pT1a,bN0M0 (measuring 1 cm or less, with disease-free axillary lymph nodes and no distant metastasis). Needless to say, early detection is of paramount importance in reducing mortality from this major public health burden.
  • Current breast cancer detection methods are based on physical examination and imaging (for example, mammography, ultrasound, and MRI). These methods can produce a substantial percentage of false positive and false negative results especially in women with dense parenchymal breast tissue. Consequently, screening results in a number of negative biopsy results yielding a high percentage of false positives. There is also a demonstrated lack of sensitivity in detecting cancerous lesions in younger women yielding a significant percentage of false negatives. Accordingly, a clear need exists for added modalities of screening for breast cancer.
  • In the last decade, biomarker discoveries for breast cancer detection have focused on blood/or tissue, using proteomic, transcriptomic, and genomic approaches. In comparison to prognostic biomarkers, the development of detection biomarkers has been limited, mainly due to a lack of sensitivity and specificity for this clinical context. Most importantly, the use of tissue biomarkers for early detection will be limited to patients at very high risk because they rely on invasive procedures.
  • As such, a need exists for methods useful for detecting breast cancer, and in particular biomarkers that can detect early stages of the disease and are largely non-invasive.
  • BRIEF SUMMARY OF THE INVENTION
  • In accordance with some embodiments of the invention, a method of determining the likelihood of the presence or occurrence of breast cancer in a test subject is provided. The disclosed method includes analyzing a saliva sample from the subject with an assay that specifically detects at least two biomarkers in the saliva sample. The biomarkers are selected from the group of: S100A8 (S100 calcium binding protein A8) (SEQ ID NO: 1), CSTA (cystatin A) (SEQ ID NO:2), GRM1 (glutamate receptor, metabotropic 1) (SEQ ID NO: 3), TPT1 (tumor protein, translationally-controlled 1) (SEQ ID NO:4), GRIK1 (glutamate receptor, ionotropic, kainate 1) (SEQ ID NO: 5), H6PD (hexose-6-phosphate dehydrogenase) (SEQ ID NO: 6), IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) (SEQ ID NO: 7), MDM4 (3T3 cell double minute 4) (SEQ ID NO: 8), and CA6 (carbonic anhydrase VI) (SEQ ID NO:8). The relative occurrence of at least two of these biomarkers is determined and compared to a control, thereby allowing the breast cancer status of the test subject to be determined.
  • In some embodiments, one of the biomarkers of the at least two biomarkers is cystatin A (CSTA). In other embodiments, two of the at least two biomarkers is CSTA and transformed 3T3 cell double minute 4 (MDM4). The relative occurrence of these biomarkers or these biomarkers and others in these instances is determined and compared to a control, for example, thereby allowing the breast cancer status of the test subject to be determined.
  • In some embodiments, the method of determining the likelihood of the presence or occurrence of breast cancer entails measuring at least three biomarkers. In some embodiments, two of the at least three biomarkers are CSTA and MDM4. The relative occurrence of these biomarkers or these biomarkers and others in these instances is determined and compared to a control, for example, thereby allowing the breast cancer status of the test subject to be determined.
  • In some embodiments, one of the biomarkers of the at least two biomarkers is anhydrase VI (CA6) polypeptide.
  • In other embodiments, the method of determining the likelihood of the presence or occurrence of breast cancer in a test subject includes an assay in which a nucleic acid encoding at least one biomarker is detected. The nucleic acid can be detected by, for example, mass spectroscopy, polymerase chain reaction (PCR), microarray hybridization, thermal sequencing, capillary array sequencing, or solid phase sequencing.
  • In other embodiments, the method of determining the likelihood of the presence or occurrence of breast cancer in a test subject includes an assay in which a polypeptide encoding at least one biomarker is detected. The polypeptide can be detected by, for example, enzyme-linked immunosorbent assay (ELISA), Western blot, flow cytometry, immunofluorescence, immunohistochemistry, or mass spectroscopy.
  • In accordance with other embodiments of the invention, a method for assessing the efficacy of a therapy is disclosed. This method includes analyzing a first saliva sample from the subject with an assay that specifically detects at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and CA6. This first analysis provides a first expression profile. A therapy is applied to a subject. An analysis of a second saliva sample from the subject is undertaken with an assay that specifically detects at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and CA6 thereby providing a second expression profile. The first and second expression profiles are compared in order to assess the efficacy of a therapy.
  • In another embodiment, a solid support is provided, wherein the solid support includes a capture binding probe selective for at least two biomarkers selected from the group of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4. In some embodiments, a first and a second solid support are provided, wherein the first solid support includes a capture binding probe selective for at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and wherein the second solid support includes a capture binding ligand for CA6.
  • In some embodiments, the capture binding ligand of the kit is an antibody. In another embodiment the kit provides one or more primers for the selective amplification of at least two biomarkers, wherein at least two of the biomarkers are selected from the group of: S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4. In some embodiments one or more of the primers possess a detectable label.
  • In accordance with some embodiments of the invention, a method of determining the likelihood of the presence or occurrence of breast cancer in a test subject is provided. The disclosed method includes analyzing a saliva sample from the subject with an assay that specifically detects at least nine biomarkers in the saliva sample. The biomarkers are selected from the group of: S100A8 (S100 calcium binding protein A8) (SEQ ID NO: 1), CSTA (cystatin A) (SEQ ID NO:2), GRM1 (glutamate receptor, metabotropic 1) (SEQ ID NO: 3), TPT1 (tumor protein, translationally-controlled 1) (SEQ ID NO:4), GRIK1 (glutamate receptor, ionotropic, kainate 1) (SEQ ID NO: 5), H6PD (hexose-6-phosphate dehydrogenase) (SEQ ID NO: 6), IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) (SEQ ID NO: 7), MDM4 (3T3 cell double minute 4) (SEQ ID NO: 8), and CA6 (carbonic anhydrase VI) (SEQ ID NO:8). The relative occurrence of at least nine biomarkers is determined and compared to a control, thereby allowing the breast cancer status of the test subject to be determined.
  • In any of the embodiments above, wherein a method for determining the likelihood of the presence or occurrence of breast cancer in a test subject, the number of biomarkers used can be 2, 3, 4, 5, 6, 7, 8, 9, or more.
  • These and other embodiments, features and potential advantages will become apparent with reference to the following description and drawings.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic of the study designed to identify and validate biomarkers associated with breast cancer.
  • FIG. 2 is a schematic representation of the protocol for saliva collection.
  • FIG. 3 represents the demographic information of all the subjects used.
  • FIG. 4 represents biomarkers for breast cancer detection and effect of confounding factors (sample set n=93). The Mann-Whitney rank sum test was used to determine marker validation. Possible confounding factors, including age, ethnicity, smoking status, menopausal status, and HRT treatment, were evaluated for the biomarkers by logistic regression model. Linear regression model was constructed for each marker and used the factors cancer/normal and one of the confounders. cv.err:cross validation error rate.
  • FIG. 5 demonstrates the sensitivity achieved using a combination of the identified biomarkers. (A) The shading of the contigency table boxes reflects the fraction of each samples type in each quandrant. “Cancer” and “Non’ headings indicate subjects with and withour cancer, respectively. SB+ and SB−, salivary biomarker test positive or negative; NPV, negative predictive value; PPV, positive predictive value; Sen, sensitivity; Spec, specificity. (B) Score plot of principle component analysis (PCA). Combining the nine biomarkers, the control subjects (light shaded) separate from breast cancer patients (dark shading) with cumulative proportions of 66.9% for PC1 and 21.6% for PC2.
  • FIG. 6 represents cross-disease comparisons of the salivary mRNA biomarkers. The identified mRNA biomarkers for breast cancer detection were checked against other microarray datasets. t-test p-values were calculated for the identified breast cancer genes to other microarray datasets to check for significant variation (*after Boneferonni correction, P<0.0006) between patients and controls in those diseases. Sample sizes were 10 versus 10 for oral cancer, 10 versus 10 for lung cancer, 12 versus 12 for pancreatic cancer, 11 versus 11 for ovarian cancer, 13 versus 13 for diabetes, 8 versus 10 for primary Sjögren's Syndrome, and 10 versus 10 for breast cancer.
  • DETAILED DESCRIPTION OF THE INVENTION Introduction
  • Early detection of breast cancer offers the promise of easier treatment (smaller surgeries, less radiation or chemotherapy) and improved survival. Conventional screening (physical examination and mammography) has a less-than desirable sensitivity and specificity. A sensitive assay to identify biomarkers using non-invasively collected specimens is therefore ideal for breast cancer detection.
  • While saliva is a source of easily accessible bodily fluids, there has been little effort to study its value in cancer diagnosis. Protein, as well as RNA, can be detected in saliva.
  • The present invention discloses the diagnostic/prognostic significance of nine salivary biomarkers S100A8 (SEQ ID NO: 1) (S100 calcium binding protein A8, also referred to as myloid-related protein 8 (MRP8) or S100A9 (MRP14)), CSTA (SEQ ID NO: 2)(cystatin A), GRM1 (SEQ ID NO: 3)(glutamate receptor, metabotropic 1), TPT1 (SEQ ID NO: 4)(tumor protein, translationally-controlled 1), GRIK1 (SEQ ID NO: 5) (glutamate receptor, ionotropic, kainate 1), H6PD (SEQ ID NO: 6)(hexose-6-phosphate dehydrogenase or glucose 1-dehydrogenase), IGF2BP1 (SEQ ID NO: 7)(insulin-like growth factor 2 mRNA binding protein 1), MDM4 (SEQ ID NO: 8)(Mdm4, transformed 3T3 cell double minute 4; HDMX; MDMX; MRP1; MGC132766; DKFZp781B1423), and CA6 (carbonic anhydrase VI) and combinations thereof, in breast cancer detection. Detection of these and other biomarkers in saliva are useful for diagnosis and prognosis of breast cancer.
  • Methods for detecting salivary biomarkers (proteins and nucleic acids) include techniques such as ELISA, PCR, for example, RT-PCR or mass spectroscopy, alone or in combination with other markers. Any specific probe can be used for detection, such as an antibody, a receptor, a ligand, RT-PCR etc. Mass spectroscopy can also be used for protein detection. Thus, the present invention can be used alone or as a complement to traditional antigen analysis to enhance the diagnosis of breast and other cancers.
  • Definitions
  • “S100A8,” “CSTA,” “GRM1,” “TPT1,” “GRIK1,” “H6PD,” “IGF2BP1,” “MDM4,” and “CA6” refer to nucleic acids, e.g., gene, pre-mRNA, mRNA, and polypeptides, polymorphic variants, alleles, mutants, and interspecies homologs that have an amino acid sequence that has greater than about 60% amino acid sequence identity, 65%, 70%, 75%, 80%, 85%, 90%, preferably 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or greater amino acid sequence identity, preferably over a region of over a region of at least about 25, 50, 100, 200, 500, 1000, or more amino acids, to a polypeptide encoded by a referenced nucleic acid or an amino acid sequence described herein. The nucleic acids and proteins of the invention include both naturally occurring or recombinant molecules. The nucleic acid or protein sequence is provided, for example, in SEQ ID NOs: 1-9.
  • “Cancer” refers to human cancers and carcinomas, sarcomas, adenocarcinomas, lymphomas, leukemias, etc., including solid and lymphoid cancers, kidney, breast, lung, kidney, bladder, colon, ovarian, prostate, pancreas, stomach, brain, head and neck, skin, uterine, testicular, esophagus, and liver cancer, including hepatocarcinoma, lymphoma, including non-Hodgkin's lymphomas (e.g., Burkitt's, Small Cell, and Large Cell lymphomas) and Hodgkin's lymphoma, leukemia, and multiple myeloma.
  • “Therapeutic treatment” and “cancer therapies” refers to chemotherapy, hormonal therapy, radiotherapy, and immunotherapy.
  • The terms “overexpress,” “overexpression” or “overexpressed” interchangeably refer to a protein that is transcribed or translated at a detectably greater level, usually in a cancer cell, in comparison to a normal cell. The term includes overexpression due to transcription, post transcriptional processing, translation, post-translational processing, cellular localization (e.g, organelle, cytoplasm, nucleus, cell surface), and RNA and protein stability, as compared to a normal cell. Overexpression can be detected using conventional techniques for detecting mRNA (i.e., RT-PCR, PCR, hybridization) or proteins (i.e., ELISA, immunohistochemical techniques, mass spectroscopy). Overexpression can be 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a normal cell. In certain instances, overexpression is 1-fold, 2-fold, 3-fold, 4-fold or more higher levels of transcription or translation in comparison to a normal cell.
  • The terms “cancer-associated antigen” or “tumor-specific marker” or “tumor marker” interchangeably refers to a molecule (typically protein or nucleic acid such as RNA) that is expressed in the cell, expressed on the surface of a cancer cell or secreted by a cancer cell in comparison to a normal cell, and which is useful for the diagnosis of cancer, for providing a prognosis, and for preferential targeting of a pharmacological agent to the cancer cell. Oftentimes, a cancer-associated antigen is overexpressed in a cancer cell in comparison to a normal cell, for instance, about 1.2-fold over expression, about 2-fold overexpression, about 3-fold overexpression or more in comparison to a normal cell. Oftentimes, a cancer-associated antigen is a cell surface molecule that is inappropriately synthesized in the cancer cell, for instance, a molecule that contains deletions, additions or mutations in comparison to the molecule expressed on a normal cell. Oftentimes, a cancer-associated antigen will be expressed exclusively on the cell surface of a cancer cell and not synthesized or expressed on the surface of a normal cell. Exemplified cell surface tumor markers include the proteins c-erbB-2 and human epidermal growth factor receptor (HER) for breast cancer, PSMA for prostate cancer, and carbohydrate mucins in numerous cancers, including breast, ovarian and colorectal.
  • It will be understood by the skilled artisan that markers may be used singly or in combination with other markers for any of the uses, e.g., diagnosis or prognosis of breast cancer, disclosed herein.
  • The terms “identical” or percent “identity,” in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e., about 60% identity, preferably 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or higher identity over a specified region, when compared and aligned for maximum correspondence over a comparison window or designated region) as measured using a BLAST or BLAST 2.0 sequence comparison algorithms with default parameters described below, or by manual alignment and visual inspection (see, e.g., NCBI web site hypertext transfer protocol://www.ncbi.nlm.nih.gov/BLAST/ or the like). Such sequences are then said to be “substantially identical.” This definition also refers to, or may be applied to, the compliment of a test sequence. The definition also includes sequences that have deletions and/or additions, as well as those that have substitutions. As described below, the preferred algorithms can account for gaps and the like. Preferably, identity exists over a region that is at least about 25 amino acids or nucleotides in length, or more preferably over a region that is 50-100 amino acids or nucleotides in length.
  • For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Preferably, default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • A “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well-known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Nat'l. Acad. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by manual alignment and visual inspection (see, e.g., Current Protocols in Molecular Biology (Ausubel et al., eds. 1987-2005, Wiley Interscience)).
  • An example of algorithm that is suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al., Nuc. Acids Res. 25:3389-3402 (1977) and Altschul et al., J. Mol. Biol. 215:403-410 (1990), respectively. BLAST and BLAST 2.0 are used, with the parameters described herein, to determine percent sequence identity for the nucleic acids and proteins of the invention. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (hypertext transfer protocol://www.ncbi.nlm.nih gov/). This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, an expectation (E) of 10, M=5, N=−4 and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff & Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1989)) alignments (B) of 50, expectation (E) of 10, M=5, N=−4, and a comparison of both strands.
  • “Nucleic acid” refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double-stranded form, and complements thereof.
  • Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (for example, degenerate codon substitutions) and complementary sequences, as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)). The term nucleic acid is used interchangeably with gene, cDNA, mRNA, oligonucleotide, and polynucleotide.
  • A particular nucleic acid sequence also implicitly encompasses “splice variants” and nucleic acid sequences encoding truncated forms of cancer antigens. Similarly, a particular protein encoded by a nucleic acid implicitly encompasses any protein encoded by a splice variant or truncated form of that nucleic acid. “Splice variants,” as the name suggests, are products of alternative splicing of a gene. After transcription, an initial nucleic acid transcript may be spliced such that different (alternate) nucleic acid splice products encode different polypeptides. Mechanisms for the production of splice variants vary, but include alternate splicing of exons. Alternate polypeptides derived from the same nucleic acid by read-through transcription are also encompassed by this definition. Any products of a splicing reaction, including recombinant forms of the splice products, are included in this definition. Nucleic acids can be truncated at the 5′ end or at the 3′ end. Polypeptides can be truncated at the N-terminal end or the C-terminal end. Truncated versions of nucleic acid or polypeptide sequences can be naturally occurring or recombinantly created.
  • The term “amino acid” refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, .gamma.-carboxyglutamate, and O-phosphoserine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an .alpha. carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • “Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are “silent variations,” which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid. One of skill will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid which encodes a polypeptide is implicit in each described sequence with respect to the expression product, but not with respect to actual probe sequences.
  • As to amino acid sequences, one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
  • The following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
  • A “label” or a “detectable moiety” is a composition detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means. For example, useful labels include fluorescent dyes, electron-dense reagents, enzymes (for example, as commonly used in an ELISA), biotin, digoxigenin, or haptens and proteins which can be made detectable, e.g., by incorporating a radiolabel into the peptide or used to detect antibodies specifically reactive with the peptide.
  • The tem) “recombinant” when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
  • The phrase “stringent hybridization conditions” refers to conditions under which a probe will hybridize to its target subsequence, typically in a complex mixture of nucleic acids, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Probes, “Overview of principles of hybridization and the strategy of nucleic acid assays” (1993). Generally, stringent conditions are selected to be about 5-10° C. lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength pH. The Tm is the temperature (under defined ionic strength, pH, and nucleic concentration) at which 50% of the probes complementary to the target hybridize to the target sequence at equilibrium (as the target sequences are present in excess, at Tm, 50% of the probes are occupied at equilibrium). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. For selective or specific hybridization, a positive signal is at least two times background, preferably 10 times background hybridization. Exemplary stringent hybridization conditions can be as following: 50% formamide, 5×SSC, and 1% SDS, incubating at 42° C., or, 5×SSC, 1% SDS, incubating at 65° C., with wash in 0.2×SSC, and 0.1% SDS at 65° C.
  • Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides which they encode are substantially identical. This occurs, for example, when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code. In such cases, the nucleic acids typically hybridize under moderately stringent hybridization conditions. Exemplary “moderately stringent hybridization conditions” include a hybridization in a buffer of 40% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 1×SSC at 45° C. A positive hybridization is at least twice background. Those of ordinary skill will readily recognize that alternative hybridization and wash conditions can be utilized to provide conditions of similar stringency. Additional guidelines for determining hybridization parameters are provided in numerous reference, e.g., and Current Protocols in Molecular Biology, ed. Ausubel, et al., supra.
  • For PCR, a temperature of about 36° C. is typical for low stringency amplification, although annealing temperatures may vary between about 32° C. and 48° C. depending on primer length. For high stringency PCR amplification, a temperature of about 62° C. is typical, although high stringency annealing temperatures can range from about 50° C. to about 65° C., depending on the primer length and specificity. Typical cycle conditions for both high and low stringency amplifications include a denaturation phase of 90° C.−95° C. for 30 sec-2 min., an annealing phase lasting 30 sec.-2 min., and an extension phase of about 72° C. for 1-2 min. Protocols and guidelines for low and high stringency amplification reactions are provided, e.g., in Innis et al. (1990) PCR Protocols, A Guide to Methods and Applications, Academic Press, Inc. N.Y.).
  • “Antibody” means a protein comprising one or more polypeptides substantially encoded by all or part of the recognized immunoglobulin genes. The recognized immunoglobulin genes, for example in humans, include the kappa (κ), lambda (λ) and heavy chain genetic loci, which together compose the myriad variable region genes, and the constant region genes mu (μ), delta (δ), gamma (γ), epsilon (ε) and alpha (α), which encode the IgM, IgD, IgG, IgE, and IgA isotypes respectively. Antibody herein is meant to include full length antibodies and antibody fragments, and may refer to a natural antibody from any organism, an engineered antibody or an antibody generated recombinantly for experimental, therapeutic or other purposes as further defined below. Antibody fragments include Fab, Fab′, F(ab′)2, Fv, scFv or other antigen-binding subsequences of antibodies and can include those produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA technologies. The term “antibody” refers to both monoclonal and polyclonal antibodies. Antibodies can be antagonists, agonists, neutralizing, inhibitory or stimulatory.
  • Biomarkers
  • Biomarkers may originate from epidemiological studies, animal studies, pathophysiological considerations and end-organ experiments. Ideally, a biomarker will have a high predictive value for a meaningful outcome measure, can be or is validated in appropriately designed prospective trials, reflects therapeutic success by corresponding changes in the surrogate marker results, and should be easy to assess in clinical practice.
  • Biomarkers can be used in conjunction with other diagnostic tools or used alone.
  • The term “surrogate marker,” “biomolecular marker,” “biomarker” or “marker” (also sometimes referred to herein as a “target analyte,” “target species” or “target sequence”) refers to a molecule whose measurement provides information as to the state of a subject. In various exemplary embodiments, the biomarker is used to assess a pathological state. Measurements of the biomarker may be used alone or combined with other data obtained regarding a subject in order to determine the state of the subject. In one embodiment, the biomarker is “differentially present” in a sample taken from a subject of one phenotypic status (e.g., having a disease) as compared with another phenotypic status (e.g., not having the disease). In one embodiment, the biomarker is “differentially present” in a sample taken from a subject undergoing no therapy or one type of therapy as compared with another type of therapy. Alternatively, the biomarker may be “differentially present” even if there is no phenotypic difference, e.g. the biomarkers may allow the detection of asymptomatic risk.
  • A biomarker may be over-expressed (over-abundant) or under-expressed (under abundant) relative to a control. The biomarker can be an allelic variant, truncated or mutated form of a wild-type nucleic acid or protein. The biomarker can be a splice variant.
  • A biomarker may be determined to be “differentially present” in a variety of ways, for example, between different phenotypic statuses if the mean or median level (particularly the expression level of the associated mRNAs as described below) of the biomarker in the different groups is calculated to be statistically significant. Common tests for statistical significance include, among others, t-test, ANOVA, Kruskal-Wallis, Wilcoxon, Mann-Whitney and odds ratio.
  • As described herein, a biomarker may be, for example, a small molecule, an analyte or target analyte, a nucleic acid, a protein, a metabolite or any derivative thereof or any and all combinations of these molecules, with proteins and nucleic acids finding particular use in the invention. As will be appreciated by those in the art, a large number of analytes may be detected using the present methods; basically, any biomarker for which a binding ligand, described below, may be made may be detected using the methods of the invention.
  • In various embodiments, the biomarkers used in the panels of the invention can be detected either as proteins or as nucleic acids (e.g. mRNA or cDNA transcripts) in any combination. In various embodiments, the protein form of a biomarker is measured. As will be appreciated by those in the art, protein assays may be done using standard techniques such as ELISA assays. In various embodiments, the nucleic acid form of a biomarker (e.g., the corresponding mRNA) is measured. In various exemplary embodiments, one or more biomarkers from a particular panel are measured using a protein assay and one or more biomarkers from the same panel are measured using a nucleic acid assay.
  • As will be appreciated by those in the art, there are a large number of possible proteinaceous target analytes and target species that may be detected using the present invention. The term “protein,” “polypeptide” or “oligopeptide” refers to at least two or more peptides or amino acids joined by one or more peptide bonds. A protein or an amino acid may be naturally or nonnaturally occurring and may be also be an analog, a derivative or a peptidomimetic structure. The term “protein” refers to wild-type sequences, variants of wild-type sequences and either of these containing analogs or derivatized amino acids. In various embodiments, variants of the sequences described herein, including proteins and nucleic acids based on e.g. splice variants, variants comprising a deletion, addition, substitution, fragments, preproprotein, processed preproprotein (e.g. without a signaling peptide), processed proprotein (e.g. resulting in an active form), nonhuman sequences and variant nonhuman sequences may be used as biomarkers.
  • In various embodiments, the biomarker is a nucleic acid. The term “nucleic acid” or “oligonucleotide” or grammatical equivalents herein means at least two nucleotides covalently linked together. A nucleic acid of the present invention will generally contain phosphodiester bonds, although in some cases, as outlined below, for example in the use of binding ligand probes, nucleic acid analogs are included that may have alternate backbones.
  • Biomarkers can also be bacterial nucleic acids or proteins. Over 700 species of bacteria have been identified to exist within the mouth. The presence, absence, or level of 16S rRNA from bacteria in a sample may correlate with a disease or condition. “Bacteria” refers to small prokaryotic organisms (linear dimensions of around 1 μm) with non-compartmentalized circular DNA and ribosomes of about 70 S. “16S RNA” refers to a nucleic acid component of the 30S subunit of prokaryotic ribosomes; the gene that encodes the 16S rRNA or the 16S rRNA itself. Bacterial strains of species or phylotypes have less than about a 2% difference in 16S rRNA. Closely related species or phylotypes generally have between about a 2% and about a 4% difference in 16S rRNA, whereas a genus often has between about a 5% and about a 10% difference in 16S rRNA.
  • To resolve the identity of bacterial populations, probes on a microarray can be designed, for example, to take advantage of conserved features of the 16S rRNA gene. For example, probes complementary to the more conserved features regions identify species in a large phylogenetic group, each group corresponding to a higher taxon (for example, domain, phylum, class, order, or family). Probes complementary to more variable regions distinguish genera and species.
  • Biomarkers can also include micro RNAs. “MicroRNAs” (miRs) refers to a class of small naturally occurring non-coding RNAs (18-24 nucleotides) that regulate gene expression. Many microRNAs are well conserved across species and they are present in a broad range of species: plants, nematodes, fruit flies and humans. MicroRNAs have partially or perfect complementary sequence to one or more messenger RNA molecules (mRNAs) and their main function is to negatively regulate the expression of genes. In particular, microRNAs bind to the 3′ untranslated regions of mRNAs (3-UTR) thus leading to down regulation of mRNAs in a variety of ways such as mRNA cleavage, translational repression and deadenylation.
  • A variety of experimental approaches and different techniques have been used to identify new microRNAs, as well as to study their expression pattern in the different biological processes. The cloning and identification of new microRNAs have been successfully done from size fractioned RNA samples using small RNA cloning approaches. Other approaches is as putative microRNAs homologues to microRNAs that already have been described in other species or using computational approaches alone or in combination with microarray analysis and sequence-directed cloning.
  • One of the first techniques used for detection and profiling of microRNAs was Northern Blotting, where hybridization is done with a complementary 32P, digoxigenin-labeled oligo or modified Locked-nucleic-acid (LNA) oligonucleotides after gel separation.
  • Other techniques that have been developed to specifically detect microRNAs are a modified invader assay (a synthetic oligonucleotide, the probe, which is in an appropriate overlap-flap structure is enzymatically cleavage by a structure-specific 5* nuclease) and in situ hybridization (using fluorescent-labeled complementary probes containing chemically modified nucleotides e.g. LNAs). Another widely used technique for detection and profiling of microRNAs is the use of oligonucleotide micro-array based detection platforms either with DNA capture probes or using modified Locked-nucleic-acid (LNA) oligonucleotides in which the ribose moiety is modified with an extra bridge that connects the 2′-0 and 4′-C atoms.
  • In addition, quantitative real-time PCR (reverse transcriptase/polymerase chain reaction using Taqman or SYBR green technology) has been used for detection and profiling of precursor or mature microRNAs. This technique is sensitive and requires low amounts of starting material for the detection of individual mature microRNAs. Taqman microRNA arrays have been developed that provide the sensitivity of the qRT-PCR, while at the same time enables the simultaneously detection of different microRNAs in one sample.
  • Biomarkers can also include metabolites. “Metabolite” or “small molecule” refers to organic and inorganic molecules which are present in a sample. The term does not include large macromolecules, such as large proteins (e.g., proteins with molecular weights over 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000), large nucleic acids (e.g., nucleic acids with molecular weights of over 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000), or large polysaccharides (e.g., polysaccharides with a molecular weights of over 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, or 10,000).
  • The metabolites of the cell are generally found free in solution. A “metabolic profile”, or “small molecule profile”, means a complete or partial inventory of small molecules within a targeted cell, tissue, organ, organism, or fraction thereof (e.g., cellular compartment). The inventory may include the quantity and/or type of small molecules present. The “small molecule profile” may be determined using a single technique or multiple different techniques.
  • A metabolic profile can be developed by analyzing a sample using for example, techniques such as GC-MS (gas chromatography-mass spectrometry) and LC-MS (liquid chromatography-mass spectrometry).
  • Biomarker Panels
  • Any combination of the biomarkers described herein is used to assemble a biomarker panel, which is detected or measured as described herein. As is generally understood in the art, a combination may refer to an entire set or any subset or subcombination thereof. The term “biomarker panel,” “biomarker profile,” or “biomarker fingerprint” refers to a set of biomarkers. As used herein, these terms can also refer to any form of the biomarker that is measured. Thus, if cystatin A is part of a biomarker panel, then either cystatin A mRNA, for example, or protein could be considered to be part of the panel. While individual biomarkers are useful as diagnostics, combination of biomarkers can sometimes provide greater value in determining a particular status than single biomarkers alone. Specifically, the detection of a plurality of biomarkers in a sample can increase the sensitivity and/or specificity of the test. Thus, in various embodiments, a biomarker panel may include 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more types of biomarkers. In various exemplary embodiments, the biomarker panel consists of a minimum number of biomarkers to generate a maximum amount of information. Thus, in various embodiments, the biomarker panel consists of 2, 3, 4, 5, 6, 7, 8, 9 or more types of biomarkers. Where a biomarker panel “consists of” a set of biomarkers, no biomarkers other than those of the set are present. In exemplary embodiments, the biomarker panel consists of 2 biomarkers disclosed herein. In various embodiments, the biomarker panel consists of 3 biomarkers disclosed herein. In various embodiments, the biomarker panel consists of 4 biomarkers disclosed herein. In various embodiments, the biomarker paenl consists of 5 biomarkers disclosed herein.
  • In various exemplary embodiments, the biomarker panel comprises cystatin A. In various exemplary embodiments, the biomarker panel comprises carbonic anhydrase VI.
  • In various exemplary embodiments, the biomarker panel comprises or consists of two or more of the biomarkers selected from the group of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and CA6. In various exemplary embodiments two or more of the biomarkers selected from the group of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and CA6 can be combined with 1, 2, 3, 4 or more additional biomarkers. It should be understood that in this embodiment, the biomarker panel can include any combination of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4 and the remainder of these markers.
  • A biomarker can also be a clinical parameter. The term “clinical parameter” refers to all non-sample or non-analyte biomarkers of subject health status or other characteristics, such as, without limitation, age, ethnicity, gender, family history, height, and weight.
  • The biomarkers of the invention show a statistically significant difference in breast cancer diagnosis. In various embodiments, diagnostic tests that use these biomarkers alone or in combination show a sensitivity and specificity of at least about 85%, at least about 90%, at least about 95%, at least about 98% and about 100%.
  • Measurement and Detection of Biomarkers
  • Biomarkers generally can be measured and detected through a variety of assays, methods and detection systems known to one of skill in the art. The term “measuring,” “detecting,” or “taking a measurement” refers to a quantitative or qualitative determination of a property of an entity, for example, quantifying the amount or concentration of a molecule or the activity level of a molecule. The term “concentration” or “level” can refer to an absolute or relative quantity. Measuring a molecule may also include determining the absence or presence of the molecule. Various methods include but are not limited to refractive index spectroscopy (RI), ultra-violet spectroscopy (UV), fluorescence analysis, electrochemical analysis, radiochemical analysis, near-infrared spectroscopy (near-IR), infrared (IR) spectroscopy, nuclear magnetic resonance spectroscopy (NMR), light scattering analysis (LS), mass spectrometry, pyrolysis mass spectrometry, nephelometry, dispersive Raman spectroscopy, gas chromatography, liquid chromatography, gas chromatography combined with mass spectrometry, liquid chromatography combined with mass spectrometry, matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) combined with mass spectrometry, ion spray spectroscopy combined with mass spectrometry, capillary electrophoresis, colorimetry and surface plasmon resonance (such as according to systems provided by Biacore Life Sciences). See also PCT Publications WO/2004/056456 and WO/2004/088309. In this regard, biomarkers can be measured using the above-mentioned detection methods, or other methods known to the skilled artisan. Other biomarkers can be similarly detected using reagents that are specifically designed or tailored to detect them.
  • Different types of biomarkers and their measurements can be combined in the compositions and methods of the present invention. In various embodiments, the protein form of the biomarkers is measured. In various embodiments, the nucleic acid form of the biomarkers is measured. In exemplary embodiments, the nucleic acid form is mRNA. In various embodiments, measurements of protein biomarkers are used in conjunction with measurements of nucleic acid biomarkers.
  • Methods for detecting mRNA, such as RT-PCR, real time PCR, branch DNA, NASBA and others, are well known in the art. Using sequence information provided by the database entries for the biomarker sequences, expression of the biomarker sequences can be detected (if present) and measured using techniques well known to one of ordinary skill in the art. For example, sequences in sequence database entries or sequences disclosed herein can be used to construct probes for detecting biomarker RNA sequences in, e.g., Northern blot hybridization analyses or methods which specifically, and, preferably, quantitatively amplify specific nucleic acid sequences. As another example, the sequences can be used to construct primers for specifically amplifying the biomarker sequences in, e.g., amplification-based detection methods such as reverse-transcription based polymerase chain reaction (RT-PCR). When alterations in gene expression are associated with gene amplification, deletion, polymorphisms and mutations, sequence comparisons in test and reference populations can be made by comparing relative amounts of the examined DNA sequences in the test and reference cell populations. In addition to Northern blot and RT-PCR, RNA can also be measured using, for example, other target amplification methods (e.g., TMA, SDA, NASBA), signal amplification methods (e.g., bDNA), nuclease protection assays, in situ hybridization and the like.
  • In one embodiment in the present invention are biochip assays. By “biochip” or “chip” herein is meant a composition generally comprising a solid support or substrate to which a capture binding ligand (also called an adsorbent, affinity reagent or binding ligand, or when nucleic acid is measured, a capture probe) is attached and can bind either proteins, nucleic acids or both. Generally, where a biochip is used for measurements of protein and nucleic acid biomarkers, the protein biomarkers are measured on a chip separate from that used to measure the nucleic acid biomarkers. For nonlimiting examples of additional platforms and methods useful for measuring nucleic acids, see Publications US/2006/0275782, US/2005/0064469 and DE10201463. In various embodiments, biomarkers are measured on the same platform, such as on one chip. In various embodiments, biomarkers are measured using different platforms and/or different experimental runs.
  • By “binding ligand,” “capture binding ligand,” “capture binding species,” “capture probe” or grammatical equivalents herein is meant a compound that is used to detect the presence of or to quantify, relatively or absolutely, a target analyte, target species or target sequence (all used interchangeably) and that will bind to the target analyte, target species or target sequence. Generally, the capture binding ligand or capture probe allows the attachment of a target species or target sequence to a solid support for the purposes of detection as further described herein. Attachment of the target species to the capture binding ligand may be direct or indirect. In exemplary embodiments, the target species is a biomarker. As will be appreciated by those in the art, the composition of the binding ligand will depend on the composition of the biomarker. Binding ligands for a wide variety of biomarkers are known or can be readily found using known techniques. For example, when the biomarker is a protein, the binding ligands include proteins (particularly including antibodies or fragments thereof (Fabs, etc.) as discussed further below) or small molecules. The binding ligand may also have cross-reactivity with proteins of other species. Antigen-antibody pairs, receptor-ligands, and carbohydrates and their binding partners are also suitable analyte-binding ligand pairs. In various embodiments, the binding ligand may be nucleic acid. Nucleic acid binding ligands find particular use when proteins are the targets; alternatively, as is generally described in U.S. Pat. Nos. 5,270,163; 5,475,096; 5,567,588; 5,595,877; 5,637,459; 5,683,867; 5,705,337 and related patents, hereby incorporated by reference, nucleic acid “aptamers” can be developed for binding to virtually any biomarker. Nucleic acid binding ligands also find particular use when nucleic acids are binding targets. There is a wide body of literature relating to the development of binding partners based on combinatorial chemistry methods. In these embodiments, when the binding ligand is a nucleic acid, preferred compositions and techniques are outlined in PCT Publication WO/1998/020162, hereby incorporated by reference.
  • In various exemplary embodiments, the capture binding ligand is an antibody. These embodiments are particularly useful for the detection of the protein form of a biomarker.
  • Detecting or measuring the level (e.g. the transcription level) of a biomarker involves binding of the biomarker to a capture binding ligand, generally referred to herein as a “capture probe” when the mRNA of the biomarker is to be detected on a solid support. In that sense, the biomarker is a target sequence. The term “target sequence” or “target nucleic acid” or grammatical equivalents herein means a nucleic acid sequence that may be a portion of a gene, a regulatory sequence, genomic DNA, cDNA, RNA including mRNA and rRNA, or others. As is outlined herein, the target sequence may be a target sequence from a sample, or a secondary target such as a product of an amplification reaction such as PCR etc. In some embodiments, measuring a nucleic acid can thus refer to measuring the complement of the nucleic acid. It may be any length, with the understanding that longer sequences are more specific.
  • The target sequence may also comprise different target domains; for example, a first target domain of the sample target sequence may hybridize to a first capture probe, a second target domain may hybridize to a label probe (e.g. a “sandwich assay” format), etc. The target domains may be adjacent or separated as indicated. Unless specified, the terms “first” and “second” are not meant to confer an orientation of the sequences with respect to the 5′-3′ orientation of the target sequence. For example, assuming a 5′-3′ orientation of the target sequence, the first target domain may be located either 5′ to the second domain, or 3′ to the second domain.
  • When nucleic acids are used as the target analyte, the assays of the invention can take on a number of embodiments. In one embodiment, the assays are done in solution format, using any number of solution based formats. In one embodiment, end-point or real time PCR formats are used, as are well known in the art. These assays can be done either as a panel, in individual tubes or wells, or as multiplex assays, using sets of primers and different labels within a single tube or well. In addition to PCR-based solution formats, other formats can be utilized, including, but not limited to for example ligation based assays utilizing FRET dye pairs. In this embodiment, only upon ligation of two (or more) probes hybridized to the target sequence is a signal generated.
  • In many embodiments, the assays are done on a solid support, utilizing a capture probe associated with the surface. As discussed herein, the capture probes (or capture binding ligands, as they are sometimes referred to) can be covalently attached to the surface, for example using capture probes terminally modified with functional groups, for example amino groups, that are attached to modified surfaces such as silanized glass. Alternatively, non-covalent attachment, such as electrostatic, hydrophobic/hydrophilic adhesion can be utilized. As is appreciated by those in the art and discussed herein, a large number of attachments are possible on a wide variety of surfaces.
  • In this embodiment, the assays can take on a number of formats. In one embodiment, the target sequence comprises a detectable label, as described herein. In this embodiment, the label is generally added to the target sequence during amplification of the target in one of two ways: either labeled primers are utilized during the amplification step or labeled dNTPs are used, both of which are well known in the art. The label can either be a primary or secondary label as discussed herein. For example, in one embodiment, the label on the primer and/or a dNTP is a primary label such as a fluorophore. Alternatively, the label may be a secondary label such as biotin or an enzyme; for example, in one embodiment, the primers or dNTPs are labeled with biotin, and then a streptavidin/label complex is added. In one embodiment, the streptavidin/label complex contains a label such as a fluorophore. In an alternative embodiment, the streptavidin/label complex comprises an enzymatic label. For example, the complex can comprise horseradish peroxidase, and upon addition of TMB, the action of the horseradish peroxidase causes the TMB to precipitate, causing an optically detectable event. This has a particular benefit in that the optics for detection does not require the use of a fluorimeter.
  • In alternate embodiments, the solid phase assay relies on the use of a labeled soluble capture ligand, sometimes referred to as a “label probe” or “signaling probe” when the target analyte is a nucleic acid. In this format, the assay is a “sandwich” type assay, where the capture probe binds to a first domain of the target sequence and the label probe binds to a second domain. In this embodiment, the label probe can also be either a primary (e.g. a fluorophore) or a secondary (biotin or enzyme) label. In one embodiment, the label probe comprises biotin, and a streptavidin/enzyme complex is used, as discussed herein. As above, for example, the complex can comprise horseradish peroxidase, and upon addition of TMB, the action of the horseradish peroxidase causes the TMB to precipitate, causing an optically detectable event.
  • Detection of a target species in some embodiments requires a “label” or “detectable marker” (as described below) that can be incorporated in a variety of ways. Thus, in various embodiments, the composition comprises a “label” or a “detectable marker.” In one embodiment, the target species (or target analyte or target sequence) is labeled; binding of the target species thus provides the label at the surface of the solid support.
  • In embodiments finding particular use herein, a sandwich format is utilized, in which target species are unlabeled. In these embodiments, a “capture” or “anchor” binding ligand is attached to the detection surface as described herein, and a soluble binding ligand (frequently referred to herein as a “signaling probe,” “label probe” or “soluble capture ligand”) binds independently to the target species and either directly or indirectly comprises at least one label or detectable marker.
  • By “label” or “labeled” herein is meant that a compound has at least one molecule, element, isotope or chemical compound attached to enable the detection of the compound. In general, labels fall into four classes: a) isotopic labels, which may be radioactive or heavy isotopes; b) magnetic, electrical, thermal; c) colored or luminescent dyes; and d) enzymes; although labels include particles such as magnetic particles as well. The dyes may be chromophores or phosphors but are preferably fluorescent dyes, which due to their strong signals provide a good signal-to-noise ratio for decoding. Suitable dyes for use in the invention include, but are not limited to, fluorescent lanthanide complexes, including those of Europium and Terbium, fluorescein, rhodamine, tetramethylrhodamine, eosin, erythrosin, coumarin, methyl-coumarins, pyrene, Malacite green, stilbene, Lucifer Yellow, Cascade Blue, Texas Red, Alexa dyes and others described in the 6th Edition of the Molecular Probes Handbook by Richard P. Haugland, hereby expressly incorporated by reference. Additional labels include nanocrystals or Q-dots as described in U.S. Pat. No. 6,544,732 incorporated by reference.
  • In various embodiments, a secondary detectable label is used. A secondary label is one that is indirectly detected; for example, a secondary label can bind or react with a primary label for detection, can act on an additional product to generate a primary label (e.g. enzymes), or may allow the separation of the compound comprising the secondary label from unlabeled materials, etc. Secondary labels include, but are not limited to, one of a binding partner pair; chemically modifiable moieties; nuclease inhibitors, enzymes such as horseradish peroxidase, alkaline phosphatases, lucifierases, etc. Secondary labels can also include additional labels.
  • In various embodiments, the secondary label is a binding partner pair. For example, the label may be a hapten or antigen, which will bind its binding partner. For example, suitable binding partner pairs include, but are not limited to: antigens (such as proteins (including peptides)) and antibodies (including fragments thereof (Fabs, etc.)); proteins and small molecules, including biotin/streptavidin; enzymes and substrates or inhibitors; other protein-protein interacting pairs; receptor-ligands; and carbohydrates and their binding partners. Nucleic acid-nucleic acid binding proteins pairs are also useful. In general, the smaller of the pair is attached to the NTP for incorporation into the primer. Preferred binding partner pairs include, but are not limited to, biotin (or imino-biotin) and streptavidin, digeoxinin and Abs, and Prolinx™ reagents.
  • In the sandwich formats of the invention, an enzyme serves as the secondary label, bound to the soluble capture ligand. Of particular use in some embodiments is the use of horseradish peroxidase, which when combined with 3,3′,5,5′-tetramethylbenzidine (TMB) forms a colored precipitate which is then detected. In some cases, the soluble capture ligand comprises biotin, which is then bound to a enzyme-streptavidin complex and forms a colored precipitate with the addition of TMB.
  • In various embodiments, the label or detectable marker is a conjugated enzyme (for example, horseradish peroxidase). In various embodiments, the system relies on detecting the precipitation of a reaction product or on a change in, for example, electronic properties for detection. In various embodiments, none of the compounds comprises a label.
  • As used herein, the term “fluorescent signal generating moiety” or “fluorophore” refers to a molecule or part of a molecule that absorbs energy at one wavelength and re-emits energy at another wavelength. Fluorescent properties that can be measured include fluorescence intensity, fluorescence lifetime, emission spectrum characteristics, energy transfer, and the like.
  • Signals from single molecules can be generated and detected by a number of detection systems, including, but not limited to, scanning electron microscopy, near field scanning optical microscopy (NSOM), total internal reflection fluorescence microscopy (TIRFM), and the like. Abundant guidance is found in the literature for applying such techniques for analyzing and detecting nanoscale structures on surfaces, as evidenced by the following references that are incorporated by reference: Reimer et al, editors, Scanning Electron Microscopy: Physics of Image Formation and Microanalysis, 2nd Edition (Springer, 1998); Nie et al, Anal. Chem., 78: 1528-1534 (2006); Hecht et al, Journal Chemical Physics, 112: 7761-7774 (2000); Zhu et al, editors, Near-Field Optics: Principles and Applications (World Scientific Publishing, Singapore, 1999); Drmanac, PCT Publication WO/2004/076683; Lehr et al, Anal. Chem., 75: 2414-2420 (2003); Neuschafer et al, Biosensors & Bioelectronics, 18: 489-497 (2003); Neuschafer et al, U.S. Pat. No. 6,289,144; and the like.
  • Thus, a detection system for fluorophores includes any device that can be used to measure fluorescent properties as discussed above. In various embodiments, the detection system comprises an excitation source, a fluorophore, a wavelength filter to isolate emission photons from excitation photons and a detector that registers emission photons and produces a recordable output, in some embodiments as an electrical signal or a photographic image. Examples of detection devices include without limitation spectrofluorometers and microplate readers, fluorescence microscopes, fluorescence scanners (including e.g. microarray readers) and flow cytometers.
  • In various exemplary embodiments, the binding of the biomarker to the binding ligand is specific or selective, and the binding ligand is part of a binding pair. By “specifically bind” or “selectively bind” or “selective for” a biomarker herein is meant that the ligand binds the biomarker with specificity sufficient to differentiate between the biomarker and other components or contaminants of the test sample.
  • The term “solid support” or “substrate” refers to any material that can be modified to contain discrete individual sites appropriate for the attachment or association of a capture binding ligand. Suitable substrates include metal surfaces such as gold, electrodes, glass and modified or functionalized glass, plastics (including acrylics, polystyrene and copolymers of styrene and other materials, polypropylene, polyethylene, polybutylene, polycarbonate, polyurethanes, Teflon, derivatives thereof, etc.), polysaccharides, nylon or nitrocellulose, resins, mica, silica or silica-based materials including silicon and modified silicon, carbon, metals, inorganic glasses, fiberglass, ceramics, GETEK (a blend of polypropylene oxide and fiberglass) and a variety of other polymers. Of particular use in the present invention are the ClonDiag materials described below.
  • Frequently, the surface of a biochip comprises a plurality of addressable locations, each of which comprises a capture binding ligand. An “array location,” “addressable location,” “pad” or “site” herein means a location on the substrate that comprises a covalently attached capture binding ligand. An “array” herein means a plurality of capture binding ligands in a regular, ordered format, such as a matrix. The size of the array will depend on the composition and end use of the array. Arrays containing from about two or more different capture binding ligands to many thousands can be made. Generally, the array will comprise 3, 4, 5, 6, 7 or more types of capture binding ligands depending on the end use of the array. In the present invention, the array can include controls, replicates of the markers and the like. Exemplary ranges are from about 3 to about 50. In some embodiments, the compositions of the invention may not be in array format; that is, for some embodiments, compositions comprising a single capture ligand may be made as well. In addition, in some arrays, multiple substrates may be used, either of different or identical compositions. Thus for example, large arrays may comprise a plurality of smaller substrates.
  • Accordingly, in one aspect, the invention provides a composition comprising a solid support comprising a capture binding ligand for each biomarker of a biomarker panel. In various embodiments, the capture ligand is a nucleic acid. In various embodiments, the capture binding ligand is an antibody. In various embodiments, the composition further comprises a soluble binding ligand for each biomarker of a biomarker panel.
  • A number of different biochip array platforms as known in the art may be used. For example, the compositions and methods of the present invention can be implemented with array platforms such as GeneChip® (Affymetrix), CodeLink™ Bioarray (Amersham), Expression Array System (Applied Biosystems), SurePrint microarrays (Agilent), Sentrix® LD BeadChip or Sentrix® Array Matrix (Illumina) and Verigene (Nanosphere).
  • In various exemplary embodiments, detection and measurement of biomarkers utilizes colorimetric methods and systems in order to provide an indication of binding of a target analyte or target species. In colorimetric methods, the presence of a bound target species such as a biomarker will result in a change in the absorbance or transmission of light by a sample or substrate at one or more wavelengths. Detection of the absorbance or transmission of light at such wavelengths thus provides an indication of the presence of the target species.
  • A detection system for colorimetric methods includes any device that can be used to measure colorimetric properties as discussed above. Generally, the device is a spectrophotometer, a colorimeter or any device that measures absorbance or transmission of light at one or more wavelengths. In various embodiments, the detection system comprises a light source; a wavelength filter or monochromator; a sample container such as a cuvette or a reaction vial; a detector, such as a photoresistor, that registers transmitted light; and a display or imaging element.
  • In various exemplary embodiments, a ClonDiag chip platform is used for the colorimetric detection of biomarkers. In various embodiments, a ClonDiag ArrayTube (AT) is used. One unique feature of the ArrayTube is the combination of a micro probe array (the biochip) and micro reaction vial. In various embodiments, where a target sequence is a nucleic acid, detection of the target sequence is done by amplifying and biotinylating the target sequence contained in a sample and optionally digesting the amplification products. The amplification product is then allowed to hybridize with probes contained on the ClonDiag chip. A solution of a streptavidin-enzyme conjugate, such as Poly horseradish peroxidase (HRP) conjugate solution, is contacted with the ClonDiag chip. After washing, a dye solution such as o-dianisidine substrate solution is contacted with the chip. Oxidation of the dye results in precipitation that can be detected colorimetrically. Further description of the ClonDiag platform is found in Monecke S, Slickers P, Hotzel H et al., Clin Microbiol Infect 2006, 12: 718-728; Monecke S, Berger-Bächi B, Coombs C et al., Clin Microbiol Infect 2007, 13: 236-249; Monecke S, Leube I and Ehricht R, Genome Lett 2003, 2: 106-118; Monecke S and Ehricht R, Clin Microbiol Infect 2005, 11: 825-833; German Patent DE 10201463; US Publication US/2005/0064469 and ClonDiag, ArrayTube (AT) Experiment Guideline for DNA-Based Applications, version 1.2, 2007, all incorporated by reference in their entirety. One of skill in the art will appreciate that numerous other dyes that react with a peroxidase can be utilized to produce a colorimetric change, such as 3,3′,5,5′-tetramethylbenzidine (TMB). For information on specific assay protocols, see www.clondiag.com/technologies/publications.php.
  • In various embodiments, where a target species is a protein, the ArrayTube biochip comprises capture binding ligands such as antibodies. A sample is contacted with the biochip, and any target species present in the sample is allowed to bind to the capture binding ligand antibodies. A soluble capture binding ligand or a detection compound such as a horseradish peroxidase conjugated antibody is allowed to bind to the target species. A dye, such as TMB, is then added and allowed to react with the horseradish peroxidase, causing precipitation and a color change that is detected by a suitable detection device. Further description of protein detection using ArrayTube is found in, for example, Huelseweh B, Ehricht R and Marschall H-J, Proteomics, 2006, 6, 2972-2981; and ClonDiag, ArrayTube (AT) Experiment Guideline for Protein-Based Applications, version 1.2, 2007, all incorporated by reference in their entirety.
  • Transmission detection and analysis is performed with a ClonDiag AT reader instrument. Suitable reader instruments and detection devices include the ArrayTube Workstation ATS and the ATR 03.
  • In addition to ArrayTube, the ClonDiag ArrayStrip (AS) can be used. The ArrayStrip provides a 96-well format for high volume testing. Each ArrayStrip consists of a standard 8-well strip with a microarray integrated into the bottom of each well. Up to 12 ArrayStrips can be inserted into one microplate frame enabling the parallel multiparameter testing of up to 96 samples. The ArrayStrip can be processed using the ArrayStrip Processor ASP, which performs all liquid handling, incubation, and detection steps required in array based analysis. In various embodiments, where a protein is detected, a method of using the ArrayStrip to detect the protein comprises conditioning the AS array with buffer or blocking solution; loading of up to 96 sample solutions in the AS wells to allow for binding of the protein; 3×washing; conjugating with a secondary antibody linked to HRP; 3×washing; precipitation staining with TMB; and AS array imaging and optional data storage.
  • Those skilled in the art will be familiar with numerous additional immunoassay formats and variations thereof which may be useful for carrying out the method disclosed herein. See generally E. Maggio, Enzyme-Immunoassay, (CRC Press, Inc., Boca Raton, Fla., 1980); see also U.S. Pat. Nos. 4,727,022; 4,659,678; 4,376,110; 4,275,149; 4,233,402; and 4,230,767.
  • In general, immunoassays carried out in accordance with the present invention may be homogeneous assays or heterogeneous assays. In a homogeneous assay the immunological reaction usually involves the specific antibody (e.g., anti-biomarker protein antibody), a labeled analyte, and the sample of interest. The signal arising from the label is modified, directly or indirectly, upon the binding of the antibody to the labeled analyte. Both the immunological reaction and detection of the extent thereof can be carried out in a homogeneous solution. Immunochemical labels which may be employed include free radicals, radioisotopes, fluorescent dyes, enzymes, bacteriophages, or coenzymes.
  • In a heterogeneous assay approach, the reagents are usually the sample, the antibody, and means for producing a detectable signal. Samples as described above may be used. The antibody can be immobilized on a support, such as a bead (such as protein A and protein G agarose beads), plate or slide, and contacted with the specimen suspected of containing the antigen in a liquid phase. The support is then separated from the liquid phase and either the support phase or the liquid phase is examined for a detectable signal employing means for producing such signal. The signal is related to the presence of the analyte in the sample. Means for producing a detectable signal include the use of radioactive labels, fluorescent labels, or enzyme labels. For example, if the antigen to be detected contains a second binding site, an antibody which binds to that site can be conjugated to a detectable group and added to the liquid phase reaction solution before the separation step. The presence of the detectable group on the solid support indicates the presence of the antigen in the test sample. Examples of suitable immunoassays include immunoblotting, immunofluorescence methods, immunoprecipitation, chemiluminescence methods, electrochemiluminescence (ECL) or enzyme-linked immunoassays.
  • Antibodies can be conjugated to a solid support suitable for a diagnostic assay (e.g., beads such as protein A or protein G agarose, microspheres, plates, slides or wells formed from materials such as latex or polystyrene) in accordance with known techniques, such as passive binding. Antibodies as described herein may likewise be conjugated to detectable labels or groups such as radiolabels (e.g., 35S, 125I, 131I), enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase), and fluorescent labels (e.g., fluorescein, Alexa, green fluorescent protein, rhodamine) in accordance with known techniques.
  • Using any of the methods and compositions described herein, a sample can be assayed to determine levels of a biomarker panel. Thus, in one aspect, the invention provides a method of assaying a sample from a patient to determine concentrations of a biomarker panel in the sample. In some embodiments, the method comprises contacting the sample with a composition comprising a solid support comprising a capture binding ligand or capture probe for each biomarker of a biomarker panel.
  • The invention further provides kits for use in determining breast health or breast cancer status for a number of medical (including diagnostic and therapeutic), industrial, forensic and research applications. Kits may comprise a carrier, such as a box, carton, tube or the like, having in close confinement therein one or more containers, such as vials, tubes, ampoules, bottles, pouches, envelopes and the like. In various embodiments, the kits comprise one or more components selected from one or more media or media ingredients and reagents for the measurement of the various biomarkers and biomarker panels disclosed herein. For example, kits of the invention may also comprise, in the same or different containers, one or more DNA polymerases, one or more primers, one or more suitable buffers, one or more nucleotides (such as deoxynucleoside triphosphates (dNTPs) and preferably fluorescently labeled dNTPs) and labeling components. The one or more components may be contained within the same container, or may be in separate containers to be admixed prior to use. The kits of the present invention may also comprise one or more instructions or protocols for carrying out the methods of the present invention. The kits may also comprise a computer or a component of a computer, such as a computer-readable storage medium or device. Examples of storage media include, without limitation, optical disks such as CD, DVD and Blu-ray Discs (BD); magneto-optical disks; magnetic media such as magnetic tape and internal hard disks and removable disks; semi-conductor memory devices such as EPROM, EEPROM and flash memory; and RAM. The computer-readable storage medium may comprise software encoding references to the various therapies and treatment regimens disclosed herein. The software may be interpreted by a computer to provide the practitioner with treatments according to various measured concentrations of biomarkers as provided herein. In various embodiments, the kit comprises a biomarker assay involving a lateral-flow-based point-of-care rapid test with detection of risk thresholds, or a biochip with quantitative assays for the constituent biomarkers.
  • Methods of Diagnosing and Treating
  • The compositions and methods of the present invention can be used in the prognosis, diagnosis and treatment of disease in a subject. The invention provides compositions and methods for laboratory and point-of-care tests for measuring biomarkers in a sample from a subject. The invention can be generally applied for a number of different diseases. In exemplary embodiments, the disease is breast cancer.
  • The biomarkers and biomarker panels disclosed herein can be used in methods to diagnose, identify or screen subjects that have, do not have or are at risk for having disease; to monitor subjects that are undergoing therapies for disease; to determine or suggest a new therapy or a change in therapy; to differentially diagnose disease states associated with the disease from other diseases or within sub-classifications of disease; to evaluate the severity or changes in severity of disease in a patient; to stage a subject with the disease and to select or modify therapies or interventions for use in treating subjects with the disease. In an exemplary embodiment, the methods of the present invention are used to identify and/or diagnose subjects who are asymptomatic or presymptomatic for a disease. In this context, “asymptomatic” or “presymptomatic” means not exhibiting the traditional symptoms or enough abnormality for disease.
  • In various embodiments, a method of determining a prognosis of a disease in a subject, diagnosing a disease in a subject, or treating a disease in a subject comprises taking a measurement of a biomarker panel in a sample from the subject. In various exemplary embodiments, the biomarker panel consists of two or more of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and/or CA6.
  • The term “disease status” includes any distinguishable manifestation of the disease, including non-disease. For example, disease status includes, without limitation, the presence or absence of disease, the risk of developing disease, the stage of the disease, the progression of disease (e.g., progress of disease or remission of disease over time), the severity of disease and the effectiveness or response to treatment of disease.
  • A “subject” in the context of the present invention is an animal, preferably a mammal. The mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. In various exemplary embodiments, a subject is human and may be referred to as a patient. Mammals other than humans can be advantageously used as subjects that represent animal models of a disease or for veterinarian applications. A subject can be one who has been previously diagnosed or identified as having a disease, and optionally has already undergone, or is undergoing, a therapeutic intervention for a disease. Alternatively, a subject can also be one who has not been previously diagnosed as having a disease. For example, a subject can be one who exhibits one or more risk factors for a disease, or one who does not exhibit a disease risk factor, or one who is asymptomatic for a disease. A subject can also be one who is suffering from or at risk of developing a disease. In certain embodiments, the subject can be already undergoing therapy or can be a candidate for therapy.
  • As will be appreciated by those in the art, the biomarkers may be measured in using several techniques designed to achieve more predictable subject and analytical variability.
  • The term “sample” refers to a specimen or culture obtained from a subject and includes fluids, gases and solids including for example tissue. In various exemplary embodiments, the sample comprises saliva. As will be appreciated by those in the art, virtually any experimental manipulation or sample preparation steps may have been done on the sample. For example, wash steps and/or fragmentation may be applied to a sample. In various embodiments, a biomarker panel is measured directly in a subject without the need to obtain a separate sample from the patient.
  • In one aspect, the invention provides a method of diagnosing a subject for a disease comprising taking a measurement of a biomarker panel; and correlating the measurement with the disease. The term “correlating” generally refers to determining a relationship between one type of data with another or with a state. In various embodiments, correlating the measurement with disease comprises comparing the measurement with a reference biomarker profile or some other reference value. In various embodiments, correlating the measurement with disease comprises determining whether the subject is currently in a state of disease.
  • The quantity or activity measurements of a biomarker panel can be compared to a reference value. Differences in the measurements of biomarkers in the subject sample compared to the reference value are then identified. In exemplary embodiments, the reference value is given by a risk category as described further below.
  • In various embodiments, the reference value is a baseline value. A baseline value is a composite sample of an effective amount of biomarkers from one or more subjects who do not have a disease, who are asymptomatic for a disease or who have a certain level of a disease. A baseline value can also comprise the amounts of biomarkers in a sample derived from a subject who has shown an improvement in risk factors of a disease as a result of treatments or therapies. In these embodiments, to make comparisons to the subject-derived sample, the amounts of biomarkers are similarly calculated. A reference value can also comprise the amounts of biomarkers derived from subjects who have a disease confirmed by an invasive or non-invasive technique, or are at high risk for developing a disease. Optionally, subjects identified as having a disease, or being at increased risk of developing a disease are chosen to receive a therapeutic regimen to slow the progression of a disease, or decrease or prevent the risk of developing a disease. A disease is considered to be progressive (or, alternatively, the treatment does not prevent progression) if the amount of biomarker changes over time relative to the reference value, whereas a disease is not progressive if the amount of biomarkers remains constant over time (relative to the reference population, or “constant” as used herein). The term “constant” as used in the context of the present invention is construed to include changes over time with respect to the reference value.
  • The biomarkers of the present invention can be used to generate a “reference biomarker profile” of those subjects who do not have a disease according to a certain threshold, are not at risk of having a disease or would not be expected to develop a disease. The biomarkers disclosed herein can also be used to generate a “subject biomarker profile” taken from subjects who have a disease or are at risk for having a disease. The subject biomarker profiles can be compared to a reference biomarker profile to diagnose or identify subjects at risk for developing a disease, to monitor the progression of disease, as well as the rate of progression of disease, and to monitor the effectiveness of disease treatment modalities. The reference and subject biomarker profiles of the present invention can be contained in a machine-readable medium, such as but not limited to, analog tapes like those readable by a VCR; optical media such as CD-ROM, DVD-ROM and the like; and solid state memory, among others.
  • Measurements of the biomarker panels of the invention can lead a practitioner to affect a therapy with respect to a subject. Thus, the invention provides methods of treating a disease in a subject comprising taking a measurement of a biomarker panel in a sample from the subject, and affecting a therapy with respect to the subject. The terms “therapy” and “treatment” may be used interchangeably. In certain embodiments, the therapy can be selected from, without limitation, initiating therapy, continuing therapy, modifying therapy or ending therapy. A therapy also includes any prophylactic measures that may be taken to prevent disease.
  • In certain embodiments, treatment comprises administering a disease-modulating drug to a subject. The drug can be a therapeutic or prophylactic used in subjects diagnosed or identified with a disease or at risk of having the disease. In certain embodiments, modifying therapy refers to altering the duration, frequency or intensity of therapy, for example, altering dosage levels.
  • In various embodiments, effecting a therapy comprises causing a subject to or communicating to a subject the need to make a change in lifestyle, for example, increasing exercise, changing diet, reducing or eliminating smoking and so on. The therapy can also include surgery, for example, mastectomy.
  • Measurement of biomarker levels allow for the course of treatment of a disease to be monitored. The effectiveness of a treatment regimen for a disease can be monitored by detecting one or more biomarkers in an effective amount from samples obtained from a subject over time and comparing the amount of biomarkers detected. For example, a first sample can be obtained prior to the subject receiving treatment and one or more subsequent samples are taken after or during treatment of the subject. Changes in biomarker levels across the samples may provide an indication as to the effectiveness of the therapy.
  • To identify therapeutics or drugs that are appropriate for a specific subject, a test sample from the subject can also be exposed to a therapeutic agent or a drug, and the level of one or more biomarkers can be determined. Biomarker levels can be compared to a sample derived from the subject before and after treatment or exposure to a therapeutic agent or a drug, or can be compared to samples derived from one or more subjects who have shown improvements relative to a disease as a result of such treatment or exposure. Thus, in one aspect, the invention provides a method of assessing the efficacy of a therapy with respect to a subject comprising taking a first measurement of a biomarker panel in a first sample from the subject; effecting the therapy with respect to the subject; taking a second measurement of the biomarker panel in a second sample from the subject and comparing the first and second measurements to assess the efficacy of the therapy.
  • Additionally, therapeutic or prophylactic agents suitable for administration to a particular subject can be identified by detecting a biomarker (which may be two or more) in an effective amount from a sample obtained from a subject and exposing the subject-derived sample to a test compound that determines the amount of the biomarker(s) in the subject-derived sample. Accordingly, treatments or therapeutic regimens for use in subjects having a disease or subjects at risk for developing a disease can be selected based on the amounts of biomarkers in samples obtained from the subjects and compared to a reference value. Two or more treatments or therapeutic regimens can be evaluated in parallel to determine which treatment or therapeutic regimen would be the most efficacious for use in a subject to delay onset, or slow progression of a disease. In various embodiments, a recommendation is made on whether to initiate or continue treatment of a disease.
  • Drug Treatments
  • In various exemplary embodiments, effecting a therapy comprises administering a disease-modulating drug to the subject. The subject may be treated with one or more disease-modulating drugs until altered levels of the measured biomarkers return to a baseline value measured in a population not suffering from the disease, experiencing a less severe stage or form of a disease or showing improvements in disease biomarkers as a result of treatment with a disease-modulating drug. Additionally, improvements related to a changed level of a biomarker or clinical parameter may be the result of treatment with a disease-modulating drug.
  • A number of compounds such as a disease-modulating drug may be used to treat a subject and to monitor progress using the methods of the invention. In certain embodiments, the disease-modulating drug comprises
  • The beneficial effects of these and other drugs can be visualized by assessment of clinical and laboratory biomarkers.
  • Any drug or combination of drugs disclosed herein may be administered to a subject to treat a disease. The drugs herein can be formulated in any number of ways, often according to various known formulations in the art or as disclosed or referenced herein.
  • In various embodiments, any drug or combination of drugs disclosed herein is not administered to a subject to treat a disease. In these embodiments, the practitioner may refrain from administering the drug or combination of drugs, may recommend that the subject not be administered the drug or combination of drugs or may prevent the subject from being administered the drug or combination of drugs.
  • In various embodiments, one or more additional drugs may be optionally administered in addition to those that are recommended or have been administered. An additional drug will typically not be any drug that is not recommended or that should be avoided. In exemplary embodiments, one or more additional drugs comprise one or more glucose lowering drugs.
  • Decision Matrices
  • The therapy chosen by a practitioner can depend on the concentrations of biomarkers determined in a sample. In various exemplary embodiments, the therapy depends on which category from a range of categories particular to each biomarker the measured concentration of each biomarker falls in. In various exemplary embodiments, the therapy depends on the combination of risk levels for different symptoms or diseases that are indicated by a biomarker panel.
  • With respect to concentration measurements of a biomarker, the term “category” refers to a subset of a partition of the possible concentrations that a biomarker may have. Each category may be associated with a label or classification chosen by the practitioner. The labels may be refer to, for example, the risk level of an individual for having or being subject to a disease state. The categories and labels may be derived from the current literature or according to the findings of the practitioner.
  • Each biomarker of a biomarker panel can thus be associated with a discrete set of categories, for example, risk categories. Combining one category from each biomarker forms a “decision point.” In various exemplary embodiments, the complete set of decision points comprises all possible n-tuples of categories, wherein n is the number of biomarkers in the biomarker panel. This complete set will have m1×m2× . . . mn possible decision points, wherein in is the number of categories for biomarker i.
  • Every decision point can be associated with a condition or a disease state, which is not necessarily unique. That is, one or more decision points can be associated with the same disease state. The association of every possible decision point with a condition or disease state can be referred to as a “disease classification matrix” or a “disease classification tree.” Thus, by correlating a measurement of a biomarker panel with a decision point, the practitioner can classify the condition or disease state of a patient.
  • Every decision point can also be associated with a particular therapy, which is not necessarily unique. That is, one or more decision points can be associated with the same therapy. The association of every possible decision point with one or more therapies can be referred to as a “therapy decision matrix” or “therapy decision tree.”
  • Each decision point can be associated with more than one type of information. For example, both disease state and therapy can be indicated by a decision point.
  • The articles “a,” “an” and “the” as used herein do not exclude a plural number of the referent, unless context clearly dictates otherwise. The conjunction “or” is not mutually exclusive, unless context clearly dictates otherwise. The term “include” is used to refer to non-limiting examples.
  • EXAMPLES
  • The following examples are offered to illustrate, but not to limit the invention.
  • Example 1: Salivary Transciptomic Profiling and Analysis Saliva Collection
  • Unstimulated whole saliva samples were collected with previously established protocols. Subjects were asked to refrain from eating, drinking, smoking, or oral hygiene procedures for at least 30 minutes before the collection. Lipstick was wiped off, and the subject rinsed her mouth once with plain water. Typically, patients donated approximately 5-10 ml of saliva. Samples were then centrifuged at 2,600 g for 15 minutes at 4° C. The supernatant was then stored at −80° C. until use. Of note, protease inhibitors cocktail, containing 1 μl aprotinin, 10 μl PMSF (phenylmethanesulfonyl fluoride) and 3 μl sodium orthovanadate (all from Sigma, St. Louis, Mo.) were added to each 1 ml saliva sample.
  • mRNA Isolation and Analysis
  • RNA was isolated from 330 μl of saliva supernatant using MagMax™ Viral RNA Isolation Kit (Ambion, Austin, Tex.). This process was automated using KingFisher® mL technology (Thermo Fisher Scientific, Waltham, Mass.), followed by TURBO™ DNase treatment (Ambion, Austin, Tex.) to remove contaminating DNA. 90 μl of extracted RNA (out of 100 μl) was concentrated to 11 μl and was linearly amplified using the RiboAmp® RNA Amplification kit (Molecular Devices, Sunnyvale, Calif.). After purification, cDNA was transcribed and biotinylated using GeneChip® Expression 3′-Amplification Reagents for in vitro transcription labeling (Affymetrix, Santa Clara, Calif.). Approximately 20 μg of labeled RNA were subsequently submitted for GeneChip® analysis using an Affymetrix Human Genome U133 Plus 2.0 Array. Chip hybridization and scanning were performed using the MIAME (Minimum Information About a Microarray Experiment) criteria. All Affymetrix Human Genome U133 Plus 2.0 Array data generated in this study were uploaded to the GEO database, accession number GSE20266.
  • Gene Array Statistical Analysis
  • The CEL files from all databases were imported into the statistical R 2.7.0 (hypertext transfer protocol://www.r-project.org) with same and ROC packages. The Probe Logarithmic Intensity Error Estimation (PLIER) expression measures were computed after background correction and quantile normalization for each microarray dataset. Probeset-level quantile normalization was performed across all samples to make the effect sizes similar among all datasets. Finally, for every probeset, significance analysis of microarray (SAM) was applied to identify differential expression between the cancer and healthy control samples. The probesets were then ranked by the false discovery rate (FDR) corrected p-values.
  • Screening of Biomarker Candidates
  • The biomarker candidates generated by microarray profiling were subjected to further screening by real-time quantitative RT-PCR (qPCR) on the same set of samples used for the microarray analysis. To accomplish this, total RNA was reverse-transcribed using reverse transcriptase and gene-specific primers using the following thermal cycling conditions: 1 min at 60° C., 30 min at 50° C., 2 min at 95° C., followed by 15 cycles of 15s at 95° C., 30s at 50° C., 10 s at 72° C. These steps were followed with a final extension of 5 min. at 72° C. and then cooling to 4° C. The preamplified product was cleaned using ExoSAP-IT (USB Corporation) and diluted 1/40 in water. 2 μl of the cDNA was used for qPCR.
  • qPCR was carried out in a 96-well plate in a reaction volume of 10 μl using power SYBR®-Green Master Mix (Applied Biosystems, Foster City, Calif.) for 15 min at 95° C. for initial denaturing, followed by 40 cycles of 95° C. for 30 s and 60° C. for 30 s in the ABI 7500HT Fast Real Time PCR system (Applied Biosystems, Foster City, Calif.). All qPCRS were performed in duplicate for all candidate mRNA. The specificity of the PCR was confirmed according to the melting curve of each gene, and the average threshold cycle (Ct) was examined.
  • Amplicon lengths were around 100-130 by for the outer primer pairs used in preamplification and 60-80 bp for the inner primer pairs used in qPCR. RT-qPCR primers were designed using Primer Express 3.0 software (Applied Biosystems, Foster City, Calif.). All primers were synthesized by Sigma-Genosys (Woodlands, Tex.), and the amplicons were intron spanning whenever possible.
  • Raw data were normalized by subtracting GAPDH Ct values from the biomarker Ct values to generate ΔCt. The Mann-Whitney rank sum test was used for between-group biomarker comparisons.
  • Primers for 11 Candidate Biomarkers and GAPDH
  • Gene symbol Primer name Primer sequences (5′-3′)
    ATXN3
    ATXN3-OF 
    GAAAAACAGCAGCAAAAGCA
    ATXN3-IF 
    GGGGGACCTATCAGGACAGA
    ATXN3-IR 
    CAAGTGCTCCTGAACTGGTG
    ATXN3-OR 
    CCAAGTGCTCCTGAACTGGT
    GRIK1
    GRIK1-OF 
    CCGGACTGGTCCTTTCTGTA
    GRIK1-IF 
    CCGGACTGGTCCTTTCTGTA
    GRIK1-IR 
    AGCGTTGAAAGAGAGACACTG
    GRIK1-OR 
    CAGTGAGATTCCCAGTTCTTCC
    GRM1 
    GRM1-OF 
    GCAGGGAATGCCAATTCTAA
    GRM1-IF 
    TGGCAAGTCTGTGTCATGGT
    GRM1-IR 
    GCCACATATGCTGTCCCTTG
    GRM1-OR
    GCCGTCTCATTGGTCTTCAC
    TPT1 
    TPT1-OF 
    TACCGTGAGGATGGTGTGAC
    TPT1-IF 
    CAAATGTGGCAATTATTTTGGA
    TPT1-IR 
    GATGACAAGCAGAAGCCAGTT
    TPT1-OR 
    GATGACAAGCAGAAGCCAGT
    RGS13  
    RGS13-OF
    CTCACGGTGGAGCAGAATTT
    RGS13-IF 
    CTCACGGTGGAGCAGAATTT
    RGS13-IR 
    GGGACTGTGGCTGGATGTAA
    RGS13-OR 
    TGGGTTCCTGAATGTTCCTG
    S100A8 
    S100A8-OF 
    TCAGGAAAAAGGGTGCAGAC
    S100A8-IF 
    TCAGGAAAAAGGGTGCAGAC
    S100A8-IR 
    TGGAAGTTAACTGCACCATCA
    S100A8-OR 
    ACGCCCATCTTTATCACCAG
    CLDN15 
    CLDN15-OF 
    TTGTACCCCGGAACCAAGTA
    CLDN15-IF 
    CGGAACCAAGTACGAGCTG
    CLDN15-IR 
    CACCCAGGATGGAGATCAGT
    CLDN15-OR 
    CTGGGTCCTCGTCAGAGC
    IGF2BP1 
    IGF2B P1-OF 
    AGAATTTGACGGCAGCTGAG
    IGF2BP1-IF 
    CCAGGTCATCGTGAAAATCA
    IGF2BP1-IR 
    ATCTTCCGTTGAGCCATCTG
    IGF2BP1-OR
    ATGTCTCGGATCTTCCGTTG
    CSTA 
    CSTA-OF
    ACGGAAAATTGGAAGCTGTG
    CSTA-IF 
    CATTAAGGTACGAGCAGGTGA
    CSTA-IR 
    TTTGTCCGGGAAGACTTTTG
    CSTA-OR 
    TTTGTCCGGGAAGACTTTTG
    MDM4  
    MDM4-OF
    GTGGCAGTGTACTGAATGCAA
    MDM4-IF 
    TGGCAGTGTACTGAATGCAA
    MDM4-IR
    AAGGCCCAACAACGAAAAC
    MDM4-OR
    TCAGACGTGGAGAGAGAATGG
    H6PD 
    H6PD-OF 
    GGCACAAGCTTCAGGTCTTC
    H6PD-IF 
    GTCGTGGGCCAGTACCAGT
    H6PD-IR 
    GTGGAAGCTGTCTGGCTTCT
    H6PD-OR 
    GTGGAAGCTGTCTGGCTTCT
    GAPDH 
    GAPDH-OF 
    CATTGCCCTCAACGACCACTT
    GAPDH-IF 
    ACCACTTTGTCAAGCTCATTTCCT
    GAPDH-IR 
    CACCCTGTTGCTGTAGCCAAAT
    GAPDH-OR 
    ATGTGGGCCATGAGGTCCA

    OF=Outer forward, IF=Inner forward, IR=Inner reverse, OR=Outer reverse. All primers were designed using Primer Express 3.0 software (Applied Biosystems, FosterCity, Calif.). The specificity of primers was checked using NCBI's GenBank BLAST search.
  • The data analysis for qPCR was performed using the 2−Ct method, where GAPDH is used as the reference gene. The qPCR based gene expression values between two groups were compared using the non-parametric Wilcoxon test. To normalize for RNA input, qPCR was also performed for GAPDH. Raw data were normalized by subtracting GAPDH Ct values from the marker Ct values to provide ΔCt and then analyze with the stats, utilities packages from R 2.7.0 (world wide web.r-project.org) and the ROC package from Bioconductor 2.2 (world wide web.bioconductor.org). Statistical comparisons were made with the use of the Mann-Whitney U test with consideration of two different distributions for control and pancreatic cancer groups. Biomarkers that differentiated between groups of subjects (P value <0.05) were identified and compared by Area Under Curve (AUC) value. The AUC is based on constructing a receiver operating characteristic (ROC) curve which plots the sensitivity versus one minus the specificity. The AUC value is computed by numerical integration of the ROC curve. The range of this value can be 0.5 to 1.0. A value of 0.5 indicates that the biomarker is no better that a coin toss, while 1.0 indicates the relatively best diagnostic accuracy.
  • Example 2: Salivary Proteomic Profiling and Analysis Protein Isolation and Analysis
  • Saliva from 13 healthy control subjects and 13 breast cancer subjects were centrifuged at 2600 g at 4° C. for 15 minutes. Saliva supernatant from the 13 health control subjects and 13 breast cancer subjects were pooled to form a control sample and a cancer sample for proteomic profiling. 250 μg of proteins in the pooled saliva samples were precipitated by methanol and then resuspended in 2-D cell lysis buffer (30 mM Tris-HCl, pH 8.8, containing 7M urea, 2M thiourea and 4% CHAPS detergent). The total proteins of each pooled sample, breast cancer and control, were labeled with the cyanine dyes Cy2 and Cy5 respectively. The two labeled sample sets were then combined and subjected to two-dimensional difference gel electrophoresis. After loading the labeled samples, isoelectric focusing (IEF) (pH3-10) was run following the protocol provided by Amersham BioSciences. The IPG strips were rinses in the SDS-gel running buffer before transferring to 13.5% SDS-gels. The SDS-gels were run at 15° C. until the dye front ran out of the gels. Gel images were scanned immediately following the SDS-PAGE using Typhoon TRIO™ (Amersham BioSciences). The fold change of the protein expression levels was obtained from in-gel DeCyder™ analysis.
  • Spots with fold changes larger than 1.5 on the gel were cut and then were washed multiple times to remove staining dye and other chemicals. Gel spots were dried to absorb maximum volume of digestion buffer. Dried 2D gel spots were rehydrated in digestion buffer containing sequencing grade modified trypsin (Promega, USA). Proteins were digested in-gel at 37° C. overnight. Digested peptides were extracted from the gel with TFA extraction buffer and with shaking. The digested tryptic peptides were desalted using C-18 Zip-tips (Millipore). The desalted peptides were mixed with CHCA matrix (α-cyano-4-hydroxycinnamic acid) and spotted into wells of a MALDI plate for MALDI-TOF MS (ABI4800) identification. Protein identification was based on peptide fingerprint mass mapping (using MS spectra) and peptide fragmentation mapping (using MS/MS spectra). Combined MS and MS/MS spectra were submitted for database search using GPS Explorer software equipped with the MASCOT search engine to identify proteins from primary sequence databases.
  • Screening of Biomarker Candidates
  • Four proteins (carbonic anhydrase VI, psoriasin, Transthyretin and Cyclophilin A) identified in the 2-D gel analysis (above) were subjected to Western blot analysis on the original sample set. Reduced protein samples (15 μs total protein per lane) were loaded onto a 10% bis-Tris gel and run at 150V in MES SDS running buffer for one hour. Pre-stained protein standard (Invitrogen) was used to track protein migration. The proteins were transferred to nitrocellulose membrane by using iBlot® (Invitrogen). The membrane was then washed in wash buffer containing 10 mM Tris-HCl, pH 7.6, 150 mM NaCl, and 0.1% (v/v) Tween®-20 (Sigma-Aldrich) before blocking for one hour in wash buffer containing 5% non-fat dry milk. After further washes in wash buffer, the membrane was incubated with primary antibody (mouse anti-human carbonic anhydrase VI (Lifespan Biotech) at 1 μg/ml, mouse anti-psoriasin (Abeam) at 1 μg/ml, mouse anti-actin (Sigma-Aldrich) at 1 μg/ml according to manufacturers instructions in blocking buffer at room temperature for 2 h. The membrane was then washed before applying the secondary antibody (anti-mouse IgG peroxidase-linked species specific whole antibody from sheep, GE Healthcare) according to manufacturer's instructions for one hour at room temperature. Finally, the membrane was washed and visualized using ECL Plus™ detection kit (GE Healthcare). The signal intensity of the bands was measured using Image J software (NIH, Bethesda, Md., USA). The intensity of a band representing the protein of interest was divided by the intensity of it corresponding (3-actin expression on the same blot for normalization.
  • The protein expression pattern of carbonic anhydrase VI and psoriasin was further tested by Western blot with a new subject sample set including 31 cancer subject samples and 62 control subject samples. All the samples were coded with a random number from 1 to 93 and used for blind testing by Western blot. The distribution of carbonic anhydrase VI shows significant difference in the cancer group as compared to the control group (p=0.009949).
  • Example 4: Screening Method
  • A patient undergoing routine dental care is screened during the visit. For example, a 62 year old female patient, and former smoker, prior to oral exam is asked to provide a saliva sample. A saliva sample is collected and analyzed either at the point of care or is submitted for analysis by a reference laboratory. The saliva sample is tested for the biomarkers of the instant invention and optionally other biomarkers. Results from the analysis are provided to the dental professional and the patient is informed as to whether she has breast cancer.
  • (S100A8)(NM_002964.4) 
    SEQ ID NO: 1
    gagaaaccag agactgtagc aactctggca gggagaagct
    gtctctgatg gcctgaagctgtgggcagct ggccaagcct
    aaccgctata aaaaggagct gcctctcagc
    cctgcatgtctcagtcagc tgtattcag aagacctggt
    ggggcaagtc cgtgggcatc atgttgaccgagctggagaa
    agccttgaac tctatcatcg acgtctacca caagtactcc
    ctgataaaggggaatttcca tgccgtctac agggatgacc
    tgaagaaatt gctagagacc gagtgtcctcagtatatcag
    gaaaaagggt gcagacgtct ggttcaaaga gttggatatc
    aacactgatggtgcagttaa cttccaggag ttcctcattc
    tggtgataaa gatgggcgtg gcagcccaca aaaaaagcca
    tgaagaaagc cacaaagagt agctgagtta ctgggcccag
    aggctgggcc cctggacatg tacctgcaga ataataaagt
    catcaatacc tcaaaaaaaa aa
    (CSTA)(NM_005213) 
    SEQ ID NO: 2
    tgctgtttgt ggaaaataaa gcattctata ggcggagcta
    gtgaacgcct cttttaaaacacgagtctcc acacttccct
    gttcactttg gttccagcat cctgtccagc aaagaagcaa
    tcagccaaaa tgatacctgg aggcttatct gaggccaaac
    ccgccactcc agaaatccag gagattgttg ataaggttaa
    accacagctt gaagaaaaaa caaatgagac ttacggaaaa
    ttggaagctg tgcagtataa aactcaagtt gttgctggaa
    caaattacta cattaaggta cgagcaggtg ataataaata
    tatgcacttg aaagtattca aaagtcttcc cggacaaaat
    gaggacttgg tacttactgg ataccaggtt gacaaaaaca 
    aggatgacga gctgacgggc ttttagcagc atgtacccaa
    agtgttctga ttccttcaac tggctactga gtcatgatcc
    ttgctgataa atataaccat caataaagaa gcattctttt
    ccaaagaaat tatttatca attatttctc atttattgta
    ttaagcagaa attacctttt ctttctcaaa atcagtgtta
    ttgctttaga gtataaactc catataaatt gatggcaatt
    ggaaatctta taaaaactag tcaagcctaa tgcaactggc
    taaaggatag taccaccctc acccccacca taggcaggct
    ggatcgtgga ctatcaattc accagcctcc ttgttccctg
    tggctgctga taacccaaca ttccatctct accctcatac
    ttcaaaatta aatcaagtat tttacaaaaa aaaaaaaa 
    (GRM1)(NM_001114329)
    SEQ ID NO: 3
    agtgctgaag aaagagggca ctagtgtaca gcccagatcg
    catccttgca ccgtctggat tagagctgag gcgtctgcaa
    gccgagcgtg gccacggtcc tctggccccg ggaccatagc
    gctgtctacc ccgactcagg tactcagcag catctagctc 
    accgctgcca acacgacttc cactgtactc ttgatcaatt
    taccttgatg cactaccggt gaagaacggg gactcgaatt
    cccttacaaa cgcctccagc ttgtagaggc ggtcgtggag
    gacccagagg aggagacgaa ggggaaggag gcggtggtgg
    aggaggcaaa ggccttggac gaccattgtt ggcgaggggc
    accactccgg gagaggcggc gctgggcgtc ttgggggtgc
    gcgccgggag cctgcagcgg gaccagcgtg ggaacgcggc 
    tggcaggctg tggacctcgt cctcaccacc atggtcgggc
    tccttttgtt ttttttccca gcgatctttt tggaggtgtc 
    ccttctcccc agaagccccg gcaggaaagt gttgctggca
    ggagcgtcgt ctcagcgctc ggtggccaga atggacggag
    atgtcatcat tggagccctc ttctcagtcc atcaccagcc
    tccggccgag aaagtgcccg agaggaagtg tggggagatc
    agggagcagt atggcatcca gagggtggag gccatgttcc
    acacgttgga taagatcaac gcggacccgg tcctcctgcc
    caacatcacc ctgggcagtg agatccggga ctcctgctgg 
    cactatccg tggctctgga acagagcatt gagttcatta
    gggactctct gatttccatt cgagatgaga aggatgggat
    caaccggtgt ctgcctgacg gccagtccct ccccccaggc
    aggactaaga agcccattgc gggagtgatc ggtcccggct
    ccagctctgt agccattcaa gtgcagaacc tgctccagct
    cttcgacatc ccccagatcg cttattcagc cacaagcatc
    gacctgagtg acaaaacttt gtacaaatac ttcctgaggg
    ttgtcccttc tgacactttg caggcaaggg ccatgcttga
    catagtcaaa cgttacaatt ggacctatgt ctctgcagtc 
    cacacggaag ggaattatgg ggagagcgga atggacgctt
    tcaaagagct ggctgcccag gaaggcctct gtatcgccca
    ttctgacaaa atctacagca acgctgggga gaagagcta
    gaccgactct tgcgcaaact ccgagagagg cttcccaagg
    ctagagtggt ggtctgcttc tgtgaaggca tgacagtgcg
    aggactcctg agcgccatgc ggcgccttgg cgtcgtgggc
    gagttctcac tcattggaag tgatggatgg gcagacagag 
    atgaagtcat tgaaggttat gaggtggaag ccaacggggg
    aatcacgata aagctgcagt ctccagaggt caggtcattt
    gatgattatt tcctgaaact gaggctggac actaacacga
    ggaatccctg gttccctgag ttctggcaac atcggttcca
    gtgccgcctt ccaggacacc ttctggaaaa tcccaacttt
    aaacgaatct gcacaggcaa tgaaagctta gaagaaaact
    atgtccagga cagtaagatg gggtttgtca tcaatgccat 
    ctatgccatg gcacatgggc tgcagaacat gcaccatgcc
    ctctgccctg gccacgtggg cctctgcgat gccatgaagc
    ccatcgacgg cagcaagctg ctggacttcc tcatcaagtc
    ctcattcatt ggagtatctg gagaggaggt gtggtttgat
    gagaaaggag acgctcctgg aaggtatgat atcatgaatc
    tgcagtacac tgaagctaat cgctatgact atgtgcacgt
    tggaacctgg catgaaggag tgctgaacat tgatgattac 
    aaaatccaga tgaacaagag tggagtggtg cggtctgtgt
    gcagtgagcc ttgcttaaag ggccagatta aggttatacg
    gaaaggagaa gtgagctgct gctggatttg cacggcctgc
    aaagagaatg aatatgtgca agatgagttc acctgcaaag
    cttgtgactt gggatggtgg cccaatgcag atctaacagg
    ctgtgagccc attcctgtgc gctatcttga gtggagcaac
    atcgaatcca ttatagccat cgccttttca tgcctgggaa
    tccttgttac cttgtttgtc accctaatct ttgtactgta
    ccgggacaca ccagtggtca aatcctccag tcgggagctc
    tgctacatca tcctagctgg catcttcctt ggttatgtgt
    gcccattcac tctcattgcc aaacctacta ccacctcctg
    ctacctccag cgcctcttgg ttggcctctc ctctgcgatg
    tgctactctg ctttagtgac taaaaccaat cgtattgcac
    gcatcctggc tggcagcaag aagaagatct gcacccggaa
    gcccaggttc atgagtgcct gggctcaggt gatcattgcc 
    tcaattctga ttagtgtgca actaaccctg gtggtaaccc
    tgatcatcat ggaaccccct atgcccattc tgtcctaccc 
    aagtatcaag gaagtctacc ttatctgcaa taccagcaac
    ctgggtgtgg tggccccttt gggctacaat ggactcctca
    tcatgagctg tacctactat gccttcaaga cccgcaacgt
    gcccgccaac ttcaacgagg ccaaatatat cgcgttcacc
    atgtacacca cctgtatcat ctggctagct tttgtgccca
    tttactttgg gagcaactac aagatcatca caacttgat
    tgcagtgagt ctcagtgtaa cagtggctct ggggtgcatg
    ttcactccca agatgtacat cattattgcc aagcctgaga
    ggaatgtccg cagtgccttc accacctctg atgttgtccg
    catgcatgtt ggcgatggca agctgccctg ccgctccaac
    actttcctca acatcttccg aagaaagaag gcaggggcag 
    ggaatgccaa gaagaggcag ccagaattct cgcccaccag
    ccaatgtccg tcggcacatg tgcagctttg aaaaccccca
    cactgcagtg aatgtttcta atggcaagtc tgtgtcatgg
    tctgaaccag gtggaggaca ggtgcccaag ggacagcata
    tgtggcaccg cctctctgtg cacgtgaaga ccaatgagac
    ggcctgcaac caaacagccg tcatcaagcc cctcactaaa
    agttaccaag gctctggcaa gagcctgacc tatcagata 
    ccagcaccaa gaccattac aacgtagagg aggaggagga
    tgcccagccg attcgcttta gcccgcctgg tagccatcc
    atggtggtgc acaggcgcgt gccaagcgcg gcgaccactc
    cgcctctgcc gtcccacctg accgcagagg agacccccct
    cttcctggcc gaaccagccc tccccaaggg cttgccccct
    cctctccagc agcagcagca accccctcca cagcagaaat
    cgctgatgga ccagctccag ggagtggtca gcaacttcag 
    taccgcgatc ccggattttc acgcggtgct ggcaggcccc
    ggtggtcccg ggaacgggct gcggtccctg tacccgcccc
    cgccacctcc gcagcacctg cagatgctgc cgctgcagct
    gagcaccttt ggggaggagc tggtctcccc gcccgcggac
    gacgacgacg acagcgagag gtttaagctc ctccaggagt
    acgtgtatga gcacgagcgg gaagggaaca cggaagaaga
    cgaactggaa gaggaggagg aggacctgca ggcggccagc 
    aaactgaccc cggatgattc gcctgcgctg acgcctccgt
    cgcctttccg cgactcggtg gcctcgggca gctcggtgcc
    cagctccccc gtgtccgagt cggtgctctg cacccctccc
    aacgtatcct acgcctctgt cattctgcgg gactacaagc
    aaagctcttc caccctgtaa gggggaaggg tccacataga
    aaagcaagac aagccagaga tctcccacac ctccagagat
    gtgcaaacag ctgggaggaa aagcctggga gtggggggcc 
    tcgtcgggag gacaggagac cgctgctgct gctgccgcta
    ctgctgctgc tgccttaagt aggaagagag ggaaggacac
    caagcaaaaa atgttccagg ccaggattcg gattcttgaa
    ttactcgaag ccttctctgg gaagaaaggg aattctgaca
    aagcacaatt ccatatggta tgtaactttt atcacaaatc
    aaatagtgac atcacaaaca taatgtcctc ttttgcacaa
    ttgtgcatag atatatatat gcccacacac actgggccat
    gcttgccaag gaacagccca cgtggacatg ccagtcggat
    catgagttca cctgatggca ttcggagtga gctggtggag 
    ccagacagag caggtgcggg gaagggaagg gcccaggcca
    gacccatccc aaacggatga tgggatgatg ggacagcagc
    tccttgctca gaagcccttc tccccgctgg gctgacagac
    tcctcatctt caggagactc aggaatggag cggcacaggg
    gtctctcttc atccactgca acccatccag tgccagatt
    gagattgcac ttgaagaaag gtgcatggac cccctgctgc
    tctgcagatt ccctttattt aggaaaacag gaataagagc 
    aaaattatca ccaaaaagtg cttcatcagg cgtgctacag
    gaggaaggag ctagaaatag aacaatccat cagcatgaga
    ctttgaaaaa aaaacacatg atcagcttct catgttccat
    attcacttat tggcgatttg gggaaaaggc cggaacaaga
    gattgttacg agagtggcag aaaccctttt gtagattgac
    ttgtgtttgt gccaagcggg ctttccattg accttcagtt
    aaagaacaaa ccatgtgaca aaattgttac cttccactta
    ctgtagcaaa taatacctac aagttgaact tctaagatgc
    gtatatgtac aatttggtgc cattatttct cctacgtatt
    agagaaacaa atccatcttt gaatctaatg gtgtactcat
    agcaactatt actggtttaa atgacaaata attctatcct
    attgtcactg aagtccttgt aactagcgag tgaatgtgtt
    cctgtgtcct tgtatatgtg cgatcgtaaa atttgtgcaa
    tgtaatgtca aattgactgg tcaatgtcaa cctagtagtc
    aatctaactg caattagaaa ttgtcttttg aatatactat
    atatattttt tatgttccaa taatgttttg tacatcattg
    tcatcaatat ctacagaagc tctttgacgg tttgaatact
    atggctcaag gttttcatat gcagctcgga tggacatttt
    tcttctaaga tggaacttat ttttcagata ttttctgatg
    tggagatatg ttattaatga agtggtttga aaatttgtta
    tattaaaagt gcacaaaaac tgagagtgaa aataaaaggt
    acattttata agcttgcaca cattattaac acataagatt
    gaacaaagca tttagattat tccaggttat atcatttttt
    taaagatttt ccacagctac ttgagtgtct aacatacagt
    aacatctaac tcagctaata atttgtaaaa tctttatcaa
    tcacattttg ccttctttta atttttatgt tcatggactt
    ttattcctgt gtcttggctg tcataacttt ttatttctgc
    tatttgctgt tgtgtaatat ccatggacat gtaatccact
    tactccatct ttacaatccc tttttaccac caataaaagg
    atttttcttg ctgttttgat ttcttctatt atttgtggaa
    tgaattatac cccccttaaa tatctttgtt tatgccttat
    gttcagtcat attttaatat gatccttca tattgaagct
    gctgatttct cagccaaaaa tcatcttaga atctttaaat
    atccattgca tcatttgac agaatttaac atccattcca
    atgttggagg cttgtattac ttatatttca tcatattcta
    ttgccaagtt tagtcagttc cacaccaaga atgaactgca
    tttcctttaa aaattatttt aaaacacctt tattgaaaag
    atctcatgac tgagatgtgg actttggttc catgttttca
    ttgtaagaaa gcagagagcg gaaaatcaat ggctccagtg
    attaatagat gggtttttag taattgacaa attcatgagg
    gaaagcatat gatctcttta ttagtgaatc atgcttattt
    tttactata atgccactaa tatacatccc taatatcaca
    gggcttgtgc attcagattt ttaaaaaatt aggatagata
    aggaaacaac ttatattcaa gtgtaagatg atatcaggtt
    ggtctaagac ttttggtgaa cacgttcatt caactgtgat
    cactttatta ctctgaatgc ctactattat cctgattatg
    gggtctcctg aataaataga gtattagtcc ttatgtcatc
    attgttcaaa attggagatg tacacataca taccctatac
    caagagggcc gaaactcttc accttgatgt atgttctgat
    acaagttgtt cagcttcttg taaatgtgtt ttccttcggc
    ttgttactgc cttttgtcaa ataatcttga caatgctgta
    taataaatat tttctattt 
    (TPT1)(NM_003295.2)
    SEQ ID NO: 4
    ccccccgagc gccgctccgg ctgcaccgcg ctcgctccga
    gtttcaggct cgtgctaagctagcgccgtc gtcgtctccc
    ttcagtcgcc atcatgatta tctaccggga cctcatcagc
    cacgatgaga tgttctccga catctacaag atccgggaga
    tcgcggacgg gttgtgcctg gaggtggagg ggaagatggt
    cagtaggaca gaaggtaaca ttgatgactc gctcattggt
    ggaaatgcct ccgctgaagg ccccgagggc gaaggtaccg
    aaagcacagt aatcactggt gtcgatattg tcatgaacca 
    tcacctgcag gaaacaagtt tcacaaaaga agcctacaag
    aagtacatca aagattacat gaaatcaatc aaagggaaac
    ttgaagaaca gagaccagaa agagtaaaac cttttatgac
    aggggctgca gaacaaatca agcacatcct tgctaatttc
    aaaaactacc agttctttat tggtgaaaac atgaatccag
    atggcatggt tgctctattg gactaccgtg aggatggtgt
    gaccccatat atgattact ttaaggatgg tttagaaatg
    gaaaaatgtt aacaaatgtg gcaattattt tggatctatc
    acctgtcatc ataactggct tctgcttgtc atccacacaa
    caccaggact taagacaaat gggactgatg tcatcttgag
    ctcttcattt attttgactg tgatttattt ggagtggagg
    cattgttttt aagaaaaaca tgtcatgtag gttgtctaaa
    aataaaatgc atttaaactc atttgagag 
    (GRIK1)(NM_000830.3; mRNA variant 1 of 2 shown) 
    SEQ ID NO: 5
    agagcccctg caccaactca ccctgtaccc tctctccttc
    ttcgttagtc ttctttcccc cttttccctc ctctgtctgt 
    gcctatcccc cgacttttgc atctgaccaa aggacgaatg
    agggagacgt tcctgcagat cggggcagca actttcctca
    gctggtctct gggctccggg agccagagag cgctgatcct
    ccgcggtctg cggcccatgg aagaggagga ggaggagccg
    tgatgggcta gcgacagcac tgaggagccc cgagagagct
    cagccttgcc agccagctcc gcggtcccac gcgggttccc
    tcgagctcgc tccgtgggga gcgcgcagcg tgcttggaac 
    cggagcatcc agagaggatg aggcggggac ccggcccaag
    ttgggtgcat ctctcgggcg tccggcagcg gctgtatctc
    ggcatgaatt aagaagctag gaagatggag cacggcacac
    tcctcgccca gcccgggctc tggaccaggg acaccagctg
    ggcactcctc tatttcctct gctatatcct ccctcagacc
    gccccgcaag tactcaggat cggagggatt tttgaaacag
    tggaaaatga gcctgttaat gttgaagaat tagctttcaa
    gtttgcagtc accagcatta acagaaaccg aaccctgatg
    cctaacacca cattaaccta tgacatccag agaattaacc 
    tttttgatag ttttgaagcc tcgcggagag catgtgacca
    gctggctctt ggtgtggctg ctctctttgg cccttcccat 
    agctcctccg tcagtgctgt gcagtctatt tgcaatgctc
    tcgaagttcc acacatacag acccgctgga aacacccctc
    ggtggacaac aaagatttgt tttacatcaa cctttaccca
    gattatgcag ctatcagcag ggcgatcctg gatctggtcc
    tctattacaa ctggaaaaca gtgacagtgg tgtatgaaga
    cagcacaggt ctaattcgtc tacaagagct catcaaagct
    ccctccagat ataatattaa aatcaaaatc cgccagctgc 
    cctctgggaa taaagatgcc aagcctttac tcaaggagat
    gaagaaaggc aaggagttct atgtgatatt tgattgttca
    catgaaacag ccgctgaaat ccttaagcag attctgttca
    tgggcatgat gaccgagtac tatcactact ttttcacaac
    cctggactta tttgctttgg atctggaact ctataggtac
    agtggcgtaa acatgaccgg gtttcggctg cttaacattg
    acaaccctca cgtgtcatcc atcattgaga agtggtccat
    ggagagactg caggccccac ccaggcccga gactggcctt
    ttggatggca tgatgacaac tgaagcggct ctgatgtacg
    atgctgtgta catggtggcc attgcctcgc accgggcatc
    ccagctgacc gtcagctccc tgcagtgcca tagacataag 
    ccatggcgcc tcggacccag atttatgaac ctgatcaaag
    aggcccggtg ggatggcttg actgggcata tcacctttaa
    taaaaccaat ggcttgagga aggattttga tctggacatt
    attagtctca aagaggaagg aactgaaaag gctgctggcg
    aagtgtctaa acacttgtat aaagtgtgga agaagattgg
    gatttggaat tccaacagtg ggcttaacat gacggacagc
    aacaaagaca agtccagcaa tatcactgat tcattggcca 
    acagaacact cattgtcacc accattctgg aagaacccta
    tgttatgtac aggaaatctg ataagcctct atatggaaat
    gacagatttg aaggatattg cctagacctg ttgaaagaat
    tgtcaaacat cctgggtttc atttatgatg ttaaactagt 
    tcccgatggc aaatatgggg cccagaatga caaaggggag
    tggaacggga tggttaaaga actcatagat cacagggctg
    acctggcagt ggctcctctt accatcacct acgtgcggga
    gaaagtcatt gacttctcca aacccttcat gaccctaggc
    atcagcattc tctaccggaa gcccaatggt accaatccag
    gcgttttctc cttcctcaac cccctgtctc cagatatttg
    gatgtatgtg ctcttagcct gcttgggagt cagctgtgta
    ctctttgtga ttgcaaggtt tacaccctac gagtggtata
    acccccaccc atgcaaccct gactcagacg tggtggaaaa
    caattttact ttactaaata gtttctggtt tggagttgga
    gctctcatgc agcaaggatc agagctgatg cccaaagctc 
    tatcgaccag aatagttgga gggatatggt ggtttttcac
    cctaatcatc atttcatcct acacggccaa tctggctgcc 
    ttcttgacag tagagagaat ggaatccccc atagattcgg
    cagatgatct ggcaaagcaa accaagatag aatatggggc
    ggttagagat ggatcaacaa tgaccttctt caagaaatca
    aaaatctcca cctatgagaa gatgtgggct ttcatgagca
    gcaggcagca gaccgccctg gtaagaaaca gtgatgaggg
    gatccagaga gtgctcacca cagactacgc gctgctgatg
    gagtccacca gcattgagta tgtgacgcag agaaactgca 
    acctcactca gatcgggggc ctcattgact ccaaaggtta
    cggagtggga acacctattg gttctcctta ccgggataaa
    attactattg ctattcttca actccaagaa gaagggaagc
    tgcatatgat gaaagagaag tggtggcgtg ggaatggctg
    ccccgaggaa gacaacaaag aagccagtgc cctgggagtg
    gaaaatattg gaggcatctt cattgttctg gctgccggac
    tggtcctttc tgtatttgta gctattggag aattcatata
    caaatcacgg aagaataatg atattgaaca ggattttgt
    ttatttatg gactgcaatg taagcaaacc catccaacca
    actccacttc tggaactact ttatctacgg atttagaatg
    tggtaaatta attcgagagg agagagggat tcgaaaacag
    tcctcagttc atactgtgta atcagtttaa a 
    (H6PD)(NM_004285)
    SEQ ID NO: 6
    tgaggcctga ggcctggggc ggggtggcgg ccgggctggc
    cttggcctcg cgccttcccc tgcggccgcc gcgggctccg
    cgggcggtat cggagtgtcg tgcggcgcgt ggccgcgtga
    cacgcgcact tgtcggagtg acgggccctg cggaagagga
    ggtgcggccc agggcgcagg ggagccctcg ggagcgggcc
    cggccctcag cgccgccccg gccgtgtccc ggaggagcgg 
    cctgcgccgc cgcgcgagag gaagcaccca ggcatgtgga
    atatgctcat agtggcgatg tgcttggccc ttctgggctg
    cctgcaagcc caggagctcc agggacatgt ctccataatc
    ctgctgggag caactgggga cctggctaag aagtacttat
    ggcagggact gttccagctg tacctggatg aagcggggag
    gggtcacagt tttagcttcc atggagctgc tctgacagcc
    ccaagcagg gtcaagagct catggccaag gccctggaat 
    ccctctcctg ccccaaggac tggcaccca gtcactgtgc
    agagcacaag gatcagttcc tgcagctgag ccagtaccgc
    aactgaaga cggccgagga ctatcaggcc ctgaacaagg
    acatcgaggc acagctccag acgcaggcc tccgggaggc
    tggcaggatc ttctacttct cagtgccacc cttcgcctat
    aagacattg cccgcaacat caacagtagc tgccggccag
    gcccgggcgc ctggctgcgg ttgtccttg agaaaccctt
    tggccatgac cacttctcag cccagcagct ggccacagaa
    tcgggacct ttttccagga ggaggagatg taccgggtgg 
    accattactt aggcaagcag ctgtggcgc agatcctgcc
    tttccgagac cagaaccgca aggctttgga cggcctctgg
    accggcacc atgtggagcg ggtggagatc atcatgaaag
    agaccgtgga tgctgaaggc gcaccagct tctatgagga
    gtacggtgtc attcgcgacg tcctccagaa ccatctgacg
    aggtcctca ccctcgtggc catggagctg ccccacaatg
    tcagcagtgc ggaggctgtg ctgcggcaca agcttcaggt 
    cttccaggcg ctgcggggcc tgcagagggg cagtgccgtc
    tgggccagt accagtctta cagtgagcag gtgcgcagag
    agctgcagaa gccagacagc tccacagcc tgacgccgac
    cttcgcagcc gtcctagtgc acattgacaa ccttcgctgg
    agggcgtgc ctttcatcct gatgtctggc aaagccttgg
    acgagagagt gggctacgct ggatcttgt tcaagaacca
    ggcctgctgt gtgcagagcg aaaagcactg ggccgcggcg 
    agagccagt gcctgccccg gcagctcgtc ttccacatcg
    gccatggcga cctgggcagc ctgccgtgc tggtcagcag
    gaacctgttc aggccctccc tgccctccag ctggaaggaa
    tggagggac cacctgggct ccgccttttc ggcagccctc
    tgtccgatta ctacgcctac gccctgtgc gggagcggga
    cgcccactcc gtcctcttat cccatatctt ccatggccgg
    agaatttct tcatcaccac agagaacttg ctggcctcct
    ggaacttctg gacccctctg tggagagcc tggcccataa
    ggccccacgc ctctaccctg gaggagctga
    gaatggccgtctgaggact ttgagttcag tagcggccgg
    ttgactat cccagcagca gccggagcagctggtgccag
    ggccagggcc ggccccaatg cccagtgact tccaggtcct
    cagggccaag taccgagaga gcccgctggt ctccgcctgg
    tccgaggagc tgatctctaa gctggctaat gacatcgagg 
    ccaccgctgt gcgagccgtg cggcgctttg gccagttcca
    cctggcactg tcggggggct cgagccccgt ggccctgttc
    cagcagctgg ccacggcgca ctatggcttc ccctgggccc
    acacgcacct gtggctggtt gacgagcgct gcgtcccact
    ctcagacccg gagtccaact tccagggcct gcaggcccac
    ctgctgcagc acgtccggat cccctactac aacatccacc
    ccatgcctgt gcacctgcag cagcggctct gcgccgagga 
    ggaccagggc gcccagatct atgccaggga gatctcagcc
    ctggtggcca acagcagctt cgacctggtg ctgctgggca
    tgggtgccga cgggcacaca gcctccctct tcccacagtc
    acccactggc ctggatggcg agcagctggt cgtgctgacc
    acgagcccct cccagccaca ccgccgcatg agccttagcc
    tgcctctcat caaccgcgcc aagaaggtgg cagtcctggt
    catgggcagg atgaagcgtg agatcaccac gctggtgagc 
    cgggtgggcc atgagcccaa gaagtggccc atctcgggtg
    tcctgccgca ctccggccag ctggtgtggt acatggacta
    cgacgccttc ctgggatgag ggcgcctgtg ccccttgccc
    gcttcgctcc tgtgctttcc ttcgcccgtg tcttccctcc
    cttctcggcc ccgccacctg cccagcgtgc cctggctctc
    cagaaccttc tatcccacag tcaggcccca gagagggcag
    gacaagcctt gtcccgatgc ctttgaccgg cagctctgtg 
    tattggtgga tagatgcaga aacaaggaag aaatggagtc
    tgctcctgag aagcttcaaa ttcaggccag gagagaagtc
    ttaagaaaag acctccagca gttacacatt catatcaacc
    agcacaacac gggatggcgc ccaaactccg gcgttcacaa
    gaggagacgt gacgtggtgg gctgaggtta atcagggaag
    gtttcctggg ggaggtgatc cttgaactgg ctcccgggga
    acattcagag catgattggt agacagaagg gtgcagaggc 
    gcccagggga gtacattgcc ccgtgcaaag caggggcatt
    ggggactgtc ttgagaccct gagggggtca agcccctcct
    tccccagctg cccctccttc tagaacctct gcacatctag
    cctctggccc tcctcttcac tgcctccacc tgctcccgct
    tgccatccct gtctcctcca tcctggctgt gcagtaggaa
    ttccaggctc ctccctgtgt ctttgctgtt cttcagactc
    catttataga gaatgagggc tgataacagg aatacagtgg
    caaagactag actgtggaaa gggttccaga aatctttttt
    cttttttaat taaaaaaaat atttgcagag atgagctctt
    gctatgttgc ccaggctggt ctcaaactcc tgggctcaag
    cgatcctccc atctcagcct cccagagtgc tgggattaca
    ggtgtgagct actgcgccca gccccagaaa tctcagtgct
    gtttggagct ccatttctca tttgatgact tgctctgcgt 
    ggggaggtgg ggtctcattc ccccaacttc ctcagggagg
    acccctgccc tccgctgctc ctctgtcctg ctagccttcc
    tccaggaagc acactgggtg cagataatca ggacattcca
    gagatcccca atttaagagg gtcatttcca tctcagggga
    ctcccggatg ggtgtttccg ctctcaatag cccctcttgt
    tttaccagga aagatccagt taaatcaccc actgaggtga
    cagctcatta gcggggagag agatggagca tcgagtgaca
    ctgggccatc caggcggctc tgctcccacc agacaggagc
    taggcctcac tggcaggggg gctgcccaca gccttttcag 
    gggctcgctt ggcgggtgac ggggccgcag ccaggccttc
    tctccctgcc ccttggtgac cccgtggctt cctgtctgct
    ggcctctcct gctacttatc acttcaccac gaactctctg
    cctgagactg gggaagtaag cgggtatctt ctcagtgagc
    ataggttggg gactgtgatc ttgagaagcc atgggccagc
    aatacctgct tttctgaagc ccccaaggag ggctctgaca
    ttctttttaa aaacaccaca aagcaaaatt cccaggacat
    gtgtagtttt gtttgttcag tatcccacaa cttaaggctg
    ggagatggaa ctcttggtta aggtcgattt ttctgtctgg
    cttctccgca ccttccactt gctctctgga tcaggcagat
    ataaactttc tagcgcattt tgagagaggg ctttcttggg
    tgagggagca tggcaaagtc ggtttctctc tggactgttt
    acacttcaag gcggtggatt tagaggaatc ctggattca
    ttttcaatgc cagtctgaga catgttccca agccggggct
    cttgttcaca ccacttactc tggccaccaa caacaaccca 
    ggccagacag agcatctat tttttttttt ttgagacaga
    gtctctgtcg cccaggctgg agcccagtgg cgagatcttg 
    gctcactaca acctccacct cccgggttca ggcaattctc
    gtgcctaagc ctcccgagta gctgcgacta caggcgccgg
    ccagcatgcc tgtctaattt ttgtatttta gtagagacag
    ggtttcacca tgttgcccag gctggtctcg aactcctgag
    ctcaggcagt ctacccacct cagcctccca aagtgctggg
    attacaggcg tgagccaccg cgcccagcca gaacatctgt
    ttttacaccc agagagcgcc cctcgttagg acagaaccac
    ggtgcccaga gccaggaagc cgccctcctg gcgcccagca
    tctgagcttc tacacgtgat gggcgggctc aggagaggac 
    agggagtcgt ggtggaagtt ccacagctgg ccgcgtgggg
    gggcccttgc accgcactgc cgcctcctga ctgcccctat
    ccccgcagcc cctgtgccgg atttcatttc cctcctctct
    cccagggtac ctggccccag cactctccca tctgttcttc
    aggaaccgac tcctctccag ttgcaacacc agggagaaag
    gggcctccac atgcccaagt acccctgcag gatgaagggc
    aggccggccc ttgatgtgcc atttctgaat aatagtcact 
    gccgccgagt ctaggatgtc ctgttctaac tcagccctgc
    ctcggatgca ccaccgatct gtgcagagtg ggtgtgggag
    tgtgggtgag ggtcgaaatg ccaaaggtct actttccaga
    atcaagtgcc ttctgcaaat catgttggaa aagtccaaac
    ctggagatgt ccctgtgcct ccgcccctac ccaccccttt
    tccttcagct gtgttaggaa ggagaagttt tcagaaccct
    ctaggctggt ggctttcaaa cttcagacca gatctgcag 
    caagaaacgt gccttccatc ataaatcagt ccatttgttt
    acaactgtgt ccaagcagg tttcataaag aaattcttaa 
    ccttagaacc tcggatatcc tctatgtttt agttttcatt
    tttttaaaat gcttcttaaa attcactaaa ttgggctagg 
    tgtggctcat gcctgtaatc ccagcactat gggaggctga
    ggtgagagga tcacttgagc ccagaaggtt gaaaccagcc
    tgggcaacat agtgagaccc catctctaca aaaagtttta
    aaaccaggta tggtggtgcc ctcctgtggt cccagctact
    cgggagtctg aggtgggagg atcacctgag cccaggagac
    tgaggctgca gtaaggtgtg attgcactat tgctctctag
    caggaaaac agagtgagac cctatctcaa aaaaaaaaaa 
    aaaaaaaaag gaaagagtga tgacaacagc ccagggagca
    gccccgctca gaacccaagt cccaagttcc agcactgtgt
    tcccaggcag gctgtttgcc tcttcctggt ctggaagccc
    ttgggtccta tggtggcggc agctcccaca tccaggttc
    cctggtgggg accaatgatt ccatccgcat ggaagcccac
    gtgtgcactt aggggcccat aaatggcaga agggcccctc
    ctttgggaga ccttgtcagt cagcatctct agggcaaccg 
    tgattgccat ttgtagaggg gaaggaatca agggacttta
    agctagatca aaatctgggg acaaattctc ctgctaactg
    caagttaaaa taggcccttc ttactgaatt tccctgtttg
    tttctctgca gacaatgctt tagccctact cttgggcccc
    caagttagca gagtaatcaa agcttcctac cgtttggcct
    actattccag actagtccct cgaggggttc ccttccaaaa
    tatgcagggc tcaggctccc aattccgggc ctgtctgctt
    tgcttgtgtt tctcctgtcc ctgttctccc ggagggccca
    ggtggaactc acgacaggga gggagacgct tcccaaaaac
    ctgcagggct atttcccaga atttggtttt caagtacaaa
    actttttgtc ctgtaagata tatgcagcct cacagaagca
    gcctctgcct ccactttacc agctacgttt ttatcttaag
    cacatggggc tcccttagaa cttactccac tgatttaaaa
    aaaaaaaact gcctggcagc atctcagtgt cagagtgagc
    acggcacagg aaaggcccgt ggtgacgagg gtgaggtggc 
    cacagtgacc ggacgacaaa tgagactctg caaatgagac
    tccagagggt gaagatctgc ggtctccaga catcataggc
    catgtgaccc actaggggcc gcttacccct ggccgtccgc
    tggctgaact gaacgcattc cctctctccg caactctccc
    gtgaggctgc acccgtgtgg gtagcactgg aagcggcact
    gtttgcattg tacataggaa ggaaggaagt tatccagcc
    tcaccagcac ctggcagcga gtcagagcct gtgagggcat 
    ccgaagcagt gatgcagtgt caacctccca gctggtgcca
    ctctgccctc gggggctcca agcattgtaa ctcagtcatg
    ggagctgcct ctttggaagt gcagatttat tcctgtaata
    atcctgcctg cttttacctc tcgtccactg accagcaagt
    gtgagtcccg gtgtcagtcg gcacagtcca gtgtccatct
    gcatttgctc atgcagaggg ggtgagttgg gcactccctg
    ttgaggttt tccttttgca gcacactggg cagtctccct
    ataaaacaaa aaccccacct tctgtgcctt ctgctttaga
    gcagagctcc ccctcccatt tcctcagtct tccctgcaaa
    atctgtccac cggggaaggc agcaggaacc ctgggcagcg
    ggtgttctgg gaaggctagt gacagcagat gtcatccagg
    aacagccaca cacggttctc caggccgccg tcagcagctc
    aaggtggggt atgagtgaga agctgaggat ctcgcagctt 
    gttgctgagc aaggtgcaac cgggctcatg ctgtcatcag
    cacaagacgg gatggcaagg gctttcagac gcatttccaa
    gagtccagca agccaggggg aagatgatcc ctttgccgaa
    gtgtaccctc tagccaactt ttgggagcgc ttctgtttgc
    aaagcgctgg ggatgtgcct gtctctgtgt gacccacgaa
    cgggaaggga gagcactgga gtaatgacac ttctgctgct
    gctttgattc tcaaggctga tattaaaac cctcgccttg
    ctgacaggtg ctttaaaggc agtctgcatc ttttatccc
    ttggtgtggg agaggtaaac actttgattt gctgaaagct
    gtatggagta tatttgaaca gctagtagtt agctttgaaa
    gtggaagtgt gaacagacac tacttgtgtc gctttgggtc
    cttcacttta cccccacaga agtctagagg cgtctgttat
    aaagcgttac ggggcgcctg catgcaggag gaaggacctg 
    tattagctgg aaatcatcag gaacccagct tgcctccatc
    tctctgagat gtgctgggta cagcctgccc ctcctagttc 
    tgtccaccgg gaagagccgg ctggcggcag atccccaggg
    gcagagcccc tgctggatcc tgggagctca tctttacctg
    tgccggagtg ggaactgtga ttccagccgg gcaggtcaga
    gtggagcagt gctaagaggc tgttgcagga gaactagacg
    ggcggggcct gctgcatctg gatcatgttt ctgtgctctg
    ccccgcgcta gggactcagg gtctgggctt ctgccaggtg
    aggagcagag agactgttcc cttgggtgga gaggtgtggg 
    catgagagcc acccattgcc aagcagcaag aatgttcgtg
    cttttttcca gagaggggaa ccccactggt ttttgtggaa
    acaatggaaa cttacagatg cctgcctggg atgatgaggc
    acattcagaa caaatgcttt tttttttttg agacagagtc
    tcgctctgac gcccaggctg gagtgcagtg gcgcgatctc
    ggctcactgc aaactttgcc tcccaggttc aagtgattct
    cctacctcag ccttccgagt agagggatt acaccaccat
    gcccagcaaa tttngtgtt tttagtagag acggagtttc
    accatgttgg ccaggctggt ctcgaactcc tgacctcagg
    tgatccatcc gccttggcct cccaaagtgc tgggattaca
    ggcgggagcc accatgcctg gccagaacaa atgccttttt 
    aaacctttta agaacatttt taaaatgtct ttttctatgt
    caaatgtaac gtttattttt ttaaacaata aaattgattt 
    gccaaaa 
    (IGF2BP1)(NM_001160423.1 version 1 of two mRNA
    speies)
    SEQ ID NO: 7
    atttagaggc ggcgccaggg cggccgcgga gaaacgtgac
    acaccagccc tctcggaggg gtttcggacc gaagggaaga
    agctgcgccg tgtcgtccgt ctccctgcgc gccgcgggca
    cttctcctgg gctctccccg aactctcccg cgacctctgc
    gcgccctcag gccgccttcc ccgccctggg ctcgggacaa 
    cttctggggt ggggtgcaaa gaaagtttgc ggctcctgcc
    gccggcctct ccgcctcttg gcctaggagg ctcgccgccc
    gcgcccgctc gttcggcctt gcccgggacc gcgtcctgcc
    ccgagaccgc caccatgaac aagattaca tcggcaacct
    caacgagagc gtgacccccg cggacttgga gaaagtgttt
    gcggagcaca agatctccta cagcggccag ttcttggtca
    aatccggcta cgccttcgtg gactgcccgg acgagcactg 
    ggcgatgaag gccatcgaaa ctttctccgg gaaagtagaa
    ttacaaggaa aacgcttaga gattgaacat tcggtgccca
    aaaaacaaag gagccggaaa attcaaatcc gaaatattcc
    accccagctc cgatgggaag tactggacag cctgctggct
    cagtatggta cagtagagaa ctgtgagcaa gtgaacaccg
    agagtgagac ggcagtggtg aatgtcacct attccaaccg
    ggagcagacc aggcaggctg acgaggttcc cctgaagatc 
    ctggcccata ataactttgt agggcgtctc attggcaagg
    aaggacggaa cctgaagaag gtagagcaag ataccgagac
    aaaaatcacc atctcctcgt tgcaagacct taccctttac
    aaccctgaga ggaccatcac tgtgaagggg gccatcgaga
    attgagcag ggccgagcag gaaataatga agaaagttcg
    ggaggcctat gagaatgatg tggctgccat gagcctgcag
    tctcacctga tccctggcct gaacctggct gctgtaggtc 
    ttttcccagc ttcatccagc gcagtcccgc cgcctcccag
    cagcgttact ggggctgctc cctatagctc ctttatgcag
    gctcccgagc aggagatggt gcaggtgttt atccccgccc
    aggcagtggg cgccatcatc ggcaagaagg ggcagcacat
    caaacagctc tcccggtttg ccagcgcctc catcaagatt
    gcaccacccg aaacacctga ctccaaagtt cgtatggtta
    tcatcactgg accgccagag gcccaattca aggctcaggg 
    aagaatctat ggcaaactca aggaggagaa cttctttggt
    cccaaggagg aagtgaagct ggagacccac atacgtgtgc
    cagcatcagc agctggccgg gtcattggca aaggtggaaa
    aacggtgaac gagttgcaga atttgacggc agctgaggtg
    gtagtaccaa gagaccagac ccctgatgag aacgaccagg
    tcatcgtgaa aatcatcgga catttctatg ccagtcagat
    ggctcaacgg aagatccgag acatcctggc ccaggttaag 
    cagcagcatc agaagggaca gagtaaccag gcccaggcac
    ggaggaagtg accagcccct ccctgtccct tcgagtccag
    gacaacaacg ggcagaaatc gagagtgtgc tctccccggc
    aggcctgaga atgagtggga atccgggaca cctgggccgg
    gctgtagatc aggtttgccc acttgattga gaaagatgtt
    ccagtgagga accctgatct ctcagcccca aacacccacc
    caattggccc aacactgtct gcccctcggg gtgtcagaaa 
    ttctagcgca aggcactttt aaacgtggat tgtttaaaga
    agctctccag gccccaccaa gagggtggat cacacctcag
    tgggaagaaa aataaaattt ccttcaggtt ttaaaaacat
    gcagagaggt gttttaatca gccttaaagg atggttcatt
    tcttgacctt aatgtttttc caatcttctt ccccctactt
    gggtaattga ttaaaatacc tccatttacg gcctattct
    atatttacac taattttttt atctttattg ctaccagaaa
    aaaatgcgaa cgaatgcatt gattgctta cagtattgac
    tcaagggaaa agaactgtca gtatctgtag attaattcca
    atcactccct aaccaatagg tacaatacgg aatgaagaag
    aggggaaaat ggggagaaag atggttaaaa tacataataa
    tccacgttta aaaggagcgc acttgtggct gatctatgcc
    agatcaccat cttcaaattg gcacaactga aatttcccca 
    ctctgttggg gcttccccac cacattcatg tccctctccc
    gtgtaggttt cacattatgt ccaggtgcac ataggtggta
    ttgaatgctc agcagggtag gggctgacca ctgtccctga
    ttcccatcgt tctcaggcgg attttatatt tttttaaagt 
    ctattttaat gattggatat gagcactggg aaggggacgc
    taactcccct tgataaagtc tcggttccat ggaggacttg
    agtggcccca aaggctgcca cggtgccctc accccagccc
    atgtgctccc ataagggctg gttcctagag gcaggggttg
    tggggcactc ccagccacgg cactgttacc ttggtggtgg
    gacttggaac ccaaccctga gctcccgata aagctaaagt
    ccatcatctg gcaaattcag taaattggag agtacttgct
    tctgtttgta tctgagagga atttttaact gacggcttct
    gtctccatga atcattatca gcatgatgaa aggtgtgtct
    aaaaaacaat tcagaatacc agcagcattg tacagcaagg
    ggtaaataag cttaatttat taatttacca ggcttaatta
    agatcccatg gagtgtttag cccttgtggg agacagaagc
    catcagttaa atgaggttag gcctctcctc ctaatatact
    gattgacaat gcatattagc caggtaatgc actttagcta
    ccctggacaa tgctatcaag tgtgctggga agggaggaag 
    gcctctctac atatggaaaa gcccatgcgt ggagttcccc
    tcctttcaac attgcaacaa cagtaacaac aagacaaccg
    caacatgtgg gcgtagtcag gcaatgctgt gtgcgaagta
    aactacctca aggtatgaag ttacctcagc aattattttc
    ctttttgttc cccccaaccc cattaaaaaa attttttttt
    gatttttgtt tttttgcagc ttgctgatat tttatataaa
    aaagaaaagc aaagcaaaag agaagctgat agtcttgaat
    attttatttt tttaatgaaa agaaaaaaca agaaagttat
    gtttcataat ttcttacaac atgagccagt aaccctttag
    gaactctcta tggagaacag gcctggtggg aaaggctttg
    ggggctgccc ccttaggagg aggctagtgc taagagggaa
    ggcccaggtt tgagagagcc cagaggggca gagcccagag
    ccttgtttgg ccctgatctc tgacttctag agccccagct 
    gctggcggct gaggaatat cctacctgat aggattaaaa
    ggcctagtgg agctgggggc tctcagtggt taaacaatgc
    ccaacaacca accagctggc cttggtctc ctctctttcc
    tcctttggtt aaagagcatc tcagccagct tttcccacca
    gtggtgctgt tgagatattt taaaatattg cctccgtttt
    atcgaggaga gaaataataa ctaaaaaata taccattaa
    aaaaacctat atttctctgt ctaaaaatat gggagctgag
    attccgttcg tggaaaaaag acaaggccac cctctcgccc
    tcagagaggt ccacctggtt tgtcattgca atgcttttca
    tttttttttt ttgttattgt ttcatttcag ttccgtcttg
    tattatcc taatctatat ccatagatct aaggggcaaa
    cagatactag ttaactgccc cacctctgt ctccctgtct
    tctttagatc ggtctgattg attttaaaag tggacccaaa
    ttagggaat tcttgattta gggtggctgg tggcaaggag
    gggcagggga tatggggacg tgactgggac aggttcctgc
    cttatcattt tctccctagg acattccctt gtagccccca
    gaattgtctg gcccaaattg aatagaagca gaaaaacatt
    tagggataac atcaggccag tagaattaag cctctccacc
    tgtcccaacc ataaaaaggg tctcccagct ttccatctct
    ggctctatat gctttatccc aaaacaaagc agataacgtt
    cagacgtcgg ccatttagta atttaaagcg aatttccagc 
    agcaagcatg ctttgatatc tggttcagac tatcatcagg
    aagaaaaaaa aatcccacag tacctgaaat gtgattgttg
    cagtgttcag tttccttggg ggcctgctcc cttcacacct
    tgagcccaag tccttttccg ttggctgatt cagctcccag
    aagagacgag gaagtgtgtg gcaagggact ggaaaacttc
    acttgcttgg attaggcaag gctccactca ttgttgatat
    ttgcccagca ggaaaatcat gtaagttata ccaccagaaa
    gcaaaaggag catggtttgg tggttaaggt ttagtgggat
    gaaggacctg tcttggtggg cogggccctc ttgtgccccg 
    taggctaggt cttagggcaa ctccttgccc tcctgctcag
    cacctccatt tccccatcct tggtgagata acaagctatc 
    gcgaaaagca cttgggagat ttggatgatt tgagaagagt
    gacttaaaaa aaatgcttct gtgctctaag atatatatgt 
    gtgtgtgtgt gctacatata tattataag aaaggaccat
    ctattagga tatattata aattattga aacacataac 
    caaaatggtt tgattcactg actgactttg aagctgcatc
    tgccagttac accccaaatg gattaatcc cctctcgggt 
    ctggttgcct tttgcagttt gggttgtgga ctcagctcct
    gtgaggggtc tggttaggag agagccattt ttaaggacag 
    ggagttttat agcccttttc tactttcctc ccctcctccc
    agtccttatc aatctttttt cctttttcct gaccccctcc 
    ttctggaggc agttgggagc tatccttgtt tatgcctcac
    tattggcaga aaagacccca tttaaaaccc agagaacact
    ggagggggat gctctagttg gttctgtgtc cattttcctc
    tgtgccaaag acagacagac agaggctgag agaggctgtt
    cctgaatcaa agcaatagcc agctttcgac acatacctgg
    ctgtctgagg aggaaggcct cctggaaact gggagctaag
    ggcgaggccc ttcccttcag aggctcctgg gggattaggg 
    tgtggtgttt gccaagccaa ggggtaggga gccgagaaat
    tggtctgtcg gctcctggtt gcactttggg gaaggagagg
    aagtttgggg ctccaggtag ctccctgttg tgggactgct
    ctgtcccctg cccctactgc agagatagca ctgccgagtt
    cccttcaggc ctggcagacg ggcagtgagg aggggcctca
    gttagctctc aagggtgcct tcccctcctc ccaacccaga
    cataccctct gccaaactgg gaaccagcag tgctagtaac 
    tacctcacag agccccagag ggcctgcttg agccttcttg
    ctccacagga gaagctggtg cctctaggca accccttcct
    cccacctctc atcaggggtg ggggttctcc tttctttccc
    ctgaagtgtt tatggggaga tcctagtggc tttgccattc
    aaaccactcg actgtttgcc tgtttcttga aaaccagtag
    aagggaaaca gcacagcctg tcacagtaat tgcaggaaga
    ttgaagaaaa atcctcatca atgccagggg acataaaagc
    catttccctt ccaaatactc gacaatttag atgcagaaca
    tttctctgta ttcagactta gagtaacacc agctgaaaac
    tgcagtttct ttcattgga tacataaggc ttctctatcg
    gggtacggga cagggaggag gcctcatgtc tgaaggggga
    ttaggggcg agagccccag ccctgaccct cggtcctgtg
    caccgctttg gggcacagtc tgatggcgcc tttgctggcg 
    ccttagtatg gttgactccg gatggacaaa agaaaaaaaa
    ttttttttct tgaatgaaat agcaggaagc tcctcgggag
    catgtgatt gattaaccgc aggtgatgga tgctacgagt
    ataaatggat taactacctc aatccttaca gtaagattgg
    aactaagggc agggactcat gcataagggt atgaatccca
    gccaggacaa gtgagttgag gcttgtgcca caaaaggttt
    gtccttgggg aacaggcagg cctgccagga tcccccccat
    atcgattggg ctgggagggc tggccatgag gtccccactt
    tctgctttcc ttgcccatgt gtcacccctt tggcctccag
    cttgtccctc tctcactttc tatagctttg ttggaccaga
    tggtgaggaa aggaatggcc tcttcccttc tagagggggc 
    tggctggagt gagacctggg gcttggcctg gaacccacca
    cacagcccca aagtcaggaa gcctggggaa accagagctg
    agacctcttc aacagggttt ctttgagatc ctacacctcc
    attgggccct ttttcagtct tcaatggggg cccagttggc
    tctagaagga gaagaggtga agcaggatcc tttgccctgg
    gggagtctga gggcgcggtc cttggactca ttcaggccgt
    ctttgtagtt gggggagttc cactgggcga tcccagcccc
    tccccaccca ccctctaatg gacctcctca tagaagcccc
    atttcacttt tgttttatct acctcttagc aaaacaatag
    ataaattagg tagtggcagc tccacttgct taggttaggg
    ggggaaaaag atttcttttt ccaaaggaaa aaaatattac
    cttgagaata ctttccaaaa aataaaatta aaaaaaaaaa
    aaccaaaaaa aaaaattttt ttttaaaagg gagacatttt
    ccagtgacca ctggattgtt ttaatttccc aagctttttt
    ttcccccata aataagtttc actctttggc gattttcttc
    acttgtttaa gataacgtgc tagctattcc aacaggtaac
    agctttcaca gtctgcccct ggcctgtctc accccatccc
    ccaccctatt cctgccagtg agtccttcct gtgcttact
    cccttaccc ctcccagcca gctgacttca gtcacccctg
    tcccccctcc cctgccaata agctccccca ggaataaagg
    ctttgttttg gggatgctta aatcttgact ggcacttccc 
    ggctgtgggg gctggggagc cacttgtaac atttctgtgc
    agattttatg ttagccactg ctatgtaaaa gcacgttcaa 
    aatgaatttc agcagattat gtgttaccat aatgaataaa
    cgtcctctat caccatttgg agtctccctt ttctccagga 
    tcttgatcct ggtccccaaa accagagtga atcaaaagag
    cttcctcccc tgaggcaaag tggatttgta agcagttctg
    aaacatcact tactcagaag agggaacgat gtattttgat
    gagtgcaaat tgggaagagc tggaggccta ctgcttggga
    cagttttttt tttttttttt tttttaaata tgagtgctag
    cttattctgt aattgcggca actttgaaaa ttgtatttta
    ctggaaatct gccagccatc accacccgat tttgattgta
    tccttcctcc catcctttaa tctgttcatt gctttggggg
    aggtggggca gctggctcac acgttggagt ttgttctttg
    atggatgaac gaacactcca gttttctttc ccgtgaaggt
    tgtttcagcc acaaaccact tcattttgct gtttcaattt
    caaaataaaa ggaaacttat attgaaagac aa 
    (MDM4)(NM_002393; protein is NP_002384.1) 
    SEQ ID NO: 8
    gggaggccgg aagttgcggc ttcattactc gccatttcaa
    aatgctgccg aggccctagg atctgtgact gccacccctc
    cccccacccg ggctcggcgg gggagcgact catggagctg
    ccgtaagttt taccaacaga ctgcagtttc ttcactacca
    aaatgacatc attttccacc tctgctcagt gttcaacatc
    tgacagtgct tgcaggatct ctcctggaca aatcaatcag
    gtacgaccaa aactgccgct tttgaagatt ttgcatgcag
    caggtgcgca aggtgaaatg ttcactgtta aagaggtcat
    gcactattta ggtcagtaca taatggtgaa gcaactttat
    gatcagcagg agcagcatat ggtatattgt ggtggagatc
    ttttgggaga actactggga cgtcagagct tctccgtgaa 
    agacccaagc cctctctatg atatgctaag aaagaatctt
    gtcactttag ccactgctac tacagatgct gctcagactc 
    tcgctctcgc acaggatcac agtatggata ttccaagtca
    agaccaactg aagcaaagtg cagaggaaag ttccacttcc
    agaaaaagaa ctacagaaga cgatatcccc acactgccta
    cctcagagca taaatgcata cattctagag aagatgaaga
    cttaattgaa aatttagccc aagatgaaac atctaggctg
    gaccttggat ttgaggagtg ggatgtagct ggcctgcctt
    ggtggttat aggaaacttg agaagcaact atacacctag 
    aagtaatggc tcaactgatt tacagacaaa tcaggatgtg
    ggtactgcca ttgtttcaga tactacagat gacttgtggt 
    attgaatga gtcagtatca gagcagttag gtgaggaat
    aaaagttgaa gctgctgata ctgaacaaac aagtgaagaa
    gtagggaaag taagtgacaa aaaggtgatt gaagtgggaa
    aaaatgatga cctggaggac tctaagtcct taagtgatga
    taccgatgta gaggttacct ctgaggatga gtggcagtgt
    actgaatgca agaaatttaa ctctccaagc aagaggtact
    gttttcgttg ttgggccttg aggaaggatt ggtattcaga
    ttgttcaaag ttaacccatt ctctctccac gtctgatatc
    actgccatac ctgaaaagga aaatgaagga aatgatgtcc
    ctgattgtcg aagaaccatt tcggctcctg tcgttagacc
    taaagatgcg tatataaaga aagaaaactc caaacttttt
    gatccctgca actcagtgga attcttggat ttggctcaca
    gttctgaaag ccaagagacc atctcaagca tgggagaaca 
    gttagataac ctttctgaac agagaacaga tacagaaaac
    atggaggatt gccagaatct cttgaagcca tgtagcttat
    gtgagaaaag accacgagac gggaacatta ttcatggaag
    gacgggccat cttgtcactt gttttcactg tgccagaaga
    ctaaagaagg ctggggcttc atgccctatt tgcaagaaag
    agattcagct ggttattaag gtttttatag cataatggta
    gtacgaacat aaaaatgcat ttattccgtt cacttaccac
    attatttgaa aatcaatcct ttatttaatt ttatttccaa
    cctgtcagag aatgttctta ggcatcaaaa tccaaggtag
    ctgtaagaaa aatactggag ctaacaatga agaacagaag
    taatctgatt agtcaaatta ttaagtgcca tggattactt
    tatgcagcag tcaggtacat agttaggtga acccaaaaga
    aaaactcttg aaaacaagag atttcttcca tgcacattta
    caatattgag gtataattaa catgataaag tgtttccttc
    taacgagttg tagaaatctg agtaaccacc caaaaaagca
    atagaatgtt tctgtcaccc caaaacactc ccttctgccc
    ctcttcagac agtccttcag ctatttcatg gctctcaccc
    tagttattt tttttttgca cttttttttt tccgggggta
    taggggaggt gtggggcgac agggtctgtc ttgttctgtc
    tcccaggctg aagtgcagtg cagtggtatg atcatggctc
    actgcagcct tggtttcctg ggcataagtg gtatcccac
    ttcagcctcc tgagtagctg agactataga ctagcataac
    cacactggct aattttttgt ggagatgaag tctcactatg
    ttgcccaggc tggtctcgaa ctcctgggct caaacaatcc
    tcccgcctca gccttccaaa ttgctgggat tatagtcatg 
    aggcacctag tctggccctt ttgcaagact ttaatctgaa
    atctaaattt ttaaaattta agtacttaca aaggatatac 
    tatccaacat attgcatatt atatatgtgc tttaaagttt
    tttttttttt ttgagagacg gtctcacttt gtcatccaag 
    ctggagtgca gtggtgcaaa cacggcccac ctcctgggct
    caagtgatcc tccagcctca gatccctca caggcattca
    ctatcactcc cagctaatta aaataatttg tagacggtgt
    ctcgttatgt tgcccaggct ggtctcgaac tcctgggttt
    aagtgattcc cccgcctcag cctcccaaag tgttgggctt
    acagccttga gccactatgc ttggctcaaa gatattttta
    tgaaagccct gggactatag atttagctga ttaaatttat
    agaaaaagtc ctgtcatata aactggcaaa gtctgttctt
    aatttaatta gccaaatcag acttaacttc cgtcagaaca
    tgtcttggtt ttaattcaga taaacacaca aacatacttc
    tctggcacag ccttcagaag catcagtttt tgttttgttt
    tgttttgat tttgagacag ggtcttgctc tgtcgcccag
    gctggagtgc actggcacaa tcacagttca ctgcagcctc
    gacctcccag atccaagcaa tcctcccacc taagcctccc
    aagtagctgg gtctataggc gcgtgccacc accatgccca
    gctgaatttt gtattttttg tacagacagc attttgccat
    gttgcccagg ctggtcccaa acttctagcc tcaagcaacc
    ctcctgcctc agcctctcaa agtgctagga ttgcagtcct
    gagctactgc cccctaccct ctttgcgtct taggagtcat
    ttagattttt tttgatcctt ttgtttagtg cctctggagc
    tgcttacacc aaggcaatac gccttgatat actggatggt
    tgagaggcag cctctttttt tttttttttt tttattttt
    tttggaggat agggagtatg gctgttgtga aaagggaggt
    aaagagaaat ggtagatctg aagaggcctc atcagagcac
    atattttagg acaacacata tggaaattgg acatctttaa
    gttggtttcc atagagctat gcatgtatcc ttacccccat
    gggaaaatgt tggtgtgttc tcaagggtat gcatgtgtca
    ttttgaagac caaggcccta gaattgtcaa acttaaggat
    cataaaaatc atgagggttg cttgttaaaa atgtccaaac 
    gtgcagagac tgatattga gatctggacc aggaatttgc
    atttgaacaa gtgttcctgg aatctctatg caagttttat
    acagaacata cattggaat ccttgcccta gacaggggtg
    tccaatcat tggcttccct ggtccacaat ggaagaagaa
    ttgtcttgga ccacacataa aatacactaa cactaacaat
    agctgatgag ctaaaaaaaa aaaaaaaaaa aatcgtggac
    cgggcgtagt ggctcacgcc tgtaatccca acactttggg
    agatcaccta ggtcgggagt ttgagaccag cctgaccgac
    atggagaaac cccattttta ctaaaaatac aaaaaattag 
    ctgggcatgg tggtgcatgc ctgtagtccc agctactcag
    gaggctgagg caggagaatc gcttgaacct gagaggggga
    gattgcggtg agctgagatt gcgccattgc accccagcct
    gggcaacaat agcgaaactg tctcagaaaa aagaaaaaaa
    aaatcgcaaa aagaaaaatc tcataatgtc gttgttggtt
    tttttttttt tttttgagac agtctcactc tgttgcccag
    gctggagtgc aatggcatga tctctgctca ccgcaacctc
    tgcctcccgg gttcaggtga ttctcctgcc tcagcctccc
    agatagctgg gactacaggc acataccacc atgcctggct
    aatttttgta tttttagtag agatgggggt ttcactgtgt
    tggccaggct ggtctcgaac tcctgacctc atgatccaca
    cacctcggcc tcccaaagtc ctgcgattac aggcgtgagc
    taccgcaccc agccaagttg taatttttaa taaaacttaa 
    gaagtaaaca ttttacttat gtttataggt atttgatcct
    aaatttgaca catcattgcc catgaaagaa tcctcttagg 
    ctgctcagct tcactcttcc tgcttgccca ccggggtttt
    tcactgcttc tgttagcact aagtacttag acgatcctaa 
    gatatgtgct tgagccgaat ttcatcttta cttgtaggaa
    actttaaact atttcttttc ttttcttttt tttttttttt
    tacttgagat ggagttttgc tcttgtcgcc caggctggag
    tgcagtggag tgatctcggc tcactgcaac ctctgcctcc 
    cgggttcaaa tgattctcct gcctcagcct cccaagtagc
    tgggattaca ggtgtgcacc accatgtctg gctaattttg 
    tatttttagt agagatggtt tcaccatgtt ggtcaggctg
    gtctcgaact cctgacctca ggtcatccac ccacctcagc 
    ctcgcaaagt gctgagatta caggcatgag ccacagcgcc
    cagcttaaac tattttcttg gtctgttttt gattttcttt 
    taccttgcc actgcggtac agattattt tactcactgc
    cactaaacta aagcaaggca tagatatat gtgaagtgtt 
    cagagtttac tgctataagg aaacttccaa atactgacat
    ttacctata gctgtagtta ttgggaccat gtgctctggt 
    tttctggaga ctgccaaatt gctcccattt ttctgcatcc
    cacctggttt ctttctgcat gtcccctttc actttcaaac 
    ctatcattt ggatgttaaa ttatatggtc acctagttat
    aggtaagcct tgttcgagtt gatatcttga ttgtgaggaa 
    ggatctgtgt cattggagct tgtttctgct gcaacgtgct
    gtagactatg aataatgaaa tcacaccaca ttaccatcag 
    atttcttgtt ttagttgtca aattaatatt tatgattgtt
    atcttgggcg aaaagttcag agcagagatg acaaatcatt 
    agaacaacga tgaatttcag tattacggct aaaaagttct
    tctgtctgaa tattaactca ctctccttcc agtgtacttc 
    acagtaattg gtatgctttt ttatttaatg cttaaatcaa
    actttataaa aatcttagac cagatcttta atatggtatg 
    ccatttcccc agtctaccaa tggaatagta tgggtttcta
    atcctaggct tgtacaatgg attggagttg agccatgcca 
    gcctccacac tgccactaac ttctgtaatg taagattgag
    tcactgccaa gcatttgaaa tatgcagttg tgttttaatt 
    ataatttatg tatagttaga tgtatgtagt gcattgtgtg
    gtattatttg gtttgtaaga atttattttt aagggtcaag 
    gtcatttgta acattttgtg tgtgtcaatt caatgcaatg
    ttggctgcct tttgaagtct ttgatatatt ggtgaatatt 
    cttctgatct ataatacaaa gctatgtaat gttacctctt
    gactcgcttt tgaaaggaag acaattgtta actagatatt 
    tgagtttttt cccctcagaa ttatgtgaat ttctgatata
    tggctttaga tactgtgaat ctgttttcca tttagtcagt 
    tatctgctta aattgttcag aactatatcc taacgagcaa
    ttagttctga tggttctccc agtcatgagt gtgcatgtgt 
    gcaagcatgt tttgatcctg atgctacctt tgctaaaaat
    ggccatagat taggaactag ctatgttttt agaatcaaag 
    atgaaccggt aagctgtctc atgtaccaaa cgtgaaattt
    acagtgttta caaatgtctg gaattttgca ctgccatagg 
    gaatgttaag gttacttggc tggaatttat cagacttgtg
    agtaaacaag ttgaagttta gcagatgagg gggaatattg 
    aggcccctaa ggctaaacaa aataatcagt atctgagata
    gtggctaatg tggctcccca ggcctaattt gggaacagtt 
    tttcctgatt gctttgagaa gtactttctt
    ttgacagaaa ttttcattct gcttgccatt gctatattct 
    ccattatag gagccattgg atttattcc ttttgtggga
    aatgtcccat tagcattttc agatatttg atgtgcacta 
    atgccattat tggtaatgcc gttattggtg aatacagcat
    agttaaataa actgttacag taaatctaca cttggatttg 
    ctgcacctct accaatagcc ttttgaatga ctgaaagtgt
    taacagagaa agaggcatgt ctgcagaaag agatagctaa
    tattttttgg tactttatct gaaatccaag atgctgcttc
    ccctgcaggt tgttttcctt cttacgatcc tcattgaatc
    ccctctggga gcacaggaca gttagtagaa ctctccattt
    cttttttttt ttttttagac ggagtctctc tctgtcgccc
    cggctggagt gcagtggcgc gatctcggct cactgcaacc
    tccgcctccc gggttcaccc cattctcctg cctcagcctc
    cctagtagct gggactatag gcgcccgcca ccacgcctgg
    ctaatttttg tatttttatt ggagacgggg tttcaccgtc
    ttagccagga tggtatgat ctcctgacct cgtgatctgc
    ccacctcagc ctcccaaagt actgggatta caggcgtgag
    ccaccgcgcc cggccggaac tctccatttc ttaaggtaaa 
    gagggtcaag gatacctaaa aagggtcaaa taatgctaga
    agagcaattc ctctttcaga gcagttgctg taatttggca
    aatgctttat cgaagattga tattaggcta ggggcggtgg
    cttacgcctg taatcccagc actttgggag gccgaggtgg
    gtggattgcc tgagctcagg agttcgagac cagtctgacc
    agtatggtga aaccctgtct ctactaaaaa tacaaaaatt
    agccggtcgt ggtggcgtgc acctgtagtc ccagctactt
    ggcaggttga gacaggagaa tcgcttgaac ctgggaggtg
    gaggttgcag tgagccgaga ctgcaccact gcgctcccac 
    ctgggtgaca gagactctgt ctcaaaaaaa aggacattta
    tcattataac atcttattag agcccctaat ttcttatctg
    aaggcactgt tttttttttt aaacagttaa gtactgatgt
    caacagacaa atatttctga tcagatagtc ccctgtcaac 
    agtagcaaat gtggtttcat aaagtgggaa gaaaacagca
    ttttaaagta actttttggg agactgattt gagtaataat 
    aaaactctgg tctcccttaa gaaaaaaaaa cccttccacc
    tttactgtgt catttatatc cccttagttc caaagttaat 
    tatatattt ctggatang atttatacc aaagaccat
    atcagccat gtaactacag tatattaga taagattcct 
    ctttccagtc agtcctggga aatgtttctg ttgcagagtt
    aggcggtaga tgggaagctg tgatggcaga 
    gctactatct aataaagtaa caactcgtag ttgaggcttc
    ctttctgtgt gtgatggggg atagggagtt agctcccctg 
    ttgtctcagc actaagaaat tgaggtcagg ccaggcgcgg
    tggttcactc ctgttattcc agcactgggg 
    tggccaaagt gggcagattg cttgcgctct ggagctcgag
    accagcctgg gcaacatggt gaaaccctgt 
    ctctaccaaa aatacaaaaa aaaagctggg catggtgggt
    gcatgcttgt cccagctact gaggaggctg 
    aggtgggagg atcgcttgag cctgggaggt ggaggttgca
    gtgagctgag atggcaccac tgcaatccaa 
    ggtgggtgac agagacgctg tctcaaagaa attgaggtca
    ggcttccttc ttacagaatt atttttttct ctgtagtttg 
    cctcattftt tcactttctt ttcaatgaga atcgaagtgt
    ttcttttggg tttttttttc ccccttttaa aatcaacagg 
    aaatgtttca aaggagggat gaaatgcttc ttggatcct
    cagcacttgg caaggtagac ctcatagcaa ccttgaatat 
    gactttcttt agtctctagc tatgcactat taagtgcctc
    ttgggtagag gtagagttaa gtattgagtg ccagtcttga 
    cgtccgtatg cctcagtttt tctcatatat aaaaagcagt
    atacatacct acccttttct acctcatcat ttgttgtagg 
    gattaaatcc gggagagcaa ttctgaagcc tataaatttc
    cttgaagaga tctaagaacc tattatgctc ttggtgtacc 
    aagctctggg gtatatattc agaatacctc atgttctgga
    agctgagcac tagctcccct ttattgcctg cctggcagag 
    cctgtttgat tactgcaggc ccttttaccc atgcttctag
    tttaggtatt attattga tatgaggctc ttgaccagaa 
    aagagttctt tctctaggtg ttctgagaga agtttgtaaa
    tttggatagt acattctatc ctgataaaac caccttgctg 
    tggtatgat gtacaaaaaa aaattttttt tttgagacag
    agtatactc tgtcacccag gctggaatgc agtggcgcaa 
    tcttggttca ctgcaacccc cgcctcctgg gttcaagcga
    tcctcctgcc tcaacctctc aagtagctgg 
    gactacaggc gtgcaccacc acacctggct aattttgta
    ttttagtaga gacagggttt caccatgttg 
    gccaggctgg tcttgaactc ctgacctcag gcgatctgcc
    cgccttggcc tcccaaagta ctgggattac 
    aggcgtgagc aactgctcct ggcccaaaac atctctact 
    acatacactt gagtaggtgg cataaaatgc 
    actgtcaata tatagaaaac atgaaatttt ccaaatattt
    ccgatcagag aatcacaaga gcagcaaatg tggtttcat 
    aagtgggaag aaagcagcaa tttaaaataa ctttttggga
    gactgaattg agtaataata aaacttcagt ctttcgctaa 
    taataataat aataataata ataacaacaa cttattgaat
    gtggccagct cactagatga ggaaagagga aggcattttc 
    tgcattcttg cctagttttc cttataagca ccactaagtt
    aatagctctg tctttttggt gtttgcacta tgtaatgctt 
    ttaatacttt ttaattgtgc ttttttatgt attaaatgtt
    tttccttttg cca 
    (CA VI)(NM_001215) 
    SEQ ID NO: 9
    MRALVLLLSLELLGGQAQHVSDWTYSEGALDEAHWPQHYPACGGQRQSP 
    INLQRTKVRYNPSLKGLNMTGYETQAGEFPMVNNGHTVQISLPSTMRMT 
    VADGTVYIAQQMHFHWGGASSEISGSEHTVDGIRHVIEIHIVHYNSKYK 
    SYDIAQDAPDGLAVLAAFVEVKNYPENTYYSNFISHLANIKYPGQRTTL 
    TGLDVQDMLPRNLQHYYTYHGSLTTPPCTENVHWFVLADFVKLSRTQVW 
    KLENSLLDHRNKTIHNDYRRTQPLNHRVVESNFPNQEYTLGSEFQFYLH
    KIEEILDYLRRALN 
  • All references, publications, patent applications, issued patents, accession records and databases cited herein, including in any appendices, are incorporated by reference in their entirety for all purposes.

Claims (13)

1. A method for diagnosing breast cancer status in a subject, the method comprising:
a) analyzing a saliva sample from the subject with an assay that specifically detects at least two biomarkers in the saliva sample, wherein at least one biomarker is selected from the group consisting of the biomarkers S100A8 (S100 calcium binding protein A8) (SEQ ID NO: 1), CSTA (cystatin A) (SEQ ID NO:2), GRM1 (glutamate receptor, metabotropic 1) (SEQ ID NO: 3), TPT1 (tumor protein, translationally-controlled 1)(SEQ ID NO:4), GRIK1 (glutamate receptor, ionotropic, kainate 1) (SEQ ID NO: 5), H6PD (hexose-6-phosphate dehydrogenase) (SEQ ID NO: 6), IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) (SEQ ID NO: 7), MDM4 (3T3 cell double minute 4) (SEQ ID NO: 8), and CA6 (carbonic anhydrase VI) (SEQ ID NO: 9); and
b) determining whether or not the at least two biomarkers are differentially expressed in the sample relative to a control; thereby providing breast cancer status.
2. The method of claim 1, wherein one of the at least two biomarkers is cystatin A (CSTA).
3. The method of claim 1, wherein one of the at least two biomarkers is CSTA and the other biomarker of the at least two biomarkers is transformed 3T3 cell double minute 4 (MDM4).
4. The method of claim 1, wherein at least three biomarkers are measured.
5. The method of claim 1, wherein one of the at least two biomarkers is anhydrase VI (CA6) polypeptide.
6. The method of claim 1 wherein the assay detects a nucleic acid encoding at least one biomarker, and wherein the nucleic acid is detected by mass spectroscopy, PCR, microarray hybridization, thermal sequencing, capillary array sequencing, or solid phase sequencing.
7. The method of claim 1, wherein the assay detects a polypeptide of at least one biomarker, and wherein the polypeptide is detected by ELISA, Western blot, flow cytometry, immunofluorescence, immunohistochemistry, or mass spectroscopy.
8. A method of assessing the efficacy of a therapy on a subject comprising:
(a) analyzing a first saliva sample from the subject with an assay that specifically detects at least two biomarkers selected from the group consisting of S100A8 (SEQ ID NO: 1), CSTA (cystatin A) (SEQ ID NO:2), GRM1 (glutamate receptor, metabotropic 1) (SEQ ID NO: 3), TPT1 (tumor protein, translationally-controlled 1) (SEQ ID NO:4), GRIK1 (glutamate receptor, ionotropic, kainate 1) (SEQ ID NO: 5), H6PD (hexose-6-phosphate dehydrogenase) (SEQ ID NO: 6), IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) (SEQ ID NO: 7), MDM4 (3T3 cell double minute 4) (SEQ ID NO: 8), and CA6 (carbonic anhydrase VI) (SEQ ID NO: 9), thereby providing a first expression profile;
(b) effecting a therapy on the subject;
(c) analyzing a second saliva from the subject with an assay that specifically detects at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIM, H6PD, IGF2BP1, MDM4, and CA6; thereby providing a second expression profile;
(e) comparing the first and second expression profile, thereby assessing the efficacy of a therapy.
9. A kit comprising a solid support, wherein the solid support comprises a capture binding probe selective for at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, H6PD, IGF2BP1, MDM4.
10. A kit comprising a first and a second solid support, wherein the first solid support comprises a capture binding probe selective for at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, and wherein the second solid support comprises a capture binding ligand for CA6.
11. The kit of claim 10, wherein the capture binding ligand is an antibody.
12. A kit comprising one or more primers for the selective amplification of at least two biomarkers selected from the group consisting of S100A8, CSTA, GRM1, TPT1, GRIK1, H6PD, IGF2BP1, MDM4, wherein each of the primers optionally comprises a detectable label.
13. A method for diagnosing breast cancer status in a subject, the method comprising:
a) analyzing a saliva sample from the subject with an assay that specifically detects at least nine biomarkers in the saliva sample, wherein the at least nine biomarkers are selected from the group consisting of the biomarkers S100A8 (S100 calcium binding protein A8) (SEQ ID NO: 1), CSTA (cystatin A) (SEQ ID NO:2), GRM1 (glutamate receptor, metabotropic 1) (SEQ ID NO: 3), TPT1 (tumor protein, translationally-controlled 1) (SEQ ID NO:4), GRIK1 (glutamate receptor, ionotropic, kainate) (SEQ ID NO: 5), H6PD (hexose-6-phosphate dehydrogenase) (SEQ ID NO: 6), IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) (SEQ ID NO: 7), MDM4 (3T3 cell double minute 4) (SEQ ID NO: 8), and CA6 (carbonic anhydrase VI) (SEQ NO: 9); and
b) determining whether or not the at least nine biomarkers are differentially expressed in the sample relative to a control; thereby providing breast cancer status
US15/488,832 2010-02-10 2017-04-17 Salivary Transcriptomics and Proteomic Biomarkers for Breast Cancer Detection Abandoned US20170219586A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/488,832 US20170219586A1 (en) 2010-02-10 2017-04-17 Salivary Transcriptomics and Proteomic Biomarkers for Breast Cancer Detection

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US30320010P 2010-02-10 2010-02-10
US13/025,110 US9624547B2 (en) 2010-02-10 2011-02-10 Salivary transcriptomic and proteomic biomarkers for breast cancer detection
US15/488,832 US20170219586A1 (en) 2010-02-10 2017-04-17 Salivary Transcriptomics and Proteomic Biomarkers for Breast Cancer Detection

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/025,110 Continuation US9624547B2 (en) 2010-02-10 2011-02-10 Salivary transcriptomic and proteomic biomarkers for breast cancer detection

Publications (1)

Publication Number Publication Date
US20170219586A1 true US20170219586A1 (en) 2017-08-03

Family

ID=44368124

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/025,110 Active US9624547B2 (en) 2010-02-10 2011-02-10 Salivary transcriptomic and proteomic biomarkers for breast cancer detection
US15/488,832 Abandoned US20170219586A1 (en) 2010-02-10 2017-04-17 Salivary Transcriptomics and Proteomic Biomarkers for Breast Cancer Detection

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/025,110 Active US9624547B2 (en) 2010-02-10 2011-02-10 Salivary transcriptomic and proteomic biomarkers for breast cancer detection

Country Status (2)

Country Link
US (2) US9624547B2 (en)
WO (1) WO2011100472A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX355020B (en) 2010-07-09 2018-04-02 Somalogic Inc Lung cancer biomarkers and uses thereof.
CN106198980B (en) 2010-08-13 2018-09-07 私募蛋白质体公司 Cancer of pancreas biomarker and application thereof
EP2771692B1 (en) * 2011-10-24 2018-04-18 Somalogic, Inc. Lung cancer biomarkers and uses thereof
KR101431064B1 (en) 2012-03-08 2014-08-21 (주)바이오메디앙 Protein marker carbonic anhydrase 1 for breast cancer diagnosis, method of detecting the same, and diagnosis kit for breast cancer using antibody against the same
CN103848889B (en) * 2012-11-30 2015-12-02 北京市结核病胸部肿瘤研究所 The antigenic peptide of IGF2BP1 autoantibody identification
US10613090B2 (en) 2014-05-09 2020-04-07 Ascendant Diagnostics, LLC Methods of detecting cancer
EP3242132B1 (en) * 2016-05-05 2020-02-12 Ascendant Diagnostics, LLC Methods of detecting cancer
US20200081008A1 (en) * 2016-11-22 2020-03-12 Prime Genomics, Inc. Methods for cancer detection
CN107385026B (en) * 2017-07-06 2020-08-28 北京大学深圳医院(北京大学深圳临床医学院) Group of mutant genes related to breast cancer and auxiliary diagnostic kit thereof
CN112730836B (en) * 2020-12-24 2023-05-05 北京信息科技大学 Non-diagnostic detection method for multiple tumor markers based on SERS (surface enhanced Raman scattering) sensing substrate

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2005300688B2 (en) 2004-11-03 2012-02-02 Almac Diagnostics Limited Transcriptome microarray technology and methods of using the same
JP2009510395A (en) * 2005-09-14 2009-03-12 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Salivary proteins and RNA for breast cancer detection
CA2660286A1 (en) * 2006-08-09 2008-02-21 Homestead Clinical Corporation Organ-specific proteins and methods of their use
US9353415B2 (en) * 2006-12-19 2016-05-31 Thomson Reuters (Scientific) Llc Methods for functional analysis of high-throughput experimental data and gene groups identified therefrom
AU2008264893A1 (en) * 2007-04-16 2008-12-24 Inserm- Institut National De La Sante Et De La Recherche Medicale Methods of assessing a propensity of clinical outcome for a female mammal suffering from breast cancer
US20100279419A1 (en) * 2007-09-18 2010-11-04 Board Of Regents Of The University Of Texas System Detection of saliva proteins modulated secondary to ductal carcinoma in situ of the breast
US20100190656A1 (en) 2008-08-08 2010-07-29 Integrated Diagnostics, Inc. Breast Cancer Specific Markers and Methods of Use
AU2011277034B2 (en) 2010-07-08 2014-04-10 Prime Genomics, Inc. System for the quantification of system-wide dynamics in complex networks

Also Published As

Publication number Publication date
US20110212851A1 (en) 2011-09-01
WO2011100472A1 (en) 2011-08-18
US9624547B2 (en) 2017-04-18

Similar Documents

Publication Publication Date Title
US20170219586A1 (en) Salivary Transcriptomics and Proteomic Biomarkers for Breast Cancer Detection
JP6114035B2 (en) Salivary biomarker for lung cancer detection
DK2456889T3 (en) Markers of endometrial cancer
CA2616277C (en) Urine markers for detection of bladder cancer
US9939442B2 (en) Salivary biomarkers for gastric cancer detection
KR101566368B1 (en) Urine gene expression ratios for detection of cancer
JP5892701B2 (en) Gastric cancer detection marker
US9182383B2 (en) Biomarkers for the diagnosis and treatment of pancreatic cancer
KR20150090246A (en) Molecular diagnostic test for cancer
KR20160117606A (en) Molecular diagnostic test for predicting response to anti-angiogenic drugs and prognosis of cancer
KR20160057416A (en) Molecular diagnostic test for oesophageal cancer
KR101750146B1 (en) Use of RSPH9 for Prognostic Marker Diagnosis of Bladder Cancer
KR20130040294A (en) Composition for diagnosis of small cell lung cancer and diagnosis kit of small cell lung cancer
US20120289416A1 (en) Methods and kits used in assessing cancer risk
US20220064235A1 (en) Urine Markers and Methods for Detection of Bladder Cancer and Treatment Thereof
KR102657306B1 (en) Use of markers including filamin a in the diagnosis and treatment of prostate cancer
KR20230086462A (en) Novel Biomarker for Predicting Therapeutic Response and Prognosis of Metastatic Breast Cancer To Chemotherapeutic Agents and Uses Thereof
KR20170092671A (en) Use of markers including filamin a in the diagnosis and treatment of prostate cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF CALIFORNIA LOS ANGELES;REEL/FRAME:043702/0942

Effective date: 20170828

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: 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: 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