WO2006063133A2 - Biomarqueurs pour maladie intestinale inflammatoire - Google Patents

Biomarqueurs pour maladie intestinale inflammatoire Download PDF

Info

Publication number
WO2006063133A2
WO2006063133A2 PCT/US2005/044423 US2005044423W WO2006063133A2 WO 2006063133 A2 WO2006063133 A2 WO 2006063133A2 US 2005044423 W US2005044423 W US 2005044423W WO 2006063133 A2 WO2006063133 A2 WO 2006063133A2
Authority
WO
WIPO (PCT)
Prior art keywords
inflammatory bowel
bowel disease
markers
biomarker
biomarkers
Prior art date
Application number
PCT/US2005/044423
Other languages
English (en)
Other versions
WO2006063133A3 (fr
Inventor
Chakravarti Shukti
Feng Wu
Original Assignee
The Johns Hopkins University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Johns Hopkins University filed Critical The Johns Hopkins University
Priority to EP05853365A priority Critical patent/EP1844158A4/fr
Publication of WO2006063133A2 publication Critical patent/WO2006063133A2/fr
Priority to US11/750,114 priority patent/US20090258848A1/en
Publication of WO2006063133A3 publication Critical patent/WO2006063133A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/06Gastro-intestinal diseases
    • G01N2800/065Bowel diseases, e.g. Crohn, ulcerative colitis, IBS

Definitions

  • BACKGROUND OF THE INVENTION Crohn's disease (CD) and ulcerative colitis (UC), are complex, heterogeneous, multifactorial diseases involving genetic, environmental and microbial factors.
  • IBD inflammatory bowel disease
  • Crohn's disease and ulcerative colitis have similar symptoms, but are very different in the manner in which they affect the digestive tract.
  • UC ulcerative colitis
  • Diagnosis and classification of these diseases are primarily based on patient histories and serologic, radiological, endoscopic and histopathology findings. 6 Early, precise differentiation and diagnosis would directly influence the clinical treatment, patient management and the outcome of such diseases.
  • the present invention provides, for the first time, novel biomarkers that are differentially present in the samples of inflammatory bowel disease(IBD) subjects and in the samples of control subjects.
  • the present invention also provides sensitive and quick methods and kits that are useful for determining the inflammatory bowel disease status by measuring these novel markers.
  • the measurement of these markers alone or in combination, in patient samples provides information that a diagnostician can correlate with a probable diagnosis of inflammatory bowel disease or a negative diagnosis (e.g., normal or disease-free).
  • the markers are characterized by their known protein identities or by their m/z value or molecular weight and/or by characteristics discussed herein.
  • the markers can be resolved in a sample by using a variety of techniques, e.g., microarrays, PCT techniques (e.g., real time, reverse transcriptase, PCR), and fractionation techniques (e.g., chromatographic separation coupled with mass spectrometry, protein capture using immobilized antibodies or by traditional immunoassays).
  • PCT techniques e.g., real time, reverse transcriptase, PCR
  • fractionation techniques e.g., chromatographic separation coupled with mass spectrometry, protein capture using immobilized antibodies or by traditional immunoassays.
  • the present invention provides a method of qualifying inflammatory bowel disease status in a subject comprising measuring at least one biomarker in a sample from the subject.
  • the method of resolution involves Surface-Enhanced Laser Desorpti on/Ion ization ("SELDI”) mass spectrometry, in which the surface of the mass spectrometry probe comprises adsorbents that bind the markers.
  • the invention provides biomarkers for inflammatory bowel disease status comprising one or more of the following Markers 1-75 and combinations thereof. These Markers 1-75 are set forth in Table 1-3, which follows and are sometimes referred to herein as biomarkers of Table I or similar designations.
  • the biomarker for inflammatory bowel disease status of the invention comprises Markers 1-75.
  • markers 1 - 48 are Markers of Crohn's disease (CD).
  • markers 49 - 75 are markers of ulcerative colitis.
  • markers 49-60 are up-regulated in ulcerative colitis (UC).
  • markers 61-75 are down-regulated in ulcerative colitis.
  • markers 1, 2, 4 and 5 are correlate with CD; markers 6 and 10 correlate with CD; markers 17, 18, and 21 correlate with CD; markers 55 and 57 correlate with UC; markers 55 and 57 are up-regulated in UC; markers 69, 74 and 75 and are down- regulated in UC.
  • markers may discriminate between IBD disease state, for example, markers 1, 6, 17, 55 and 69 discriminate between UC and CD; markers 2, 10, 18, 57, and 74 also discriminate between UC and CD; as do markers 4, 6, 21, 55, and 69; and markers 1, 6, and 17; and markers 55 and 69.
  • the biomarkers may be used in combination, for example, markers 1, 2, 4 and 5; markers 6 and 10; markers 17, 18, and 21 ; markers 55 and 57; markers 69, 74 and 75; markers 1, 6, 17, 55 and 69; markers 2, 10, 18, 57, and 74; 4, 6, 21, 55, and 69; markers 1, 6, and 17; and markers 55 and 69.
  • the invention provides, in one aspect, methods for qualifying IBD status in a subject comprising measuring at least one biomarker in a sample from the subject, wherein the biomarker is selected from one or more of the biomarkers of Tables 1 - 3, and correlating the measurement with inflammatory bowel disease status.
  • the inflammatory bowel disease is ulcerative colitis (UC) and/or Crohn's disease (CD).
  • the method further comprises managing subject treatment based on the status.
  • the managing subject treatment is selected from ordering further diagnostic tests (e.g., colonoscopy and imaging techniques), administering at least one therapeutic agent, surgery, surgery followed or preceded by at least one therapeutic agent, biotherapy, and taking no further action.
  • further diagnostic tests e.g., colonoscopy and imaging techniques
  • the therapeutic agent is selected from one or more of an antibiotic, an antispasmotic, and/or an antidepressant.
  • antibiotics include, for example, rifaximin.
  • Other therapeutic agents include, for example, sulfa drugs, corticosteriods (prednisone), 5-aminosalicylates (Asacol, Pentasa, Rowasa, or 5-ASA), immunosuppressives (azathioprine, Imuran, Cyclosporine, 6-MP, Purinethol and Methotrexate), anti-TNF (Remicade), anticholinergics, dicyclomine (Bentyl), belladonna/phenobarbital (Donnatal, Antispas, bBarbidonna, donnapine, hyosophen, Spasmolin), hyoscyamine (Levsin, Anaspaz), chlordiazepoxide/clidinium (Librax), anti- diarrheals, diphenoxylate/
  • the method for qualifying inflammatory bowel disease status in a subject may further comprise measuring the at least one biomarker after subject management.
  • the inflammatory bowel disease status is selected from one or more of the subject's risk of IBD, the presence or absence of IBD, the type of IBD disease, the stage of IBD and effectiveness of treatment.
  • the inflammatory bowel disease status is selected from one or more of the presence or absence of alternating diarrhea and constipation, abdominal pain, bloating, spasms, nausea, bloody diarrhea, fever, dehydration, eye inflammation, joint pain, skin rashes or lesions, mouth ulcers, chronic diarrhea, weight loss, lack of appetite, nutritional deficiencies, and/or inflamed colon.
  • Methods may further comprise assessing the status of the inflammatory bowel disease, for example, by barium enema, upper GI series, stool culture, blood tests (to determine a white blood cell count or if anemia is present), fecal occult blood test, sigmoidoscopy, and/or colonoscopy.
  • the invention provides, in another aspect, methods for differentiating between a diagnosis of UC and CD comprising detecting in a subject sample an amount of at least one biomarker wherein the biomarker is selected from one or more of the biomarkers of Tables 1 - 3, and correlating the amount with a diagnosis of inflammatory bowel disease or noninflammatory bowel disease.
  • Marker 34 Complement component 4 binding protein, ⁇ C4BPB
  • Coagulation factor 11 (thrombin) receptor-like 1 F2RL1 Marker 68 Surfactant, pulmonary-associated protein D SFTPD Marker 69 Solute carrier family 4, sodium bicarbonate cotransporter, member SLC4A4
  • GABA Gamma-aminobuty ⁇ c acid
  • GABRGi Marker 71
  • HPGD Marker 72 TAF5-hke RNA polymerase II, p300/CBP-associated factor TAF SL
  • markers of the invention may be detected, for example, by mass spectrometry according to one embodiment.
  • the markers are detected by SELDI.
  • the marker or markers are detected by capturing the marker on a biochip having a hydrophobic surface and detecting the captured marker by SELDI. Suitable biochips include the IMAC3 ProteinChip® Array and the WCX2 ProteinChip® Array.
  • markers are detected by nucleic acid arrays, e.g., DNA arrays or by PCR methods.
  • the methods for qualifying inflammatory bowel disease status in a subject further comprise generating data on immobilized subject samples on a biochip, by subjecting the biochip to laser ionization and detecting intensity of signal for mass/charge ratio; and transforming the data into computer readable form; executing an algorithm that classifies the data according to user input parameters, for detecting signals that represent biomarkers present in inflammatory bowel disease subjects and are lacking in noninflammatory bowel disease subject controls.
  • one or more of the biomarkers are detected using laser desorption/ionization mass spectrometry, comprising providing a probe adapted for use with a mass spectrometer comprising an adsorbent attached thereto; contacting the subject sample with the adsorbent; desorbing and ionizing the biomarker or biomarkers from the probe; and detecting the desorbed/ionized markers with the mass spectrometer.
  • least one or more protein biomarkers are detected using immunoassays.
  • the sample from the subject is one or more of colon biopsy material, intestinal biopsy material, fecal material, blood, blood plasma, serum, urine, cells, organs, seminal fluids, bone marrow, saliva, stool, a cellular extract, a tissue sample, a tissue biopsy, and cerebrospinal fluid.
  • the methods for qualifying inflammatory bowel disease status in a subject further comprise measuring the amount of each biomarker in the subject sample and determining the ratio of the amounts between the markers.
  • the measuring is selected from detecting the presence or absence of the biomarkers(s), quantifying the amount of marker(s), and qualifying the type of biomarker.
  • at least two biomarkers are measured.
  • at least three biomarkers are measured.
  • at least four biomarkers are measured.
  • at least one UC and at least one CD biomarker is measured.
  • the protein biomarkers are measured by one or more of electrospray ionization mass spectrometry (ESI-MS), ESI-MS/MS, ESI-MS/(MS) n , matrix- assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS), surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF- MS), desorption/ionization on silicon (DIOS), secondary ion mass spectrometry (SIMS), quadrupole time-of-flight (Q-TOF), atmospheric pressure chemical ionization mass spectrometry (APCI-MS), APCI-MS/MS, APCI-(MS).sup.n, atmospheric pressure photoionization mass spectrometry (APPI-MS), APPI-MS/MS, and APPI-(MS) n , quadrupole mass spectrometry, fourier transform mass spectrometry,
  • kits for example, for aiding the diagnosis of inflammatory bowel disease or the diagnosis of the subtypes of inflammatory bowel disease.
  • the kits may suitably include an adsorbent, wherein the adsorbent retains one or more biomarkers selected from one or more of the markers of Tables 1 - 3, and written instructions for use of the kit for detection of inflammatory bowel disease.
  • the kit for aiding the diagnosis of the subtypes of inflammatory bowel disease comprises an adsorbent, wherein the adsorbent retains one or more biomarkers selected from each of Markers 1 - 48 and Markers 49-75, and written instructions for use of the kit for detection of the IBD or a subtype of inflammatory bowel disease, e.g., UC or CD.
  • Kits may also comprise instructions provide for contacting a test sample with the adsorbent and detecting one or more biomarkers retained by the adsorbent, wherein the adsorbent is, for example, an antibody, single or double stranded oligonucleotide, amino acid, protein, peptide or fragments thereof.
  • the adsorbent is, for example, an antibody, single or double stranded oligonucleotide, amino acid, protein, peptide or fragments thereof.
  • the one or more protein biomarkers is detected using mass spectrometry, immunoassays, or PCR.
  • the measuring is selected from detecting the presence or absence of the biomarkers(s), quantifying the amount of marker(s), and qualifying the type of biomarker.
  • the invention provides methods for identifying a candidate compound for treating inflammatory bowel disease comprising contacting one or more of the biomarkers of Tables 1 - 3 with a test compound; and determining whether the test compound interacts with the biomarker, wherein a compound that interacts with the biomarker is identified as a candidate compound for treating inflammatory bowel disease.
  • the invention also provides methods of treating inflammatory bowel disease comprising administering to a subject suffering from or at risk of developing inflammatory bowel disease a therapeutically effective amount of a compound capable of modulating the expression or activity of one or more of the biomarkers of Tables 1 - 3.
  • the invention provides methods of treating a condition in a subject comprising administering to a subject a therapeutically effective amount of a compound which modulates the expression or activity of one or more of the biomarkers of Tables 1 - 3.
  • the compound are selected from the group consisting of enzyme inhibitor, cytotoxic drug, cytokin, chemokine, antibodies, a DNA molecule, an RNA molecule, a small molecule, a peptide, and a peptidomimetic.
  • Classes of drugs include, antiinflammatory, antibiotic, antiviral, antidepressant, anticonvulsant therapeutics.
  • the invention provides methods for modulating the concentration of a biomarker, wherein the biomarker is one or more of the biomarkers listed in Tables 1 - 3.
  • the method comprises contacting a cell with a test compound, measuring at least one biomarker, wherein the biomarker is selected from one or more of the biomarkers of Tables 1 - 3, and correlating the measurement with a determination of efficacy.
  • the invention also provides, in one aspect, a method of identifying a biomarker comprising obtaining an endoscopic sample from a subject, isolating nucleic acid from the sample, analyzing the nucleic acid and correlating the results.
  • the results may be analyzed against a control database of IBD samples and/or controls.
  • the invention also provides methods of determining the inflammatory bowel disease status of a subject, comprising (a) obtaining a biomarker profile from a sample taken from the subject; and (b) comparing the subject's biomarker profile to a reference biomarker profile obtained from a reference population, wherein the comparison is capable of classifying the subject as belonging to or not belonging to the reference population; wherein the subject's biomarker profile and the reference biomarker profile comprise one or more markers listed in Tables 1 - 3.
  • the comparison of the biomarker profiles can determine inflammatory bowel disease status in the subject with an accuracy of at least about 60%, 70%, 80%, 90% or approaching 100%.
  • the sample is fractionated by one or more of chemical extraction partitioning, ion exchange chromatography, reverse phase liquid chromatography, isoelectric focusing, one-dimensional polyacrylamide gel electrophoresis (PAGE), two- dimensional polyacrylamide gel electrophoresis (2D-P AGE), thin-layer chromatography, gas chromatography, liquid chromatography, and any combination thereof.
  • chemical extraction partitioning ion exchange chromatography, reverse phase liquid chromatography, isoelectric focusing, one-dimensional polyacrylamide gel electrophoresis (PAGE), two- dimensional polyacrylamide gel electrophoresis (2D-P AGE), thin-layer chromatography, gas chromatography, liquid chromatography, and any combination thereof.
  • the measuring step comprises quantifying the amount of marker(s) in the sample. In other methods, the measuring step comprises qualifying the type of biomarker in the sample.
  • the biomarkes may be sufficiently characterized by, e.g., mass and by affinity characteristics. It is noted that molecular weight and binding properties are characteristic properties of the markers and not limitations on means of detection or isolation. Furthermore, using the methods described herein or other methods known in the art, the absolute identity of markers can be determined.
  • the present invention also relates to biomarkers designated as Markers 1-75.
  • Protein markers of the invention can be characterized in one or more of several respects. In particular, in one aspect, these markers are characterized by molecular weights under the conditions specified herein, particularly as determined by mass spectral analysis. In another aspect, the markers can be characterized by features of the markers' mass spectral signature such as size (including area) and/or shape of the markers' spectral peaks, features including proximity, size and shape of neighboring peaks, etc. In yet another aspect, the markers can be characterized by affinity binding characteristics, particularly ability to binding to cation- exchange and/or hydrophobic surfaces. In preferred embodiments, markers of the invention may be characterized by each of such aspects, i.e. molecular weight, mass spectral signature and cation and/or hydrophobic absorbent binding.
  • the present invention provides for a method for detecting and diagnosing (including e.g., differentiating between) different subtypes of inflammatory bowel disease, wherein the method comprises using a biochip array for detecting at least one biomarker in a subject sample; evaluating at least one biomarker in a subject sample, and correlating the detection of one or more protein biomarkers with a inflammatory bowel disease subtype, e.g., UC and CD.
  • a biochip array for detecting at least one biomarker in a subject sample
  • evaluating at least one biomarker in a subject sample e.g., UC and CD.
  • the biomarkers of the invention may be detected in samples of blood, blood plasma, serum, urine, tissue, cells, organs, seminal fluids, bone marrow, colon biopsies, intestinal biopsies, and cerebrospinal fluid.
  • Biochip arrays useful in the invention include protein and nucleic acid arrays.
  • One or more markers are captured on the biochip array and subjected to laser ionization to detect the molecular weight of the markers. Analysis of the markers is, for example, by molecular weight of the one or more markers against a threshold intensity that is normalized against total ion current.
  • the step of correlating the measurement of the biomarkers with inflammatory bowel disease status is performed by a software classification algorithm.
  • data is generated on immobilized subject samples on a biochip array, by subjecting the biochip array to laser ionization and detecting intensity of signal for mass/charge ratio; and transforming the data into computer readable form; and executing an algorithm that classifies the data according to user input parameters, for detecting signals that represent markers present in inflammatory bowel disease subjects and are lacking in non-inflammatory bowel disease subject controls.
  • the biochip surfaces are, for example, ionic, anionic, hydrophobic; comprised of immobilized nickel or copper ions;, comprised of a mixture of positive and negative ions; and/or comprised of one or more antibodies, single or double stranded nucleic acids, proteins, peptides or fragments thereof, amino acid probes, or phage display libraries.
  • one or more of the markers are measured using laser desorption/ionization mass spectrometry, comprising providing a probe adapted for use with a mass spectrometer comprising an adsorbent attached thereto, and contacting the subject sample with the adsorbent, and desorbing and ionizing the marker or markers from the probe and detecting the deionized/ionized markers with the mass spectrometer.
  • the laser desorption/ionization mass spectrometry comprises: providing a substrate comprising an adsorbent attached thereto; contacting the subject sample with the adsorbent; placing the substrate on a probe adapted for use with a mass spectrometer comprising an adsorbent attached thereto; and desorbing and ionizing the marker or markers from the probe and detecting the desorbed/ionized marker or markers with the mass spectrometer.
  • the adsorbent can for example be, hydrophobic, hydrophilic, ionic or metal chelate adsorbent, such as nickel or copper, or an antibody, single- or double stranded oligonucleotide, amino acid, protein, peptide or fragments thereof.
  • a process for purification of a biomarker comprising fractioning a sample comprising one or more protein biomarkers by size-exclusion chromatography and collecting a fraction that includes the one or more biomarker; and/or fractionating a sample comprising the one or more biomarkers by anion exchange chromatography and collecting a fraction that includes the one or more biomarkers. Fractionation is monitored for purity on normal phase and immobilized nickel arrays.
  • Generating data on immobilized marker fractions on an array is accomplished by subjecting the array to laser ionization and detecting intensity of signal for mass/charge ratio; and transforming the data into computer readable form; and executing an algorithm that classifies the data according to user input parameters, for detecting signals that represent markers present in inflammatory bowel disease subjects and are lacking in non-inflammatory bowel disease subject controls.
  • Preferably fractions are subjected to gel electrophoresis and correlated with data generated by mass spectrometry.
  • gel bands representative of potential markers are excised and subjected to enzymatic treatment and are applied to biochip arrays for peptide mapping.
  • biomarkers are selected from gel bands representing Markers 1-75 described herein.
  • Purified proteins for detection of inflammatory bowel disease and/or screening and aiding in the diagnosis of inflammatory bowel disease and/or generation of antibodies for further diagnostic assays are provided.
  • the invention provides methods for identifying compounds (e.g., antibodies, nucleic acid molecules (e.g., DNA, RNA), small molecules, peptides, and/or peptidomimetics) capable of treating inflammatory bowel disease comprising contacting at least one or more of a biomarker selected from Markers 1-75, and combinations thereof with a test compound; and determining whether the test compound interacts with, binds to, or modulates the biomarker, wherein a compound that interacts with, binds to, or modulates the biomarker is identifies as a compound capable of treated inflammatory bowel disease.
  • compounds e.g., antibodies, nucleic acid molecules (e.g., DNA, RNA), small molecules, peptides, and/or peptidomimetics
  • the invention provides methods of treating inflammatory bowel disease comprising administering to a subject suffering from or at risk of developing inflammatory bowel disease a therapeutically effective amount of a compound (e.g., an antibody, nucleic acid molecule (e.g., DNA, RNA), small molecule, peptide, and/or peptidomimetic) capable of modulating the expression or activity of one or more of the Biomarkes 1-75.
  • a compound e.g., an antibody, nucleic acid molecule (e.g., DNA, RNA), small molecule, peptide, and/or peptidomimetic
  • the invention provides methods of determining the inflammatory bowel disease status of a subject, comprising (a) obtaining a biomarker profile from a sample taken from the subject; and (b) comparing the subject's biomarker profile to a reference biomarker profile obtained from a reference population, wherein the comparison is capable of classifying the subject as belonging to or not belonging to the reference population; wherein the subject's biomarker profile and the reference biomarker profile comprise one or more markers listed in Tables 1 - 3.
  • Methods of the invention may further comprise repeating the method at least once, wherein the subject's biomarker profile is obtained from a separate sample taken each time the method is repeated.
  • samples from the subject are taken about 24, 30, 48, 60, and/or 72 hours apart.
  • the comparison of the biomarker profiles can determine inflammatory bowel disease status in the subject with an accuracy of at least about 60% to about 99% .
  • the reference biomarker profile is obtained from a population comprising a single subject, at least two subjects, and at least 20 subjects.
  • the methods of the present invention provide and solve the need for methods of accurately assessing, i.e., diagnostically, prognostically, and therapeutically, IBD, including UC and CD.
  • Figure 1 depicts gene expression signals from CD-76-aff-l (X axis) and CD-76-aff-2 (Y axis) biopsies from one affected area. Each point represents the expression value of a probe set (defining a gene) in log-scale in the two biopsies.
  • a probe set with a "Present” call in both arrays (red), “Absent” in both (yellow), and "Present” in either one of the two arrays (blue) is shown.
  • the diagonal lines indicate fold change of 2, 3, and 10 in expression levels between two arrays. For genes expressed differentially between the two arrays, change in expression must be > 2 fold, expression > 100 arbitrary units, and "Present call" in one sample.
  • Figure 2 depicts multidimensional scaling (MDS) of 32 samples.
  • Figure 3 depicts hierarchical clustering across all arrays, of the top 50 genes whose expression patterns correlate with the distribution of samples in the MDS plot of Figure 2.
  • the inflammation score (*) for each biopsy taken from Tables 1 - 3 are shown on the top.
  • Genes with similar expression levels across samples are clustered vertically and samples with similar gene expression patterns are grouped horizontally. Genes expressed above mean (red), mean (black) and below mean (green) are as shown. To derive this set of genes, each sample was assigned to one of four groups, depending on which quadrant it occupied in the MDS map, and an analysis of variance (ANOVA) on the expression values for each gene was calculated. Genes with large F-statistics have strong quadrant specific differences in expression. The top 50 genes with the highest F-statistic scores are shown.
  • Figure 4 is a model showing distinct pathogenic events in UC and CD. Gene symbols are taken from Tables 2, 3 and Figure 4. Gene up regulations and down regulations are indicated by arrows. We speculate that in response to microbial and other environmental stimuli, CD shows a deregulated immune response that entails acute phase response, antigen presentation and macrophage activation. In contrast early events in UC suggest impaired detoxification, overload of unfolded proteins and endoplasmic reticulum stress.
  • Figure 5 depicts histology of endoscopic biopsies of colon from a healthy control (A), CD-76, a patient with Crohn's disease (B and C), and UC-55, a patient with ulcerative colitis (D). (B) is taken from unaffected mucosa showing essentially normal colon structures.
  • CD76 a view of CD76 affected biopsy, showing significant inflammatory infiltration in the mucosa and submucosa, cryptitis with crypt abscesses, and basal lymphoplasmacytosis (inflammation grade: ++).
  • D UC-55 affected demonstrates crypt distortion and dropout, and lamina limbal fibrosis (fibrosis grade: ++).
  • MM muscularis mucosa
  • SM submucosa. H&E staining, original magnification 4Ox.
  • Figure 6 depicts the expressions of selected genes that were quantified by real-time RT-PCR.
  • the relative expression value of a gene was normalized to that of GAPD.
  • the samples include unaffected (un) and affected (aff) sample from six CD cases (CD-33, 51, 53, 58, 59 and 76), five UC samples (UC-32, 35, 38, 44 and 55) and four from normal controls (N65, N66, N69 and N79). Each point represents an individual sample.
  • CXCLl chemokine (C-X-C motif) ligand 1
  • DMBTl deleted in malignant brain tumors 1
  • ADM adrenomedullin
  • STAT3 signal transducer and activator of transcription 3
  • ASMT acetylserotonin O-methyltransferase
  • IFI35 interferon-induced protein 35
  • PSME2 proteasome activator subunit 2
  • PSMB8 proteasome subunit, beta type, 8.
  • the horizontal bar indicates the mean value of each group.
  • the present invention provides biomarkers generated from comparison of protein profiles from subjects diagnosed with inflammatory bowel disease and from subjects without known neoplastic diseases, using the mass spectrometry techniques.
  • the invention provides that these biomarkers, used individually, or preferably in combination with other biomarkers from this group or with other diagnostic tests, provide a novel method of determining inflammatory bowel disease status in a subject.
  • the present invention presents markers that are differentially present in samples of inflammatory bowel disease subjects and control subjects, and the application of this discovery in methods and kits for determining inflammatory bowel disease status.
  • These protein markers are found in samples from inflammatory bowel disease subjects at levels that are different than the levels in samples from subject in whom human IBD is undetectable. Accordingly, the amount of one or more markers found in a test sample compared to a control, or the presence or absence of one or more markers in the test sample provides useful information regarding the inflammatory bowel disease status of the patient.
  • the present invention also relates to a method for identification of biomarkers for IBD, with high specificity and sensitivity.
  • a panel of biomarkers were identified that are associated with inflammatory bowel disease status.
  • IBS Inflammatory bowel disease
  • IBS refers to a functional disorder of the colon (large intestine) that causes crampy abdominal pain, bloating, constipation and/or diarrhea.
  • IBS is classified as a functional gastrointestinal disorder because no structural or biochemical cause can be found to explain the symptoms.
  • the most common symptoms of IBD include, abdominal pain, weight loss, fever, rectal bleeding, skin and eye irritations, and diarrhea. Intervals of active disease, or 'flares', and periods of remission characterize IBD.
  • the colon shows no evidence of disease such as ulcers or inflammation. Therefore, IBS preferably diagnosed only after other possible digestive disorders and diseases have been ruled out. IBS is often misdiagnosed or misnamed as colitis, mucous colitis, spastic colon, irritable bowel disease or spastic bowel (colon).
  • Ultracerative colitis refers to a disease that is a form of IBD and causes inflammation and sores, called ulcers, in the top layers of the lining of the large intestine.
  • Common symptoms of UC include bloody diarrhea, fever and abdominal pain. There can also be symptoms outside the digestive system which are known as extra-intestinal symptoms.
  • Fever is a characteristic of the inflammatory process that takes place in UC and there are several extra-intestinal symptoms that are not directly related to the inflammation in the colon and include eye inflammation, joint pains, skin rashes or lesions, and mouth ulcers.
  • UC is diagnosed, for example, by stool culture, blood tests, fecal occult blood test, sigmoidoscopy, colonoscopy, and barium enema.
  • sulfasalazine Azulfadine
  • mesalamine Asacol, Pentasa, Rowasa
  • osalazine Dipentum
  • corticosteroids prednisone
  • Surgery may also be used to treat UC, usually after all available drug treatments have failed.
  • Surgery for UC always involves a total colectomy, or a complete removal of the large intestine (colon). Resection, or removing only the diseased section of the colon, is not an option in UC, because the disease will only re-occur in the portion of the colon that is left.
  • CD ulcerative colitis
  • a fissure is a tear or ulcer in the lining of the anal canal and symptoms include painful bowel movements, bright red blood in toilet bowel or on paper, anal lump, and swollen skin tag.
  • Acute fissures may be treated with Weg baths, fiber to create softer stools, stool softeners, topical hydrocortisone, zinc oxide, petroleum jelly and topical anesthetics.
  • a chronic fissure may need more aggressive treatment including surgery.
  • a fistula is an abnormal tunnel connecting two body cavities or a body cavity to the skin. Approximately 30% of people with Crohn's Disease develop fistulas. Treatments include antibiotics, immunosuppresants, Remicade, liquid nutrition to replace solid food and surgery.
  • Treatments for CD include, for example, sulfasalazine (Azulfadine), mesalamine (Asacol, Pentasa), balsalazide disodium (colazal®), azathioprine (Imuran), 6-MP (Purinethol), cyclosporine, methotrexate, infliximab
  • tests may be used by physicians to diagnose CD, including, barium enema, upper GI series, stool culture, blood tests to determine a white blood cell count or if anemia is present, fecal occult blood test, sigmoidoscopy, colonoscopy, and other tests may be used to rule out other potential diagnoses.
  • inflammatory bowel disease status refers to the status of the disease in the patient.
  • types of inflammatory bowel disease statuses include, but are not limited to, the subject's risk of IBD, including colorectal UC or CD, the presence or absence of disease (e.g., IBD, UC or CD), the stage of disease in a patient (e.g., IBD, UC or CD), and the effectiveness of treatment of disease.
  • Other statuses and degrees of each status are known in the art.
  • Gas phase ion spectrometer refers to an apparatus that detects gas phase ions.
  • Gas phase ion spectrometers include an ion source that supplies gas phase ions.
  • Gas phase ion spectrometers include, for example, mass spectrometers, ion mobility spectrometers, and total ion current measuring devices.
  • Gas phase ion spectrometry refers to the use of a gas phase ion spectrometer to detect gas phase ions.
  • Mass spectrometer refers to a gas phase ion spectrometer that measures a parameter that can be translated into mass-to-charge ratios of gas phase ions. Mass spectrometers generally include an ion source and a mass analyzer. Examples of mass spectrometers are time-of-flight, magnetic sector, quadrupole filter, ion trap, ion cyclotron resonance, electrostatic sector analyzer and hybrids of these. “Mass spectrometry” refers to the use of a mass spectrometer to detect gas phase ions.
  • Laser desorption mass spectrometer refers to a mass spectrometer that uses laser energy as a means to desorb, volatilize, and ionize an analyte.
  • Tandem mass spectrometer refers to any mass spectrometer that is capable of performing two successive stages of m/z-based discrimination or measurement of ions, including ions in an ion mixture.
  • the phrase includes mass spectrometers having two mass analyzers that are capable of performing two successive stages of m/z-based discrimination or measurement of ions tandem-in-space.
  • the phrase further includes mass spectrometers having a single mass analyzer that is capable of performing two successive stages of m/z- based discrimination or measurement of ions tandem-in-time.
  • Mass analyzer refers to a sub-assembly of a mass spectrometer that comprises means for measuring a parameter that can be translated into mass-to-charge ratios of gas phase ions.
  • the mass analyzer comprises an ion optic assembly, a flight tube and an ion detector.
  • Ion source refers to a sub-assembly of a gas phase ion spectrometer that provides gas phase ions.
  • the ion source provides ions through a desorption/ionization process.
  • Such embodiments generally comprise a probe interface that positionally engages a probe in an interrogatable relationship to a source of ionizing energy (e.g., a laser desorption/ionization source) and in concurrent communication at atmospheric or subatmospheric pressure with a detector of a gas phase ion spectrometer.
  • a source of ionizing energy e.g., a laser desorption/ionization source
  • ionizing energy for desorbing/ionizing an analyte from a solid phase include, for example: (1) laser energy; (2) fast atoms (used in fast atom bombardment); (3) high energy particles generated via beta decay of radionucleides (used in plasma desorption); and (4) primary ions generating secondary ions (used in secondary ion mass spectrometry).
  • the preferred form of ionizing energy for solid phase analytes is a laser (used in laser desorption/ionization), in particular, nitrogen lasers, Nd- Yag lasers and other pulsed laser sources. "Fluence" refers to the energy delivered per unit area of interrogated image.
  • a high fluence source such as a laser, will deliver about 1 mJ / mm2 to 50 mJ / mm2.
  • a sample is placed on the surface of a probe, the probe is engaged with the probe interface and the probe surface is struck with the ionizing energy. The energy desorbs analyte molecules from the surface into the gas phase and ionizes them.
  • Solid support refers to a solid material which can be derivatized with, or otherwise attached to, a capture reagent.
  • Exemplary solid supports include probes, microtiter plates and chromatographic resins.
  • Probe in the context of this invention refers to a device adapted to engage a probe interface of a gas phase ion spectrometer (e.g., a mass spectrometer) and to present an analyte to ionizing energy for ionization and introduction into a gas phase ion spectrometer, such as a mass spectrometer.
  • a “probe” will generally comprise a solid substrate (either flexible or rigid) comprising a sample presenting surface on which an analyte is presented to the source of ionizing energy.
  • “Surface-enhanced laser desorption/ionization” or “SELDI” refers to a method of desorption/ionization gas phase ion spectrometry (e.g., mass spectrometry) in which the analyte is captured on the surface of a SELDI probe that engages the probe interface of the gas phase ion spectrometer.
  • SELDI MS the gas phase ion spectrometer is a mass spectrometer.
  • SELDI technology is described in, e.g., U.S. patent 5,719,060 (Hutchens and Yip) and U.S. patent 6,225,047 (Hutchens and Yip).
  • SEEC Surface-Enhanced Affinity Capture
  • SELDI probe an absorbent surface
  • Adsorbent surface refers to a surface to which is bound an adsorbent (also called a “capture reagent” or an
  • an adsorbent is any material capable of binding an analyte (e.g., a target polypeptide or nucleic acid).
  • Chrographic adsorbent refers to a material typically used in chromatography.
  • Chromatographic adsorbents include, for example, ion exchange materials, metal chelators (e.g., nitriloacetic acid or iminodiacetic acid), immobilized metal chelates, hydrophobic interaction adsorbents, hydrophilic interaction adsorbents, dyes, simple biomolecules (e.g., nucleotides, amino acids, simple sugars and fatty acids) and mixed mode adsorbents (e.g., hydrophobic attraction/electrostatic repulsion adsorbents).
  • metal chelators e.g., nitriloacetic acid or iminodiacetic acid
  • immobilized metal chelates e.g., immobilized metal chelates
  • hydrophobic interaction adsorbents e.g., hydrophilic interaction adsorbents
  • dyes e.g., simple biomolecules (e.g., nucleotides, amino acids, simple sugars and
  • Biospecific adsorbent refers an adsorbent comprising a biomolecule, e.g., a nucleic acid molecule (e.g., an aptamer), a polypeptide, a polysaccharide, a lipid, a steroid or a conjugate of these (e.g., a glycoprotein, a lipoprotein, a glycolipid, a nucleic acid (e.g., DNA)-protein conjugate).
  • the biospecific adsorbent can be a macromolecular structure such as a multiprotein complex, a biological membrane or a virus. Examples of biospecific adsorbents are antibodies, receptor proteins and nucleic acids.
  • Biospecific adsorbents typically have higher specificity for a target analyte than chromatographic adsorbents. Further examples of adsorbents for use in SELDI can be found in U.S. Patent 6,225,047 (Hutchens and Yip, "Use of retentate chromatography to generate difference maps," May 1 , 2001).
  • a SEAC probe is provided as a pre-activated surface which can be modified to provide an adsorbent of choice.
  • certain probes are provided with a reactive moiety that is capable of binding a biological molecule through a covalent bond.
  • Epoxide and carbodiimidizole are useful reactive moieties to covalently bind biospecific adsorbents such as antibodies or cellular receptors.
  • Adsorption refers to detectable non-covalent binding of an analyte to an adsorbent or capture reagent.
  • SEND Surface-Enhanced Neat Desorption
  • SEND probe. "Energy absorbing molecules”
  • EAM Energy absorbing molecules
  • the phrase includes molecules used in MALDl , frequently referred to as “matrix”, and explicitly includes cinnamic acid derivatives, sinapinic acid (“SPA”), cyano-hydroxy-cinnamic acid (“CHCA”) and dihydroxybenzoic acid, ferulic acid, hydroxyacetophenone derivatives, as well as others. It also includes EAMs used in SELDI. SEND is further described in United States patent 5,719,060 and United States patent application 60/408,255, filed September 4, 2002 (Kitagawa, "Monomers And Polymers Having Energy Absorbing Moieties Of Use In Desorption/Ionization Of Analytes").
  • SEPAR Surface-Enhanced Photolabile Attachment and Release
  • SELDI Surface-Enhanced Photolabile Attachment and Release
  • SEPAR is a version of SELDI that involves the use of probes having moieties attached to the surface that can covalently bind an analyte, and then release the analyte through breaking a photolabile bond in the moiety after exposure to light, e.g., laser light.
  • SEPAR is further described in United States Patent 5,719,060.
  • Eluant or “wash solution” refers to an agent, typically a solution, which is used to affect or modify adsorption of an analyte to an adsorbent surface and/or remove unbound materials from the surface.
  • the elution characteristics of an eluant can depend on, for example, pH, ionic strength, hydrophobicity, degree of chaotropism, detergent strength and temperature.
  • Analyte refers to any component of a sample that is desired to be detected. The term can refer to a single component or a plurality of components in the sample.
  • the "complexity" of a sample adsorbed to an adsorption surface of an affinity capture probe means the number of different protein species that are adsorbed.
  • Molecular binding partners and “specific binding partners” refer to pairs of molecules, typically pairs of biomolecules that exhibit specific binding. Molecular binding partners include, without limitation, receptor and ligand, antibody and antigen, biotin and avidin, and biotin and streptavidin.
  • Monitoring refers to recording changes in a continuously varying parameter.
  • Biochip refers to a solid substrate having a generally planar surface to which an adsorbent is attached. Frequently, the surface of the biochip comprises a plurality of addressable locations, each of which location has the adsorbent bound there. Biochips can be adapted to engage a probe interface, and therefore, function as probes.
  • Protein biochip refers to a biochip adapted for the capture of polypeptides. Many protein biochips are described in the art. These include, for example, protein biochips produced by Ciphergen Biosystems (Fremont, CA), Packard BioScience Company (Meriden CT), Zyomyx (Hayward, CA) and Phylos (Lexington, MA).
  • Protein biochips produced by Ciphergen Biosystems comprise surfaces having chromatographic or biospecific adsorbents attached thereto at addressable locations. Biochips are further described in: WO 00/66265 (Rich et al., "Probes for a Gas Phase Ion Spectrometer," November 9, 2000); WO 00/67293 (Beecher et al., "Sample Holder with Hydrophobic Coating for Gas Phase Mass Spectrometer," November 9, 2000); U.S. patent application US20030032043A1 (Pohl and Papanu, "Latex Based Adsorbent Chip," July 16, 2002) and U.S. patent application 60/350,1 10 (Um et al., "Hydrophobic Surface Chip,” November 8, 2001).
  • analytes can be detected by a variety of detection methods selected from, for example, a gas phase ion spectrometry method, an optical method, an electrochemical method, atomic force microscopy and a radio frequency method.
  • Gas phase ion spectrometry methods are described herein. Of particular interest is the use of mass spectrometry, and in particular, SELDI.
  • Optical methods include, for example, detection of fluorescence, luminescence, chemiluminescence, absorbance, reflectance, transmittance, birefringence or refractive index (e.g., surface plasmon resonance, ellipsometry, a resonant mirror method, a grating coupler waveguide method or interferometry).
  • Optical methods include microscopy (both confocal and non-confocal), imaging methods and non-imaging methods.
  • Immunoassays in various formats e.g., ELISA
  • Electrochemical methods include voltametry and amperometry methods.
  • Radio frequency methods include multipolar resonance spectroscopy.
  • Biomarker or “biomarker” in the context of the present invention refer to a polypeptide (of a particular apparent molecular weight)or nucleic acid, which is differentially present in a sample taken from subjects having human inflammatory bowel disease as compared to a comparable sample taken from control subjects (e.g., a person with a negative diagnosis or undetectable inflammatory bowel disease, normal or healthy subject).
  • biomarker is used interchangeably with the term “marker.”
  • the biomarkers are identified by molecular mass in Daltons, and include the masses centered around the identified molecular masses for each marker.
  • measuring means methods which include detecting the presence or absence of marker(s) in the sample, quantifying the amount of marker(s) in the sample, and/or qualifying the type of biomarker. Measuring can be accomplished by methods known in the art and those further described herein, including but not limited to microarray analysis (with Significance Analysis of Microarrays (SAM) software), SELDI and immunoassay. Any suitable methods can be used to detect and measure one or more of the markers described herein. These methods include, without limitation, mass spectrometry (e.g. , laser desorption/ionization mass spectrometry), fluorescence (e.g. sandwich immunoassay), surface plasmon resonance, ellipsometry and atomic force microscopy. "Detect” refers to identifying the presence, absence or amount of the object to be detected.
  • mass spectrometry e.g. , laser desorption/ionization mass spectrometry
  • fluorescence e.g. sandwich immunoassay
  • surface plasmon resonance
  • a marker refers to differences in the quantity and/or the frequency of a marker present in a sample taken from subjects having human IBD as compared to a control subject. For example, some markers described herein are present at an elevated level in samples of subjects compared to samples from control subjects. In contrast, other markers described herein are present at a decreased level in samples of inflammatory bowel disease subjects compared to samples from control subjects. Furthermore, a marker can be a polypeptide, which is detected at a higher frequency or at a lower frequency in samples of human IBD subjects compared to samples of control subjects.
  • a marker can be a polypeptide, which is detected at a higher frequency or at a lower frequency in samples of unaffected tissue from human IBD subjects compared to samples affected tissue from human IBD subjects.
  • a marker can be a polypeptide, which is detected at a higher frequency or at a lower frequency in samples of human unaffected tissue from IBD subjects compared to samples of control subjects.
  • a marker can be a polypeptide, which is detected at a higher frequency or at a lower frequency in samples of human affected tissue from IBD subjects compared to samples of control subjects.
  • a marker can be differentially present in terms of quantity, frequency or both.
  • Affected tissue refers to tissue from and IBD subject that is grossly diseased tissue (tissue that is inflamed or shows fibrosis.
  • Unaffected tissue refers to a tissue from an IBD subject that is from a portion of tissue that does not have gross disease present, for example tissue that is about 1, 2, 5, 10, 20 or more cm from grossly diseased tissue.
  • a polypeptide is differentially present between two samples if the amount of the polypeptide in one sample is statistically significantly different from the amount of the polypeptide in the other sample.
  • a polypeptide is differentially present between the two samples if it is present at least about 120%, at least about 130%, at least about 150%, at least about 180%, at least about 200%, at least about 300%, at least about 500%, at least about 700%, at least about 900%, or at least about 1000% greater than it is present in the other sample, or if it is detectable in one sample and not detectable in the other.
  • a polypeptide is differentially present between two sets of samples if the frequency of detecting the polypeptide in the IBD subjects' samples is statistically significantly higher or lower than in the control samples.
  • a polypeptide is differentially present between the two sets of samples if it is detected at least about 120%, at least about 130%, at least about 150%, at least about 180%, at least about 200%, at least about 300%, at least about 500%, at least about 700%, at least about 900%, or at least about 1000% more frequently or less frequently observed in one set of samples than the other set of samples.
  • Diagnostic means identifying the presence or nature of a pathologic condition, i.e., inflammatory bowel disease. Diagnostic methods differ in their sensitivity and specificity.
  • the "sensitivity” of a diagnostic assay is the percentage of diseased individuals who test positive (percent of "true positives”). Diseased individuals not detected by the assay are “false negatives.” Subjects who are not diseased and who test negative in the assay, are termed “true negatives.”
  • the "specificity" of a diagnostic assay is 1 minus the false positive rate, where the "false positive” rate is defined as the proportion of those without the disease who test positive. While a particular diagnostic method may not provide a definitive diagnosis of a condition, it suffices if the method provides a positive indication that aids in diagnosis.
  • test amount of a marker refers to an amount of a marker present in a sample being tested.
  • a test amount can be either in absolute amount (e.g., ⁇ g/ml) or a relative amount (e.g. , relative intensity of signals).
  • a “diagnostic amount” of a marker refers to an amount of a marker in a subject's sample that is consistent with a diagnosis of inflammatory bowel disease.
  • a diagnostic amount can be either in absolute amount (e.g., ⁇ g/ml) or a relative amount (e.g., relative intensity of signals).
  • a "control amount" of a marker can be any amount or a range of amount, which is to be compared against a test amount of a marker.
  • a control amount of a marker can be the amount of a marker in a person without inflammatory bowel disease.
  • a control amount can be either in absolute amount (e.g., ⁇ g/ml) or a relative amount (e.g., relative intensity of signals).
  • the term "sensitivity" is the percentage of subjects with a particular disease.
  • the biomarkers of the invention have a sensitivity of about 80.0%-98.6%, and preferably a sensitivity of 85%, 87.5%, 90%, 92.5%, 95%, 97%, 98%, 99% or approaching 100%.
  • the term "specificity" is the percentage of subjects correctly identified as having a particular disease i.e., normal or healthy subjects. For example, the specificity is calculated as the number of subjects with a particular disease as compared to non-IBD subjects (e.g., normal healthy subjects).
  • the specificity of the assays described herein may range from about 80% to 100%. Preferably the specificity is about 90%, 95%, or 100%.
  • polypeptide refers to a polymer of amino acid residues.
  • the terms apply to amino acid polymers in which one or more amino acid residue is an analog or mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers.
  • Polypeptides can be modified, e.g., by the addition of carbohydrate residues to form glycoproteins.
  • polypeptide include glycoproteins, as well as non- glycoproteins.
  • immunoassay is an assay that uses an antibody to specifically bind an antigen (e.g., a marker). The immunoassay is characterized by the use of specific binding properties of a particular antibody to isolate, target, and/or quantify the antigen.
  • Antibody refers to a polypeptide ligand substantially encoded by an immunoglobulin gene or immunoglobulin genes, or fragments thereof, which specifically binds and recognizes an epitope ⁇ e.g., an antigen).
  • the recognized immunoglobulin genes include the kappa and lambda light chain constant region genes, the alpha, gamma, delta, epsilon and mu heavy chain constant region genes, and the myriad immunoglobulin variable region genes.
  • Antibodies exist, e.g., as intact immunoglobulins or as a number of well- characterized fragments produced by digestion with various peptidases. This includes, e.g., Fab 1 ' and F(ab) ! ' 2 fragments.
  • antibody also includes antibody fragments either produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA methodologies. It also includes polyclonal antibodies, monoclonal antibodies, chimeric antibodies, humanized antibodies, or single chain antibodies. "Fc" portion of an antibody refers to that portion of an immunoglobulin heavy chain that comprises one or more heavy chain constant region domains, CHi, CH 2 and CH 3 , but does not include the heavy chain variable region.
  • the specified antibodies bind to a particular protein at least two times the background and do not substantially bind in a significant amount to other proteins present in the sample.
  • Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity for a particular protein.
  • polyclonal antibodies raised to marker "X" from specific species such as rat, mouse, or human can be selected to obtain only those polyclonal antibodies that are specifically immunoreactive with marker "X” and not with other proteins, except for polymorphic variants and alleles of marker "X". This selection may be achieved by subtracting out antibodies that cross-react with marker "X" molecules from other species.
  • a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein.
  • solid-phase ELISA immunoassays are routinely used to select antibodies specifically immunoreactive with a protein ⁇ see, e.g., Harlow & Lane, Antibodies, A Laboratory Manual (1988), for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity).
  • a specific or selective reaction will be at least twice background signal or noise and more typically more than 10 to 100 times background.
  • Managing subject treatment refers to the behavior of the clinician or physician subsequent to the determination of IBD status. For example, if the result of the methods of the present invention is inconclusive or there is reason that confirmation of status is necessary, the physician may order more tests. Alternatively, if the status indicates that treatment is appropriate, the physician may schedule the patient for treatment, e.g., surgery, administer one or more therapeutic agents or radiation. Likewise, if the status is negative, e.g., late stage inflammatory bowel disease or if the status is acute, no further action may be warranted. Furthermore, if the results show that treatment has been successful, a maintenance therapy or no further management may be necessary.
  • CD biomarkers include the proteins or their encoding nucleic acids for the following pathways or cellular processes: acute phase and innate immune response(IL-l and TNF ⁇ mediated induction of NF- ⁇ B), immune response, apoptosis, inflammatory cell recruitment pathways, inflammatory response (IL]B 1 S100A8), antigen presentation (MHC class II immunoproteasome members PSME2 and PSMB8, MHC class II ATP-binding antigen peptide transporter TAPl, HLA-DMA and UBD of MHC class I), inflammatory cell chemotaxis (IL8, CXCLl, CXCL3), apoptosis (CASPl, CASPlO), macrophage activation (ASMT and interferon-regulated genes IFITMl, IFITM3, ISG20, IFB 5, SPIlO), leukocyte protection (LILRB encoding a receptor for class I MHC antigens), recruitment of inflammatory cells, acute phase response (ADM, STATl, STAT3, and protea
  • CD Crohn's disease patients often require surgery due to obstruction, when disease may be well established and gene expression patterns rather static.
  • Profiling of endoscopic biopsies provide the opportunity to interrogate all stages of disease.
  • Clinical sub grouping of CD is based on anatomic site of involvement (ileum only, colon only, or upper small bowel and colon) 12 and disease behavior (inflammatory, stricturing, or fistulizing). 13 ' 14
  • Pinch biopsies are collected during endoscopy for routine evaluation of disease activity by histology ' 5
  • single endoscopic pinch biopsies were used from nine colonic Crohn's disease cases with mild to severe inflammation, five ulcerative colitis cases and four healthy controls.
  • expression patterns for a biopsy from an affected and one from an unaffected area were obtained.
  • Multidimensional scaling of the expression patterns distinguished IBD from healthy individuals, CD from UC, and also unaffected from healthy controls.
  • Crohn's colitis harbors some phenotypic overlaps with ulcerative colitis, the expression profiles identify a distinct set of differentially expressed genes, and distinct pathophysiologies, for each disease.
  • UC Biomarkers include the proteins or their encoding nucleic acids for the following pathways or cellular processes: endoplasmic reticulum stress pathway members, protein- trafficking pathway members, and detoxification and cell growth pathway members.
  • UC biomarkers include the proteins or their encoding nucleic acids for the following pathways or cellular processes: up-regulations of complement cascade activation (BF and C4A), growth regulatory (MIA) and apoptosis (ATM) pathways, detoxification
  • BF and C4A up-regulations of complement cascade activation
  • MIA growth regulatory
  • ATM apoptosis
  • UC patterns are quite dynamic showing multiple gene expression changes (REGlA, LCN2, NOS2, NNMT, for example).
  • IBD biomarkers include both the UC and CD biomarkers (see Tables 1 - 3) as well as the following genes and nucleic acids and proteins encoded by the following genes, as well as fragments and variants thereof: CASPlO at 2q33-34, HLA-DMA, TAPl, UBD, PSMB8 at 6p21.3, and PSME2 at 14ql 1.2.
  • the sequences of these biomarkers are appended to the specification, as well as exemplary primers for amplifying the biomarkers.
  • Nine genes are elevated in most CD and UC affected profiles and most likely contribute towards separation of IBD from normal controls in the MDS plot.
  • genes include several chemokine ligands produced by activated monocytes and neutrophils, indicative of an immune/inflammation process and seem to correlate well with the inflammation scoring of the samples by histology (e.g., Group 3) Certain overlaps evident between the CD and the UC over expressed gene signatures
  • biomarkers for IBD include the proteins or their encoding nucleic acids for the following pathways or cellular processes: apoptosis -regulation (CASPlO, LILRB, 1 GNGTl (7q21.3)), antigen-presenting genes (PSME2), immunoproteasome for generating MHC class I binding antigenic peptides (IBD3, HLA-DMA, TAPl, UBD and PSMB8) , and Wnt-signaling (PRKACB (Ip36.1 , IBD7)).
  • apoptosis -regulation (CASPlO, LILRB, 1 GNGTl (7q21.3)
  • PSME2 antigen-presenting genes
  • IBD3, HLA-DMA, TAPl, UBD and PSMB8 immunoproteasome for generating MHC class I binding antigenic peptides
  • PRKACB Wnt-signaling
  • Corresponding proteins or fragments of proteins for these biomarkers may be represented as intensity peaks in SELDI (surface enhanced laser desorption/ionization) protein chip/mass spectra with molecular masses centered around the values.
  • Markers 1-75 also may be characterized based on affinity for an adsorbent, particularly binding to a cation-exchange or hydrophobic surface under the conditions specified in the Examples, which follow.
  • biomarkers are examples of biomarkers, as determined by identity, identified by the methods of the invention and serve merely as an illustrative example and are not meant to limit the invention in any way.
  • a major advantage of identification of these markers is their high specificity and ability to differentiate between different inflammatory bowel disease states (e.g., between UC and CD).
  • the present invention is based upon the discovery of protein markers that are differentially present in samples of human inflammatory bowel disease subjects and control subjects, and the application of this discovery in methods and kits for aiding a human inflammatory bowel disease diagnosis.
  • Some of these protein markers are found at an elevated level and/or more frequently in samples from human inflammatory bowel disease subjects compared to a control (e.g., subjects with diseases other than inflammatory bowel disease). Accordingly, the amount of one or more markers found in a test sample compared to a control, or the mere detection of one or more markers in the test sample provides useful information regarding probability of whether a subject being tested has inflammatory bowel disease or not, and/or whether a subject being tested has a particular inflammatory bowel disease subtype or not.
  • the protein of the present invention have a number of other uses.
  • the markers can be used to screen for compounds that modulate the expression of the markers in vitro or in vivo, which compounds in turn may be useful in treating or preventing human inflammatory bowel disease in subjects.
  • markers can be used to monitor responses to certain treatments of human inflammatory bowel disease.
  • the markers can be used in heredity studies. For instance, certain markers may be genetically linked. This can be determined by, e.g., analyzing samples from a population of human inflammatory bowel disease subjects whose families have a history of inflammatory bowel disease.
  • the results can then be compared with data obtained from, e.g., inflammatory bowel disease subjects whose families do not have a history of inflammatory bowel disease.
  • the markers that are genetically linked may be used as a tool to determine if a subject whose family has a history of inflammatory bowel disease is pre-disposed to having inflammatory bowel disease.
  • the invention provides methods for detecting markers which are differentially present in the samples of an inflammatory bowel disease patient and a control (e.g., subjects in non-inflammatory bowel disease subjects).
  • the markers can be detected in a number of biological samples.
  • the sample is preferably a biological biopsy sample. Any suitable methods can be used to detect one or more of the markers described herein.
  • mass spectrometry e.g., laser desorption/ionization mass spectrometry
  • fluorescence e.g. sandwich immunoassay
  • surface plasmon resonance e.g., ellipsometry
  • atomic force microscopy e.g., atomic force microscopy
  • Methods may further include, by one or more of microarrays, PCR methods, electrospray ionization mass spectrometry (ESI-MS), ESI-MS/MS, ESI-MS/(MS) n , matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS), surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF-MS), desorption/ionization on silicon (DIOS), secondary ion mass spectrometry (SIMS), quadrupole time-of-flight (Q-TOF), atmospheric pressure chemical ionization mass spectrometry (APCI-MS), APCI-MS/MS, APCI-(MS) 11 , atmospheric pressure photoionization mass spectrometry (APPI-MS), APPI-MS/MS, and APPI-(MS).sub.n, quadrupole mass spectrometry, four
  • a sample such as for example, serum from a subject or patient, is immobilized on a biochip.
  • the biochip comprises a functionalized, cross-linked polymer in the form of a hydrogel physically attached to the surface of the biochip or covalently attached through a silane to the surface of the biochip.
  • any biochip which can bind samples from subjects can be used.
  • the surfaces of the biochips are comprised of, for example, hydrophilic adsorbent to capture hydrophilic proteins (e.g.
  • carboimidizole functional groups that can react with groups on proteins for covalent binding
  • epoxide functional groups for covalent binding with proteins e.g. antibodies, receptors, lectins, heparin, Protein A, biotin/streptavidin and the like
  • anionic exchange groups e.g. antibodies, receptors, lectins, heparin, Protein A, biotin/streptavidin and the like
  • anionic exchange groups e.g. antibodies, receptors, lectins, heparin, Protein A, biotin/streptavidin and the like
  • anionic exchange groups e.g. antibodies, receptors, lectins, heparin, Protein A, biotin/streptavidin and the like
  • anionic exchange groups e.g. antibodies, receptors, lectins, heparin, Protein A, biotin/streptavidin and the like
  • anionic exchange groups e.g. antibodies, receptors,
  • samples are pre-fractionated prior to immobilization as discussed below.
  • Analytes or samples captured on the surface of a biochip can be detected by any method known in the art. This includes, for example, mass spectrometry, fluorescence, surface plasmon resonance, ellipsometry and atomic force microscopy.
  • Mass spectrometry, and particularly SELDI mass spectrometry is a particularly useful method for detection of the biomarkers of this invention.
  • Immobilized samples or analytes are preferably subjected to laser ionization and the intensity of signal for mass/charge ratio is detected.
  • the data obtained from the mass/charge ratio signal is transformed into data which is read by any type of computer.
  • An algorithm is executed by the computer user that classifies the data according to user input parameters for detecting signals that represent biomarkers present in, for example, inflammatory bowel disease subjects and are lacking in non-inflammatory bowel disease subject controls.
  • the biomarkers are most preferably identified by their molecular weights.
  • Samples are collected from subjects to establish inflammatory bowel disease status.
  • the subjects may be subjects who have been determined to have a high risk of inflammatory bowel disease based on their family history, a previous treatment, subjects with physical symptoms known to be associated with inflammatory bowel disease, subjects identified through screening assays (e.g., sigmoidoscopy) or rectal digital exam or rigid or flexible colonoscopy or CT scan or other x-ray techniques.
  • Other subjects include subjects who have inflammatory bowel disease and the test is being used to determine the effectiveness of therapy or treatment they are receiving.
  • subjects could include healthy people who are having a test as part of a routine examination, or to establish baseline levels of the biomarkers.
  • Samples may be collected from subjects who had been diagnosed with inflammatory bowel disease and received treatment to eliminate the inflammatory bowel disease, or perhaps are in remission. TYPES OF SAMPLE AND PREPARATION OF THE SAMPLE
  • the markers can be measured in different types of biological samples.
  • the sample is preferably a biological tissue or fluid sample.
  • biological tissue sample is a colon or intestinal biopsy sample, from for example a endoscopic examination.
  • a biological fluid sample useful in this invention include blood, blood serum, plasma, vaginal secretions, urine, tears, saliva, urine, tissue, cells, organs, seminal fluids, bone marrow, cerebrospinal fluid, etc. Because the markers are found in intestinal and/or colon tissue, these are preferred sample sources for embodiments of the invention.
  • Nucleic acids may be obtained from the samples in many ways known to one of skill in the art. For example, extraction methods, including for example, solvent extraction, affinity purification and centrifugation. Selective precipitation can also purify nucleic acids. Chromatography methods may also be utilized including, gel filtration, ion exchange, selective adsorption, or affinity binding.
  • the nucleic acids may be, for example, RNA, DNA or may be synthesized into cDNA.
  • the nucleic acids may be detected using microarray techniques that are well known in the art, for example, Affymetrix arrays followed by multidimensional scaling techniques. See R. Ekins and F.W. Chu, Microarrays: their origins and applications. Trends in Biotechnology, 1999, 17, 217-218; D. D. Shoemaker, et al., Experimental annotation of the human genome using microarray technology, Nature Volume 409 Number 6822 Page 922 - 927 (2001) and US Patent 5,750,015.
  • the markers can be resolved in a sample by using a variety of techniques, e.g., nucleic acid chips, PCR, real time PCR, reverse transcriptase PCR, real time reverse transcriptase PCR, in situ PCR, chromatographic separation coupled with mass spectrometry, protein capture using immobilized antibodies or by traditional immunoassays.
  • a variety of techniques e.g., nucleic acid chips, PCR, real time PCR, reverse transcriptase PCR, real time reverse transcriptase PCR, in situ PCR, chromatographic separation coupled with mass spectrometry, protein capture using immobilized antibodies or by traditional immunoassays.
  • Biomarker expression may also be by PCR methods, including for example, real time PCR. See for example, U.S. Patents 5,723,591; 5,801 ,155 and 6,084,102 and Higuchi, 1992 and 1993.
  • PCR assays may be done, for example, in a multi-well plate formats or in chips, such as the BioTrove OpenArrayTM Chips (BioTrove, Woburn, MA). If desired, the sample can be prepared to enhance detectability of the markers.
  • a blood serum sample from the subject can be preferably fractionated by, e.g., Cibacron blue agarose chromatography and single stranded DNA affinity chromatography, anion exchange chromatography, affinity chromatography (e.g., with antibodies) and the like.
  • the method of fractionation depends on the type of detection method used. Any method that enriches for the protein of interest can be used.
  • preparation involves fractionation of the sample and collection of fractions determined to contain the biomarkers. Methods of pre-fractionation include, for example, size exclusion chromatography, ion exchange chromatography, heparin chromatography, affinity chromatography, sequential extraction, gel electrophoresis and liquid chromatography.
  • the analytes also may be modified prior to detection. These methods are useful to simplify the sample for further analysis. For example, it can be useful to remove high abundance proteins, such as albumin, from blood before analysis.
  • a sample can be pre-fractionated according to size of proteins in a sample using size exclusion chromatography.
  • a size selection spin column is used for a biological sample wherein the amount of sample available is small.
  • a K30 spin column available from Princeton Separation, Ciphergen Biosystems, Inc., etc. can be used.
  • fraction 1 the first fraction that is eluted from the column
  • fraction 2 has a lower percentage of high molecular weight proteins
  • fraction 3 has even a lower percentage of high molecular weight proteins
  • fraction 4 has the lowest amount of large proteins; and so on.
  • Each fraction can then be analyzed by gas phase ion spectrometry for the detection of markers.
  • a sample can be pre-fractionated by anion exchange chromatography.
  • Anion exchange chromatography allows pre-fractionation of the proteins in a sample roughly according to their charge characteristics.
  • a Q anion-exchange resin can be used ⁇ e.g , Q HyperD F, Biosepra), and a sample can be sequentially eluted with eluants having different pH's.
  • Anion exchange chromatography allows separation of biomolecules in a sample that are more negatively charged from other types of biomolecules. Proteins that are eluted with an eluant having a high pH is likely to be weakly negatively charged, and a fraction that is eluted with an eluant having a low pH is likely to be strongly negatively charged.
  • anion exchange chromatography separates proteins according to their binding characteristics.
  • a sample can be pre-fractionated by heparin chromatography.
  • Heparin chromatography allows pre-fractionation of the markers in a sample also on the basis of affinity interaction with heparin and charge characteristics.
  • Heparin, a sulfated mucopolysaccharide will bind markers with positively charged moieties and a sample can be sequentially eluted with eluants having different pH's or salt concentrations. Markers eluted with an eluant having a low pH are more likely to be weakly positively charged. Markers eluted with an eluant having a high pH are more likely to be strongly positively charged.
  • heparin chromatography also reduces the complexity of a sample and separates markers according to their binding characteristics.
  • a sample can be pre-fractionated by removing proteins that are present in a high quantity or that may interfere with the detection of markers in a sample.
  • serum albumin is present in a high quantity and may obscure the analysis of markers.
  • a blood serum sample can be pre- fractionated by removing serum albumin.
  • Serum albumin can be removed using a substrate that comprises adsorbents that specifically bind serum albumin.
  • a column which comprises, e.g. , Cibacron blue agarose (which has a high affinity for serum albumin) or anti-serum albumin antibodies can be used.
  • a sample can be pre-fractionated by isolating proteins that have a specific characteristic, e.g. are glycosylated.
  • a blood serum sample can be fractionated by passing the sample over a lectin chromatography column (which has a high affinity for sugars). Glycosylated proteins will bind to the lectin column and non- glycosylated proteins will pass through the flow through. Glycosylated proteins are then eluted from the lectin column with an eluant containing a sugar, e.g., N-acetyl-glucosamine and are available for further analysis.
  • a sugar e.g., N-acetyl-glucosamine
  • affinity adsorbents which are suitable for pre-fractionating blood serum samples.
  • An example of one other type of affinity chromatography available to pre- fractionate a sample is a single stranded DNA spin column. These columns bind proteins which are basic or positively charged. Bound proteins are then eluted from the column using eluants containing denaturants or high pH.
  • a sample can be fractionated using a sequential extraction protocol.
  • sequential extraction a sample is exposed to a series of adsorbents to extract different types of biomolecules from a sample. For example, a sample is applied to a first adsorbent to extract certain proteins, and an eluant containing non-adsorbent proteins (i.e., proteins that did not bind to the first adsorbent) is collected. Then, the fraction is exposed to a second adsorbent. This further extracts various proteins from the fraction. This second fraction is then exposed to a third adsorbent, and so on. Any suitable materials and methods can be used to perform sequential extraction of a sample.
  • a series of spin columns comprising different adsorbents can be used.
  • a multi-well comprising different adsorbents at its bottom can be used.
  • sequential extraction can be performed on a probe adapted for use in a gas phase ion spectrometer, wherein the probe surface comprises adsorbents for binding biomolecules.
  • the sample is applied to a first adsorbent on the probe, which is subsequently washed with an eluant. Markers that do not bind to the first adsorbent is removed with an eluant.
  • the markers that are in the fraction can be applied to a second adsorbent on the probe, and so forth.
  • biomolecules in a sample can be separated by high- resolution electrophoresis, e.g., one or two-dimensional gel electrophoresis.
  • a fraction containing a marker can be isolated and further analyzed by gas phase ion spectrometry.
  • two-dimensional gel electrophoresis is used to generate two-dimensional array of spots of biomolecules, including one or more markers. See, e.g., Jungblut and Thiede, Mass Spectr. Rev. 16: 145-162 (1997).
  • the two-dimensional gel electrophoresis can be performed using methods known in the art. See, e.g., Guider ed., Methods In Enzymology vol. 182.
  • biomolecules in a sample are separated by, e.g., isoelectric focusing, during which biomolecules in a sample are separated in a pH gradient until they reach a spot where their net charge is zero (i.e., isoelectric point).
  • This first separation step results in one-dimensional array of biomolecules.
  • the biomolecules in one- dimensional array is further separated using a technique generally distinct from that used in the first separation step.
  • biomolecules separated by isoelectric focusing are further separated using a polyacrylamide gel, such as polyacrylamide gel electrophoresis in the presence of sodium dodecyl sulfate (SDS-PAGE).
  • SDS-PAGE gel allows further separation based on molecular mass of biomolecules.
  • two-dimensional gel electrophoresis can separate chemically different biomolecules in the molecular mass range from 1000-200,000 Da within complex mixtures.
  • Biomolecules in the two-dimensional array can be detected using any suitable methods known in the art.
  • biomolecules in a gel can be labeled or stained (e.g., Coomassie Blue or silver staining).
  • the spot can be is further analyzed by gas phase ion spectrometry.
  • spots can be excised from the gel and analyzed by gas phase ion spectrometry.
  • the gel containing biomolecules can be transferred to an inert membrane by applying an electric field. Then a spot on the membrane that approximately corresponds to the molecular weight of a marker can be analyzed by gas phase ion spectrometry.
  • the spots can be analyzed using any suitable techniques, such as MALDI or SELDI (e.g., using ProteinChip " array) as described in detail below.
  • cleaving reagents such as proteases (e.g., trypsin).
  • the digestion of biomolecules into small fragments provides a mass fingerprint of the biomolecules in the spot, which can be used to determine the identity of markers if desired.
  • HPLC high performance liquid chromatography
  • HPLC instruments typically consist of a reservoir of mobile phase, a pump, an injector, a separation column, and a detector. Biomolecules in a sample are separated by injecting an aliquot of the sample onto the column. Different biomolecules in the mixture pass through the column at different rates due to differences in their partitioning behavior between the mobile liquid phase and the stationary phase. A fraction that corresponds to the molecular weight and/or physical properties of one or more markers can be collected. The fraction can then be analyzed by gas phase ion spectrometry to detect markers. For example, the spots can be analyzed using either MALDI or SELDI (e.g., using ProteinChip array) as described in detail below.
  • a marker can be modified before analysis to improve its resolution or to determine its identity.
  • the markers may be subject to proteolytic digestion before analysis. Any protease can be used. Proteases, such as trypsin, that are likely to cleave the markers into a discrete number of fragments are particularly useful. The fragments that result from digestion function as a fingerprint for the markers, thereby enabling their detection indirectly. This is particularly useful where there are markers with similar molecular masses that might be confused for the marker in question. Also, proteolytic fragmentation is useful for high molecular weight markers because smaller markers are more easily resolved by mass spectrometry.
  • biomolecules can be modified to improve detection resolution.
  • neuraminidase can be used to remove terminal sialic acid residues from glycoproteins to improve binding to an anionic adsorbent (e.g., cationic exchange ProteinChip ® arrays) and to improve detection resolution.
  • the markers can be modified by the attachment of a tag of particular molecular weight that specifically bind to molecular markers, further distinguishing them.
  • the identity of the markers can be further determined by matching the physical and chemical characteristics of the modified markers in a protein database (e.g., SwissProt).
  • any suitable method can be used to measure a marker or markers in a sample.
  • markers can be detected and/or measured by a variety of detection methods including for example, gas phase ion spectrometry methods, optical methods, electrochemical methods, atomic force microscopy, radio frequency methods, surface plasmon resonance, ellipsometry and atomic force microscopy.
  • SELDI Surface-enhanced laser desorption/ionization
  • SELDI refers to a method of desorption/ionization gas phase ion spectrometry (e.g., mass spectrometry) in which the analyte is captured on the surface of a SELDI probe that engages the probe interface.
  • gas phase ion spectrometer is a mass spectrometer. SELDI technology is described in more detail above and as follows.
  • a laser desorption time-of-flight mass spectrometer is used in embodiments of the invention.
  • a substrate or a probe comprising markers is introduced into an inlet system.
  • the markers are desorbed and ionized into the gas phase by laser from the ionization source.
  • the ions generated are collected by an ion optic assembly, and then in a time-of-flight mass analyzer, ions are accelerated through a short high voltage field and let drift into a high vacuum chamber. At the far end of the high vacuum chamber, the accelerated ions strike a sensitive detector surface at a different time.
  • the elapsed time between ion formation and ion detector impact can be used to identify the presence or absence of markers of specific mass to charge ratio.
  • Markers on the substrate surface can be desorbed and ionized using gas phase ion spectrometry. Any suitable gas phase ion spectrometers can be used as long as it allows markers on the substrate to be resolved. Preferably, gas phase ion spectrometers allow quantitation of markers.
  • a gas phase ion spectrometer is a mass spectrometer.
  • a substrate or a probe comprising markers on its surface is introduced into an inlet system of the mass spectrometer.
  • the markers are then desorbed by a desorption source such as a laser, fast atom bombardment, high energy plasma, electrospray ionization, thermospray ionization, liquid secondary ion MS, field desorption, etc.
  • the generated desorbed, volatilized species consist of preformed ions or neutrals which are ionized as a direct consequence of the desorption event.
  • Generated ions are collected by an ion optic assembly, and then a mass analyzer disperses and analyzes the passing ions. The ions exiting the mass analyzer are detected by a detector.
  • the detector then translates information of the detected ions into mass-to-charge ratios. Detection of the presence of markers or other substances will typically involve detection of signal intensity. This, in turn, can reflect the quantity and character of markers bound to the substrate.
  • a mass spectrometer e.g., a desorption source, a mass analyzer, a detector, etc.
  • a laser desorption time-of-flight mass spectrometer is used in embodiments of the invention.
  • a substrate or a probe comprising markers is introduced into an inlet system.
  • the markers are desorbed and ionized into the gas phase by laser from the ionization source.
  • the ions generated are collected by an ion optic assembly, and then in a time-of-flight mass analyzer, ions are accelerated through a short high voltage field and let drift into a high vacuum chamber. At the far end of the high vacuum chamber, the accelerated ions strike a sensitive detector surface at a different time. Since the time-of-flight is a function of the mass of the ions, the elapsed time between ion formation and ion detector impact can be used to identify the presence or absence of markers of specific mass to charge ratio. In another embodiment, an ion mobility spectrometer can be used to detect markers.
  • ion mobility spectrometry is based on different mobility of ions. Specifically, ions of a sample produced by ionization move at different rates, due to their difference in, e.g. , mass, charge, or shape, through a tube under the influence of an electric field. The ions (typically in the form of a current) are registered at the detector which can then be used to identify a marker or other substances in a sample.
  • ions of a sample produced by ionization move at different rates, due to their difference in, e.g. , mass, charge, or shape, through a tube under the influence of an electric field.
  • the ions typically in the form of a current
  • One advantage of ion mobility spectrometry is that it can operate at atmospheric pressure.
  • a total ion current measuring device can be used to detect and characterize markers. This device can be used when the substrate has a only a single type of marker. When a single type of marker is on the substrate, the total current generated from the ionized marker reflects the quantity and other characteristics of the marker. The total ion current produced by the marker can then be compared to a control (e.g., a total ion current of a known compound). The quantity or other characteristics of the marker can then be determined.
  • a control e.g., a total ion current of a known compound
  • an immunoassay can be used to detect and analyze markers in a sample. This method comprises: (a) providing an antibody that specifically binds to a marker; (b) contacting a sample with the antibody; and (c) detecting the presence of a complex of the antibody bound to the marker in the sample.
  • An immunoassay is an assay that uses an antibody to specifically bind an antigen (e.g., a marker).
  • the immunoassay is characterized by the use of specific binding properties of a particular antibody to isolate, target, and/or quantify the antigen.
  • the phrase "specifically (or selectively) binds" to an antibody or “specifically (or selectively) immunoreactive with,” when referring to a protein or peptide, refers to a binding reaction that is determinative of the presence of the protein in a heterogeneous population of proteins and other biologies.
  • the specified antibodies bind to a particular protein at least two times the background and do not substantially bind in a significant amount to other proteins present in the sample.
  • Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity for a particular protein.
  • polyclonal antibodies raised to a marker from specific species such as rat, mouse, or human can be selected to obtain only those polyclonal antibodies that are specifically immunoreactive with that marker and not with other proteins, except for polymorphic variants and alleles of the marker. This selection may be achieved by subtracting out antibodies that cross-react with the marker molecules from other species.
  • antibodies that specifically bind to a marker can be prepared using any suitable methods known in the art. See, e.g., Coligan, Current Protocols in Immunology (1991); Harlow & Lane, Antibodies: A Laboratory Manual (1988); Goding, Monoclonal Antibodies : Principles and Practice (2d ed. 1986); and Kohler & Milstein, Nature 256:495-497 (1975).
  • Such techniques include, but are not limited to, antibody preparation by selection of antibodies from libraries of recombinant antibodies in phage or similar vectors, as well as preparation of polyclonal and monoclonal antibodies by immunizing rabbits or mice ⁇ see, e.g., Huse et ai, Science 246: 1275-1281 (1989); Ward et ah, Nature 341 :544-546 (1989)).
  • a specific or selective reaction will be at least twice background signal or noise and more typically more than 10 to 100 times background.
  • a sample obtained from a subject can be contacted with the antibody that specifically binds the marker.
  • the antibody can be fixed to a solid support to facilitate washing and subsequent isolation of the complex, prior to contacting the antibody with a sample.
  • solid supports include glass or plastic in the form of, e.g., a microtiter plate, a stick, a bead, or a microbead.
  • Antibodies can also be attached to a probe substrate or ProteinChip ® array described above.
  • the sample is preferably a biological fluid sample taken from a subject.
  • biological fluid samples include blood, serum, plasma, nipple aspirate, urine, tears, saliva etc.
  • the biological fluid comprises blood serum.
  • the sample can be diluted with a suitable eluant before contacting the sample to the antibody.
  • the mixture is washed and the antibody- marker complex formed can be detected.
  • This detection reagent may be, e.g., a second antibody which is labeled with a detectable label.
  • detectable labels include magnetic beads ⁇ e.g., DYNABEADS T ), fluorescent dyes, radiolabels, enzymes ⁇ e.g., horse radish peroxide, alkaline phosphatase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold or colored glass or plastic beads.
  • the marker in the sample can be detected using an indirect assay, wherein, for example, a second, labeled antibody is used to detect bound marker-specific antibody, and/or in a competition or inhibition assay wherein, for example, a monoclonal antibody which binds to a distinct epitope of the marker is incubated simultaneously with the mixture.
  • an indirect assay wherein, for example, a second, labeled antibody is used to detect bound marker-specific antibody, and/or in a competition or inhibition assay wherein, for example, a monoclonal antibody which binds to a distinct epitope of the marker is incubated simultaneously with the mixture.
  • Methods for measuring the amount of, or presence of, antibody-marker complex include, for example, detection of fluorescence, luminescence, chemiluminescence, absorbance, reflectance, transmittance, birefringence or refractive index (e.g., surface plasmon resonance, ellipsometry, a resonant mirror method, a grating coupler waveguide method or interferometry).
  • Optical methods include microscopy (both confocal and non- confocal), imaging methods and non-imaging methods.
  • Electrochemical methods include voltametry and amperometry methods.
  • Radio frequency methods include multipolar resonance spectroscopy. Methods for performing these assays are readily known in the art.
  • Useful assays include, for example, an enzyme immune assay (EIA) such as en2yme-linked immunosorbent assay (ELISA), a radioimmune assay (RIA), a Western blot assay, or a slot blot assay.
  • EIA enzyme immune assay
  • ELISA en2yme-linked immunosorbent assay
  • RIA radioimmune assay
  • Western blot assay e.g., Western blot assay, or a slot blot assay.
  • incubation and/or washing steps may be required after each combination of reagents. Incubation steps can vary from about 5 seconds to several hours, preferably from about 5 minutes to about 24 hours. However, the incubation time will depend upon the assay format, marker, volume of solution, concentrations and the like. Usually the assays will be carried out at ambient temperature, although they can be conducted over a range of temperatures, such as 10 0 C to 40 0 C.
  • Immunoassays can be used to determine presence or absence of a marker in a sample as well as the quantity of a marker in a sample.
  • the amount of an antibody-marker complex can be determined by comparing to a standard.
  • a standard can be, e.g., a known compound or another protein known to be present in a sample.
  • the test amount of marker need not be measured in absolute units, as long as the unit of measurement can be compared to a control.
  • the methods for detecting these markers in a sample have many applications. For example, one or more markers can be measured to aid humaninflammatory bowel disease diagnosis or prognosis. In another example, the methods for detection of the markers can be used to monitor responses in a subject to inflammatory bowel disease treatment. In another example, the methods for detecting markers can be used to assay for and to identify compounds that modulate expression of these markers in vivo or in vitro. In a preferred example, the biomarkers are used to differentiate between the different stages of tumor progression, thus aiding in determining appropriate treatment and extent of metastasis of the tumor. USE OF MODIFIED FORMS OF A BIOMARKER
  • Pre-translational modified forms include allelic variants, slice variants and RNA editing forms.
  • Post-translationally modified forms include forms resulting from proteolytic cleavage (e.g., fragments of a parent protein), glycosylation, phosphorylation, lipidation, oxidation, methylation, cystinylation, sulphonation and acetylation.
  • modified protein cluster The collection of proteins including a specific protein and all modified forms of it is referred to herein as a "protein cluster.”
  • Modified forms of any biomarker of this invention (including any of Markers I through XIII) also may be used, themselves, as biomarkers. In certain cases the modified forms may exhibit better discriminatory power in diagnosis than the specific forms set forth herein.
  • Modified forms of a biomarker including any of Markers 1 -75 can be initially detected by any methodology that can detect and distinguish the modified from the biomarker.
  • a preferred method for initial detection involves first capturing the biomarker and modified forms of it, e.g., with biospecific capture reagents, and then detecting the captured proteins by mass spectrometry. More specifically, the proteins are captured using biospecific capture reagents, such as antibodies, aptamers or Affibodies that recognize the biomarker and modified forms of it. This method also will also result in the capture of protein interactors that are bound to the proteins or that are otherwise recognized by antibodies and that, themselves, can be biomarkers. Preferably, the biospecific capture reagents are bound to a solid phase.
  • the captured proteins can be detected by SELDI mass spectrometry or by eluting the proteins from the capture reagent and detecting the eluted proteins by traditional MALDI or by SELDI.
  • SELDI mass spectrometry is especially attractive because it can distinguish and quantify modified forms of a protein based on mass and without the need for labeling.
  • the biospecific capture reagent is bound to a solid phase, such as a bead, a plate, a membrane or a chip.
  • a solid phase such as a bead, a plate, a membrane or a chip.
  • Methods of coupling biomolecules, such as antibodies, to a solid phase are well known in the art. They can employ, for example, bifunctional linking agents, or the solid phase can be derivatized with a reactive group, such as an epoxide or an imidizole, that will bind the molecule on contact.
  • Biospecific capture reagents against different target proteins can be mixed in the same place, or they can be attached to solid phases in different physical or addressable locations. For example, one can load multiple columns with derivatized beads, each column able to capture a single protein cluster.
  • antibody-derivatized bead-based technologies such as xMAP technology of Luminex (Austin, TX) can be used to detect the protein clusters.
  • the biospecific capture reagents must be specifically directed toward the members of a cluster in order to differentiate them.
  • the surfaces of biochips can be derivatized with the capture reagents directed against protein clusters either in the same location or in physically different addressable locations.
  • the modified form can be used as a biomarker in any of the methods of this invention.
  • detection of the modified from can be accomplished by any specific detection methodology including affinity capture followed by mass spectrometry, or traditional immunoassay directed specifically the modified form.
  • Immunoassay requires biospecific capture reagents, such as antibodies, to capture the analytes.
  • the assay must be designed to specifically distinguish protein and modified forms of protein. This can be done, for example, by employing a sandwich assay in which one antibody captures more than one form and second, distinctly labeled antibodies, specifically bind, and provide distinct detection of, the various forms.
  • Antibodies can be produced by immunizing animals with the biomolecules.
  • This invention contemplates traditional immunoassays including, for example, sandwich immunoassays including ELISA or fluorescence-based immunoassays, as well as other enzyme immunoassays.
  • the methods for detecting these markers in a sample have many applications. For example, one or more markers can be measured to aid human inflammatory bowel disease diagnosis or prognosis. In another example, the methods for detection of the markers can be used to monitor responses in a subject to inflammatory bowel disease treatment. In another example, the methods for detecting markers can be used to assay for and to identify compounds that modulate expression of these markers in vivo or in vitro.
  • Non-inflammatory bowel disease and inflammatory bowel disease status may be by the detection of one or more of the Markers listed in Tables 1 - 3 or the Markers described as proteins or pathways for IBD, UC, or CD.
  • an exemplary marker that may independently discriminate between colorectal and non-colorectal status is Markers 1- 75.
  • Combinations of markers are also useful in the methods of the invention for the discrimination of on-inflammatory bowel disease and inflammatory bowel disease status, for example, Markers may also be used to discriminate or distinguish or diagnose between UC and CD and between unaffected and affected tissue of a UC and/or CD subject.
  • Markers may be detected, determined, monitored in a sample by molecular biological methods, including, arrays (nucleic acid, protein), PCR methods (real-time, reverse transcriptase, PCR).
  • Detection of markers can be analyzed using any suitable means, including arrays.
  • Nucleic acid arrays may be analyzed using software, for example, Applied Maths, Belgium.
  • GenExploreTM 2-way cluster analysis, principal component analysis, discriminant analysis, self-organizing maps; BioDiscovery, Inc., Los Angeles, California (ImaGeneTM, special image processing and data extraction software, powered by MatLab®; GeneSight: hierarchical clustering, artificial neural network (SOM?), principal component analysis, time series; AutoGeneTM; CloneTrackerTM); GeneData AG (Basel, Switzerland); Molecular Pattern Recognition web site at MIT's Whitehead Genome Center; Rosetta Inpharmatics, Kirkland, Washington.
  • Detection of markers can be analyzed using any suitable means.
  • data generated, for example, by desorption is analyzed with the use of a programmable digital computer.
  • the computer program generally contains a readable medium that stores codes. Certain code can be devoted to memory that includes the location of each feature on a probe, the identity of the adsorbent at that feature and the elution conditions used to wash the adsorbent.
  • the computer also contains code that receives as input, data on the strength of the signal at various molecular masses received from a particular addressable location on the probe. This data can indicate the number of markers detected, including the strength of the signal generated by each marker.
  • Data analysis can include the steps of determining signal strength (e.g., height of peaks) of a marker detected and removing "outliers" (data deviating from a predetermined statistical distribution).
  • the observed peaks can be normalized, a process whereby the height of each peak relative to some reference is calculated.
  • a reference can be background noise generated by instrument and chemicals (e.g., energy absorbing molecule) which is set as zero in the scale.
  • the signal strength detected for each marker or other biomolecules can be displayed in the form of relative intensities in the scale desired (e.g., 100).
  • a standard e.g., a serum protein
  • a standard may be admitted with the sample so that a peak from the standard can be used as a reference to calculate relative intensities of the signals observed for each marker or other markers detected.
  • the computer can transform the resulting data into various formats for displaying.
  • spectrum view or retentate map a standard spectral view can be displayed, wherein the view depicts the quantity of marker reaching the detector at each particular molecular weight.
  • peak map a standard spectral view
  • peak map only the peak height and mass information are retained from the spectrum view, yielding a cleaner image and enabling markers with nearly identical molecular weights to be more easily seen.
  • gel view each mass from the peak view can be converted into a grayscale image based on the height of each peak, resulting in an appearance similar to bands on electrophoretic gels.
  • 3-D overlays In yet another format, referred to as "3-D overlays,” several spectra can be overlaid to study subtle changes in relative peak heights.
  • difference map view two or more spectra can be compared, conveniently highlighting unique markers and markers which are up- or down-regulated between samples. Marker profiles (spectra) from any two samples may be compared visually.
  • Spotf ⁇ re Scatter Plot can be used, wherein markers that are detected are plotted as a dot in a plot, wherein one axis of the plot represents the apparent molecular of the markers detected and another axis represents the signal intensity of markers detected.
  • markers that are detected and the amount of markers present in the sample can be saved in a computer readable medium. This data can then be compared to a control (e.g., a profile or quantity of markers detected in control, e.g., men in whom human inflammatory bowel disease is undetectable).
  • a control e.g., a profile or quantity of markers detected in control, e.g., men in whom human inflammatory bowel disease is undetectable.
  • the software can comprise code that converts signal from the mass spectrometer into computer readable form.
  • the software also can include code that applies an algorithm to the analysis of the signal to determine whether the signal represents a "peak" in the signal corresponding to a marker of this invention, or other useful markers.
  • the software also can include code that executes an algorithm that compares signal from a test sample to a typical signal characteristic of "normal” and human IBD and determines the closeness of fit between the two signals.
  • the software also can include code indicating which the test sample is closest to, thereby providing a probable diagnosis.
  • multiple biomarkers are measured.
  • the use of multiple biomarkers increases the predictive value of the test and provides greater utility in diagnosis, toxicology, patient stratification and patient monitoring.
  • the process called "Pattern recognition" detects the patterns formed by multiple biomarkers greatly improves the sensitivity and specificity of clinical proteomics for predictive medicine.
  • Subtle variations in data from clinical samples e.g., obtained using SELDI, indicate that certain patterns of protein expression can predict phenotypes such as the presence or absence of a certain disease, a particular stage of IBD-progression, or a positive or adverse response to drug treatments.
  • Data generation in mass spectrometry begins with the detection of ions by an ion detector as described above.
  • Ciphergen's ProteinChip system employs an analog-to-digital converter (ADC) to accomplish this.
  • ADC analog-to-digital converter
  • the ADC integrates detector output at regularly spaced time intervals into time-dependent bins. The time intervals typically are one to four nanoseconds long.
  • time-of-flight spectrum ultimately analyzed typically does not represent the signal from a single pulse of ionizing energy against a sample, but rather the sum of signals from a number of pulses. This reduces noise and increases dynamic range.
  • This time-of- flight data is then subject to data processing.
  • data processing typically includes TOF-to-M/Z transformation, baseline subtraction, high frequency noise filtering.
  • TOF-to-M/Z transformation involves the application of an algorithm that transforms times-of-flight into mass-to-charge ratio (M/Z).
  • M/Z mass-to-charge ratio
  • the signals are converted from the time domain to the mass domain. That is, each time-of-flight is converted into mass-to- charge ratio, or M/Z.
  • Calibration can be done internally or externally.
  • the sample analyzed contains one or more analytes of known M/Z. Signal peaks at times-of- flight representing these massed analytes are assigned the known M/Z. Based on these assigned M/Z ratios, parameters are calculated for a mathematical function that converts times-of-flight to M/Z.
  • a function that converts times-of-flight to M/Z such as one created by prior internal calibration, is applied to a time-of-flight spectrum without the use of internal calibrants.
  • Baseline subtraction improves data quantification by eliminating artificial, reproducible instrument offsets that perturb the spectrum. It involves calculating a spectrum baseline using an algorithm that incorporates parameters such as peak width, and then subtracting the baseline from the mass spectrum.
  • the moving average filter is a variable width digital filter in which the bandwidth of the filter varies as a function of, e.g., peak bandwidth, generally becoming broader with increased time-of-flight. See, e.g., WO 00/70648, November 23, 2000 (Gavin et al., "Variable Width Digital Filter for Time-of-flight Mass Spectrometry"). Analysis generally involves the identification of peaks in the spectrum that represent signal from an analyte. Peak selection can, of course, be done by eye.
  • Ciphergen's ProteinChip® software that can automate the detection of peaks.
  • this software functions by identifying signals having a signal-to-noise ratio above a selected threshold and labeling the mass of the peak at the centroid of the peak signal.
  • many spectra are compared to identify identical peaks present in some selected percentage of the mass spectra.
  • One version of this software clusters all peaks appearing in the various spectra within a defined mass range, and assigns a mass (M/Z) to all the peaks that are near the mid-point of the mass (M/Z) cluster.
  • Peak data from one or more spectra can be subject to further analysis by, for example, creating a spreadsheet in which each row represents a particular mass spectrum, each column represents a peak in the spectra defined by mass, and each cell includes the intensity of the peak in that particular spectrum.
  • Various statistical or pattern recognition approaches can applied to the data.
  • the spectra that are generated in embodiments of the invention can be classified using a pattern recognition process that uses a classification model.
  • data derived from the spectra e.g., mass spectra or time-of-flight spectra
  • samples such as "known samples”
  • a "known sample” is a sample that is pre-classified (e.g., inflammatory bowel disease or not inflammatory bowel disease).
  • Data derived from the spectra (e.g., mass spectra or time-of- flight spectra) that are generated using samples such as "known samples” can then be used to "train” a classification model.
  • a "known sample” is a sample that is pre -classified.
  • the data that are derived from the spectra and are used to form the classification model can be referred to as a "training data set”.
  • the classification model can recognize patterns in data derived from spectra generated using unknown samples.
  • the classification model can then be used to classify the unknown samples into classes. This can be useful, for example, in predicting whether or not a particular biological sample is associated with a certain biological condition (e.g., diseased vs. non diseased).
  • the training data set that is used to form the classification model may comprise raw data or pre-processed data.
  • raw data can be obtained directly from time-of-flight spectra or mass spectra, and then may be optionally "pre-processed” in any suitable manner.
  • signals above a predetermined signal-to-noise ratio can be selected so that a subset of peaks in a spectrum is selected, rather than selecting all peaks in a spectrum.
  • a predetermined number of peak "clusters" at a common value e.g., a particular time-of-flight value or mass-to-charge ratio value
  • a peak at a given mass-to-charge ratio is in less than 50% of the mass spectra in a group of mass spectra, then the peak at that mass-to-charge ratio can be omitted from the training data set.
  • Pre-processing steps such as these can be used to reduce the amount of data that is used to train the classification model.
  • Classification models can be formed using any suitable statistical classification (or "learning") method that attempts to segregate bodies of data into classes based on objective parameters present in the data.
  • Classification methods may be either supervised or unsupervised. Examples of supervised and unsupervised classification processes are described in Jain, "Statistical Pattern Recognition: A Review", IEEE Transactions on Pattern Analysis and Machine Intelligence, Vol. 22, No. 1, January 2000, which is herein incorporated by reference in its entirety.
  • supervised classification training data containing examples of known categories are presented to a learning mechanism, which learns one more sets of relationships that define each of the known classes. New data may then be applied to the learning mechanism, which then classifies the new data using the learned relationships.
  • supervised classification processes include linear regression processes (e.g., multiple linear regression (MLR), partial least squares (PLS) regression and principal components regression (PCR)), binary decision trees (e.g., recursive partitioning processes such as CART - classification and regression trees), artificial neural networks such as backpropagation networks, discriminant analyses (e.g., Bayesian classifier or Fischer analysis), logistic classifiers, and support vector classifiers (support vector machines).
  • linear regression processes e.g., multiple linear regression (MLR), partial least squares (PLS) regression and principal components regression (PCR)
  • binary decision trees e.g., recursive partitioning processes such as CART - classification and regression trees
  • artificial neural networks such as backpropagation networks
  • discriminant analyses e.g.
  • a preferred supervised classification method is a recursive partitioning process.
  • Recursive partitioning processes use recursive partitioning trees to classify spectra derived from unknown samples. Further details about recursive partitioning processes are provided in U.S. 2002 0138208 Al (Paulse et al., "Method for analyzing mass spectra," September 26, 2002.
  • the classification models that are created can be formed using unsupervised learning methods.
  • Unsupervised classification attempts to learn classifications based on similarities in the training data set, without pre classifying the spectra from which the training data set was derived.
  • Unsupervised learning methods include cluster analyses. A cluster analysis attempts to divide the data into "clusters" or groups that ideally should have members that are very similar to each other, and very dissimilar to members of other clusters. Similarity is then measured using some distance metric, which measures the distance between data items, and clusters together data items that are closer to each other.
  • Clustering techniques include the MacQueen ! 's K-means algorithm and the Kohonen ! 's Self-Organizing Map algorithm.
  • the peak intensity data of samples from subjects e.g., IBD subjects, and healthy controls are used as a "discovery set.” This data were combined and randomly divided into a training set and a test set to construct and test multivariate predictive models using a non-linear version of Unified Maximum Separability Analysis ("USMA") classifiers. Details of USMA classifiers are described in U.S. 2003/0055615 Al .
  • the invention provides methods for aiding a human inflammatory bowel disease diagnosis using one or more markers, for example Markers in the tables and figures which follow, and including one or more Markers 1-75 as specified herein. These markers can be used alone, in combination with other markers in any set, or with entirely different markers in aiding human inflammatory bowel disease diagnosis.
  • the markers are differentially present in samples of a human inflammatory bowel disease patient and a normal subject in whom human inflammatory bowel disease is undetectable. For example, some of the markers are expressed at an elevated level and/or are present at a higher frequency in human inflammatory bowel disease subjects than in normal subjects, while some of the markers are expressed at a decreased level and/or are present at a lower frequency in human inflammatory bowel disease subjects than in normal subjects. Therefore, detection of one or more of these markers in a person would provide useful information regarding the probability that the person may have inflammatory bowel disease.
  • the invention provides methods for aiding a human inflammatory bowel disease diagnosis using one or more markers, for example Markers in the tables and figures which follow, and including one or more Markers 1 -75 as specified herein. These markers can be used alone, in combination with other markers in any set, or with entirely different markers in aiding human inflammatory bowel disease diagnosis.
  • the markers are differentially present in samples of a human inflammatory bowel disease patient and a normal subject in whom human inflammatory bowel disease is undetectable.
  • some of the markers are expressed at an elevated level and/or are present at a higher frequency in human inflammatory bowel disease subjects than in normal subjects, while some of the markers are expressed at a decreased level and/or are present at a lower frequency in human inflammatory bowel disease subjects than in normal subjects. Therefore, detection of one or more of these markers in a person would provide useful information regarding the probability that the person may have inflammatory bowel disease.
  • a biological sample is collected from a patient and then either left unfractionated, or fractionated using an anion exchange resin as described above.
  • the biomarkers in the sample are captured using an ProteinChip array.
  • the markers are then detected using SELDI.
  • SELDI SELDI-Semiconductor
  • the results are then entered into a computer system, which contains an algorithm that is designed using the same parameters that were used in the learning algorithm and classification algorithm to originally determine the biomarkers.
  • the algorithm produces a diagnosis based upon the data received relating to each biomarker.
  • the diagnosis is determined by examining the data produced from the tests with algorithms that are developed using the biomarkers.
  • the algorithms depend on the particulars of the test protocol used to detect the biomarkers. These particulars include, for example, sample preparation, chip type and mass spectrometer parameters. If the test parameters change, the algorithm must change. Similarly, if the algorithm changes, the test protocol must change.
  • the sample is collected from the patient.
  • the biomarkers are captured using an antibody ProteinChip array as described above.
  • the markers are detected using a biospecific SELDI test system.
  • the results are then entered into a computer system, which contains an algorithm that is designed using the same parameters that were used in the learning algorithm and classification algorithm to originally determine the biomarkers.
  • the algorithm produces a diagnosis based upon the data received relating to each biomarker.
  • the markers are captured and tested using non-
  • the sample is collected from the patient.
  • the biomarkers are captured on a substrate using other known means, e.g., antibodies to the markers.
  • the markers are detected using methods known in the art, e.g., optical methods and refractive index. Examples of optical methods include detection of fluorescence, e.g., ELISA. Examples of refractive index include surface plasmon resonance.
  • the results for the markers are then subjected to an algorithm, which may or may not require artificial intelligence.
  • the algorithm produces a diagnosis based upon the data received relating to each biomarker.
  • the data from the sample may be fed directly from the detection means into a computer containing the diagnostic algorithm.
  • embodiments of the invention include methods for aiding a human inflammatory bowel disease diagnosis, wherein the method comprises: (a) detecting at least one marker in a sample, wherein the marker is selected from any of the Markers 1-75; and (b) correlating the detection of the marker or markers with a probable diagnosis of human inflammatory bowel disease.
  • the correlation may take into account the amount of the marker or markers in the sample compared to a control amount of the marker or markers (up or down regulation of the marker or markers) (e.g., in normal subjects in whom human inflammatory bowel disease is undetectable).
  • the correlation may take into account the presence or absence of the markers in a test sample and the frequency of detection of the same markers in a control.
  • the correlation may take into account both of such factors to facilitate determination of whether a subject has a human inflammatory bowel disease or not.
  • Markers 1 -75 are used to make a correlation with inflammatory bowel disease, wherein the inflammatory bowel disease may be any subtype, e.g., Crohn's disease or ulcerative colitis.
  • a sample is a colon or intestinal biopsy, e.g., an endoscopic biopsy sample from the subject.
  • the sample can be prepared as described above to enhance detectability of the markers.
  • a sample from the subject can be preferably fractionated by, e.g., Cibacron blue agarose chromatography and single stranded DNA affinity chromatography, anion exchange chromatography and the like. Sample preparations, such as pre-fractionation protocols, are optional and may not be necessary to enhance detectability of markers depending on the methods of detection used. For example, sample preparation may be unnecessary if antibodies that specifically bind markers are used to detect the presence of markers in a sample.
  • Processes for the purification of a biomarker include fractioning a sample, as described herein, for example, by size-exclusion chromatography and collecting a fraction that includes one or more biomarkers; and/or fractionating a sample comprising the one or more biomarkers by anion exchange chromatography and collecting a fraction that includes one or more biomarkers, wherein the biomarker is selected from one or more of the biomarkes of Tables 1 - 3.
  • the invention also includes IBD candidate genes. These genes include, for example, apoptosis -regulating CASPlO at 2q33-34, LILRBl at 19ql3.4 (locus IBD6) and antigen-presenting genes PSME2 at 14ql 1.2 (locus IBD4). With respect to the IBD3 locus at 6p21, 35 HLA-DMA, TAPl, UBD and PSMB8 (immunoproteasome for generating MHC class I binding antigenic peptides), at 6p21.3, are particularly interesting.
  • GNGTl (7q21.3) functioning in apoptosis and PRKACB (Ip36.1 , IBD7), involved in Wnt-signaling from the UC signature are also good candidates.
  • the sequences of these genes are appended to the end of this specification, as well as exemplary primers for detecting or amplifying the makers.
  • any biomarker is useful in aiding in the determination of inflammatory bowel disease status.
  • the selected biomarker is measured in a subject sample using the methods described herein, e.g., capture on a SELDI biochip followed by detection by mass spectrometry. Then, the measurement is compared with a diagnostic amount or control that distinguishes a inflammatory bowel disease status from a non-inflammatory bowel disease status.
  • the diagnostic amount will reflect the information herein that a particular biomarker is up-regulated or down-regulated in a inflammatory bowel disease status compared with a non-inflammatory bowel disease status.
  • the particular diagnostic amount used can be adjusted to increase sensitivity or specificity of the diagnostic assay depending on the preference of the diagnostician. The test amount as compared with the diagnostic amount thus indicates inflammatory bowel disease status.
  • biomarkers 1-75 can be more frequently detected in human inflammatory bowel disease subjects than in normal subjects.
  • biomarkers 61-75 can be less frequently detected in human UC disease subjects than in normal subjects, and/or in subjects who have CD. The mere detection of one or more of these markers in a subject being tested indicates that the subject has a lower probability of having inflammatory bowel disease.
  • the measurement of markers can involve quantifying the markers to correlate the detection of markers with a probable diagnosis of inflammatory bowel disease.
  • a control amount i.e., higher or lower than the control, depending on the marker
  • the correlation may take into account the amount of the marker or markers in the sample compared to a control amount of the marker or markers (up or down regulation of the marker or markers) (e.g., in normal subjects or in non-inflammatory bowel disease subjects such as where inflammatory bowel disease is undetectable).
  • a control can be, e.g., the average or median amount of marker present in comparable samples of normal subjects in normal subjects or in non-inflammatory bowel disease subjects such as where inflammatory bowel disease is undetectable.
  • the control amount is measured under the same or substantially similar experimental conditions as in measuring the test amount.
  • the correlation may take into account the presence or absence of the markers in a test sample and the frequency of detection of the same markers in a control. The correlation may take into account both of such factors to facilitate determination of inflammatory bowel disease status.
  • the methods further comprise managing subject treatment based on the status.
  • management of the subject describes the actions of the physician or clinician subsequent to determining inflammatory bowel disease status.
  • the physician may order more tests (e.g., colonoscopy and imaging techniques).
  • the physician may schedule the patient for treatment.
  • the patient may receive therapeiutic treatments, either in lieu of, or in addition to, surgery. No further action may be warranted.
  • a maintenance therapy or no further management may be necessary.
  • Therapeutic agents may include, one or more of sulfa drugs, corticosteriods (prednisone), 5-aminosalicylates (Asacol, Pentasa, Rowasa, or 5-ASA), immunosuppressives (azathioprine, Imuran, Cyclosporine, 6-MP, Purinethol and Methotrexate), anti-TNF (Remicade), anticholinergics, dicyclomine (Bentyl), belladonna/phenobarbital (Donnatal,
  • the invention also provides for such methods where the biomarkers (or specific combination of biomarkers) are measured again after subject management.
  • the methods are used to monitor the status of the inflammatory bowel disease, e.g., response to inflammatory bowel disease treatment, remission of the disease or progression of the disease. Because of the ease of use of the methods and the lack of invasiveness of the methods, the methods can be repeated after each treatment the patient receives. This allows the physician to follow the effectiveness of the course of treatment. If the results show that the treatment is not effective, the course of treatment can be altered accordingly. This enables the physician to be flexible in the treatment options.
  • the methods for detecting markers can be used to assay for and to identify compounds that modulate expression of these markers in vivo or in vitro.
  • the markers can be used to screen for compounds that modulate the expression of the markers in vitro or in vivo, which compounds in turn may be useful in treating or preventing inflammatory bowel disease in subjects.
  • the markers can be used to monitor the response to treatments for inflammatory bowel disease.
  • the markers can be used in heredity studies to determine if the subject is at risk for developing inflammatory bowel disease. For instance, certain markers may be genetically linked. This can be determined by, e.g., analyzing samples from a population of inflammatory bowel disease subjects whose families have a history of inflammatory bowel disease.
  • the results can then be compared with data obtained from, e.g., -inflammatory bowel disease subjects whose families do not have a history of inflammatory bowel disease.
  • the markers that are genetically linked may be used as a tool to determine if a subject whose family has a history of inflammatory bowel disease is pre-disposed to having inflammatory bowel disease.
  • a diagnosis based on the presence or absence in a test subject of any the biomarkers of this invention is communicated to the subject as soon as possible after the diagnosis is obtained.
  • the diagnosis may be communicated to the subject by the subject's treating physician. Alternatively, the diagnosis may be sent to a test subject by email or communicated to the subject by phone.
  • a computer may be used to communicate the diagnosis by email or phone.
  • the message containing results of a diagnostic test may be generated and delivered automatically to the subject using a combination of computer hardware and software which will be familiar to artisans skilled in telecommunications.
  • a healthcare-oriented communications system is described in U.S. Patent Number 6,283,761; however, the present invention is not limited to methods which utilize this particular communications system.
  • all or some of the method steps, including the assaying of samples, diagnosing of diseases, and communicating of assay results or diagnoses may be carried out in diverse (e.g., foreign) jurisdictions.
  • Methods of the invention for determining the inflammatory bowel disease status of a subject include for example, obtaining a biomarker profile from a sample taken from the subject; and comparing the subject's biomarker profile to a reference biomarker profile obtained from a reference population, wherein the comparison is capable of classifying the subject as belonging to or not belonging to the reference population; wherein the subject's biomarker profile and the reference biomarker profile comprise one or more markers listed in Tables 1 - 3.
  • the method may further comprise repeating the method at least once, wherein the subject's biomarker profile is obtained from a separate sample taken each time the method is repeated.
  • Samples from the subject may be taken at any time, for example, the samples may be taken 24 hours apart or any other time determined useful.
  • Such comparisons of the biomarker profiles can determine inflammatory bowel disease status in the subject with an accuracy of at least about 60%, 70%, 80%, 90%, 95%, and approaching 100% as shown in the examples which follow.
  • the reference biomarker profile can be obtained from a population comprising a single subject, at least two subjects, at least 20 subjects or more. The number of subjects will depend, in part, on the number of available subjects, and the power of the statistical analysis necessary.
  • a method of treating inflammatory bowel disease comprising administering to a subject suffering from or at risk of developing inflammatory bowel disease a therapeutically effective amount of a compound capable of modulating the expression or activity of one or more of the biomarkers of Tables 1 - 3.
  • a method of treating a condition in a subject comprising administering to a subject a therapeutically effective amount of a compound which modulates the expression or activity of one or more of the biomarkers of Tables 1 - 3.
  • Compounds useful in methods disclosed herein include, for example, sulfa drugs, corticosteriods (prednisone), 5-aminosalicylates (Asacol, Pentasa, Rowasa, or 5-ASA), immunosuppressives (azathioprine, Imuran, Cyclosporine, 6-MP, Purinethol and Methotrexate), anti-TNF (Remicade), anticholinergics, dicyclomine (Bentyl), belladonna/phenobarbital (Donnatal, Antispas, bBarbidonna, donnapine, hyosophen, Spasmolin), hyoscyamine (Levsin, Anaspaz), chlordiazepoxide/clidinium (Librax), anti- diarrheals, diphenoxylate/atropine (Lomotil), alosetron hydrochloride (Lotronex), tegaserod (Zelnorm, Zelmac),
  • a method of qualifying inflammatory bowel disease status in a subject comprising:
  • the method may also comprise the step of measuring the at least one biomarker after subject management.
  • the methods of the invention may further comprise generating data on immobilized subject samples on a biochip, by subjecting the biochip to laser ionization and detecting intensity of signal for mass/charge ratio; and transforming the data into computer readable form; and executing an algorithm that classifies the data according to user input parameters, for detecting signals that represent biomarkers present in inflammatory bowel disease subjects and are lacking in non-inflammatory bowel disease subject controls.
  • Types of inflammatory bowel disease that may be identified or differentiated from one another according to this method include UC and CD.
  • kits for the analysis of IBD status include PCR primers for at least one marker selected from Markers 1 - 75.
  • the kit includes more than two or three markers selected from Markers 1 - 75.
  • the kit may further include instructions for use and correlation of the maker with disease status. For example, the presence of any one of Markers 1 - 31 indicate CD; the presence of any one of Markers 32 - 48 indicate IBD; the increased presence of any one of Markers 49- 60 indicate UC and the decreased presence of any one of Markers 61-75 indicate UC.
  • the kit may also include a DNA array containing the complement of one or more of the Markers selected from 1 - 75, reagents, and/or enzymes for amplifying or isolating sample DNA.
  • the kits may include reagents for real-time PCR, for example, TaqMan probes and/or primers, and enzymes.
  • kits for qualifying inflammatory bowel disease status and/or aiding a diagnosis of human inflammatory bowel disease wherein the kits can be used to detect the markers of the present invention.
  • the kits can be used to detect any one or more of the markers described herein, which markers are differentially present in samples of inflammatory bowel disease subjects and normal subjects.
  • the kits of the invention have many applications.
  • the kits can be used to differentiate if a subject has inflammatory bowel disease or has a negative diagnosis, thus aiding a human inflammatory bowel disease diagnosis.
  • the kits can be used to identify compounds that modulate expression of one or more of the markers in in vitro or in vivo animal models for inflammatory bowel disease.
  • a kit comprises: (a) a substrate comprising an adsorbent thereon, wherein the adsorbent is suitable for binding a marker, and (b) instructions to detect the marker or markers by contacting a sample with the adsorbent and detecting the marker or markers retained by the adsorbent.
  • the kit may comprise an eluant (as an alternative or in combination with instructions) or instructions for making an eluant, wherein the combination of the adsorbent and the eluant allows detection of the markers using gas phase ion spectrometry.
  • Such kits can be prepared from the materials described above, and the previous discussion of these materials ⁇ e.g., probe substrates, adsorbents, washing solutions, etc.) is fully applicable to this section and will not be repeated.
  • the kit may comprise a first substrate comprising an adsorbent thereon ⁇ e.g., a particle functionalized with an adsorbent) and a second substrate onto which the first substrate can be positioned to form a probe ⁇ which is removably insertable into a gas phase ion spectrometer.
  • the kit may comprise a single substrate ? which is in the form of a removably insertable probe with adsorbents on the substrate.
  • the kit may further comprise a pre-fractionation spin column ⁇ e.g., Cibacron blue agarose column, anti-HSA agarose column, K-30 size exclusion column, Q-anion exchange spin column, single stranded DNA column, lectin column, etc.).
  • a pre-fractionation spin column ⁇ e.g., Cibacron blue agarose column, anti-HSA agarose column, K-30 size exclusion column, Q-anion exchange spin column, single stranded DNA column, lectin column, etc.
  • kits comprises (a) an antibody that specifically binds to a marker; and (b) a detection reagent.
  • a kit can be prepared from the materials described above, and the previous discussion regarding the materials ⁇ e.g., antibodies, detection reagents, immobilized supports, etc.) is fully applicable to this section and will not be repeated.
  • the kit may further comprise pre-fractionation spin columns.
  • the kit may further comprise instructions for suitable operation parameters in the form of a label or a separate insert.
  • the kit may further comprise a standard or control information so that the test sample can be compared with the control information standard to determine if the test amount of a marker detected in a sample is a diagnostic amount consistent with a diagnosis of inflammatory bowel disease.
  • the biomarkers can be used to screen for compounds that modulate the expression of the biomarkers in vitro or in vivo, which compounds in turn may be useful in treating or preventing inflammatory bowel disease in subjects.
  • the biomarkers can be used to monitor the response to treatments for inflammatory bowel disease.
  • the biomarkers can be used in heredity studies to determine if the subject is at risk for developing inflammatory bowel disease.
  • kits of this invention could include a solid substrate having a hydrophobic function, such as a protein biochip (e.g., a Ciphergen ProteinChip array) and a buffer for washing the substrate, as well as instructions providing a protocol to measure the biomarkers of this invention on the chip and to use these measurements to diagnose inflammatory bowel disease.
  • Method for identifying a candidate compound for treating inflammatory bowel disease may comprise, for example, contacting one or more of the biomarkers of Tables 1 - 3 with a test compound; and determining whether the test compound interacts with the biomarker, wherein a compound that interacts with the biomarker is identified as a candidate compound for treating inflammatory bowel disease.
  • Compounds suitable for therapeutic testing may be screened initially by identifying compounds which interact with one or more biomarkers listed in identified herein.
  • screening might include recombinantly expressing a biomarker of this invention, purifying the biomarker, and affixing the biomarker to a substrate.
  • Test compounds would then be contacted with the substrate, typically in aqueous conditions, and interactions between the test compound and the biomarker are measured, for example, by measuring elution rates as a function of salt concentration.
  • Certain proteins may recognize and cleave one or more biomarkers of this invention, in which case the proteins may be detected by monitoring the digestion of one or more biomarkers in a standard assay, e.g., by gel electrophoresis of the proteins.
  • the ability of a test compound to inhibit the activity of one or more of the biomarkers of this invention may be measured.
  • One of skill in the art will recognize that the techniques used to measure the activity of a particular biomarker will vary depending on the function and properties of the biomarker. For example, an enzymatic activity of a biomarker may be assayed provided that an appropriate substrate is available and provided that the concentration of the substrate or the appearance of the reaction product is readily measurable.
  • the ability of potentially therapeutic test compounds to inhibit or enhance the activity of a given biomarker may be determined by measuring the rates of catalysis in the presence or absence of the test compounds.
  • test compounds to interfere with a non-enzymatic (e.g., structural) function or activity of one of the biomarkers of this invention may also be measured.
  • a non-enzymatic function or activity of one of the biomarkers of this invention may also be measured.
  • the self-assembly of a multi-protein complex which includes one of the biomarkers of this invention may be monitored by spectroscopy in the presence or absence of a test compound.
  • test compounds which interfere with the ability of the biomarker to enhance transcription may be identified by measuring the levels of biomarker-dependent transcription in vivo or in vitro in the presence and absence of the test compound.
  • Test compounds capable of modulating the activity of any of the biomarkers of this invention may be administered to subjects who are suffering from or are at risk of developing inflammatory bowel disease.
  • the administration of a test compound which increases the activity of a particular biomarker may decrease the risk of inflammatory bowel disease in a patient if the activity of the particular biomarker in vivo prevents the accumulation of proteins for inflammatory bowel disease.
  • the administration of a test compound which decreases the activity of a particular biomarker may decrease the risk of inflammatory bowel disease in a patient if the increased activity of the biomarker is responsible, at least in part, for the onset of inflammatory bowel disease.
  • screening a test compound includes obtaining samples from test subjects before and after the subjects have been exposed to a test compound.
  • the levels in the samples of one or more of the biomarkers of this invention may be measured and analyzed to determine whether the levels of the biomarkers change after exposure to a test compound.
  • the samples may be analyzed by mass spectrometry, as described herein, or the samples may be analyzed by any appropriate means known to one of skill in the art.
  • the levels of one or more of the biomarkers of this invention may be measured directly by Western blot using radio- or fluorescently-labeled antibodies which specifically bind to the biomarkers.
  • changes in the levels of mRNA encoding the one or more biomarkers may be measured and correlated with the administration of a given test compound to a subject.
  • the changes in the level of expression of one or more of the biomarkers may be measured using in vitro methods and materials.
  • human tissue cultured cells which-express, or are capable of expressing, one or more of the biomarkers of this invention may be contacted with test compounds.
  • Subjects who have been treated with test compounds will be routinely examined for any physiological effects which may result from the treatment.
  • the test compounds will be evaluated for their ability to decrease disease likelihood in a subject.
  • test compounds will be screened for their ability to slow or stop the progression of the disease.
  • a dataset can be analyzed by multiple classification algorithms. Some classification algorithms provide discrete rules for classification; others provide probability estimates of a certain outcome (class). In the latter case, the decision (diagnosis) is made based on the class with the highest probability. For example, consider the three-class problem: healthy, benign, and IBD. Suppose that a classification algorithm (e.g. Nearest neighbor) is constructed and applied to sample A, and the probability of the sample being healthy is 0, benign is 33%, and IBD is 67%. Sample A would be diagnosed as being IBD. This approach, however, does not take into account any "fuzziness" in the diagnosis i.e. that there was a certain probability that the sample was benign. Therefore, the diagnosis would be the same as for sample B, which has a probability of 0 of being healthy or benign and a probability of 1 of being IBD.
  • a classification algorithm e.g. Nearest neighbor
  • CD49un 21 F ileocolonic 3 rectum unaffected - +
  • CD53un 55 M colonic 15 T colon unaffected -
  • Fibrosis score based on extent of lamina intestinal involvement, splaying of the muscula ⁇ s mucosa, and crypt dropout
  • “Affected” pinch biopsies are from areas appearing affected by endoscopy, "unaffected” biopsies are from an area at least 10 cm away from any grossly diseased area (Table 4).
  • Histology of an adjacent biopsy was scored for inflammation and fibrosis (A.M.) (Table 4).
  • a four-tier grading scheme (-, +, ++, +++), based on semi-quantitative assessment of mucosal inflammation and fibrosis was used.
  • Biotinylated cRNA (10 ⁇ g per array) was hybridized to high-density oligonucleotide GeneChip Human Genome U95Av2 arrays (Affymetrix). The arrays were washed and stained (R-Phycoerythrin Streptavidin) in a GeneChip Fluidics Station 400.
  • the DNA-Chip Analyzer (dChip) software 17 was used to normalize the data from the image files for array-to-array comparisons (http://www.ncbi.nlm.nih. gov/geo) .
  • SAM Significance Analysis of Microarays
  • MDS classical Multidimensional Scaling
  • CD-33 and CD -53 Two other patients (CD-33 and CD -53) with rectal disease and high histopathologic inflammation scores co-localized with the UC affected, possibly representing a CD subgroup resembling UC.
  • CD-45 affected endoscopically, placed in the MDS plot with controls and unaffected was subsequently found to be negative for inflammation and fibrosis by histology.
  • genes were sought that were responsible for positioning of the samples in the different quadrants of the MDS map.
  • An analysis of variance on each gene identified those with significant, quadrant specific differences in expression. From the expression pattern of these genes
  • Group 3 three groups are evident.
  • Group 1 Twenty-seven genes expressed above mean in the controls and in 5 CD individuals are down regulated in four of the five individuals with UC. A majority of these genes code for membrane-bound endoplasmic reticulum-, Golgi apparatus-, or in a few cases lysozomal- proteins. These are primarily epithelial genes that regulate protein trafficking and secretion. The only two CD individuals that manifest this UC pattern are CD 33 and 53, both noted for active rectal inflammation resembling UC.
  • Group 2 Nine genes are elevated in most CD and UC affected profiles and most likely contribute towards separation of IBD from normal controls in the MDS plot.
  • inflammatory response ILlB, SlOOAS
  • antigen presentation MHC class II immunoproteasome members PSME2 and PSMB8, MHC class II ATP-binding antigen peptide transporter TAPl, HLA-DMA and UBD of MHC class I
  • IL8 inflammatory cell chemotaxis
  • CASPl CASPlO
  • macrophage activation ASMT and interferon-regulated genes IFITMl, IFITM3, ISG20, IFI35, SPIlO
  • LILRB leukocyte protection
  • ADM acute phase response
  • STATl STAT3
  • protease inhibitors SERPINAl and SPINKl to prevent tissue destruction
  • Transporter 1 ATP-binding cassette, sub B TAPl Antigen presentation 6p21 3
  • Mucin 1 transmembrane MUCI Cytoskeleton I q21 Myosin, light polypeptide 3 MYU Cytoskeleton 3p21 3-p21 2 Chymotrypsin-hke CTRL Immune response 16q22 1 Interferon induced transmembrane protein 1 IFITMI Immune response 1 I p l 5 5 Interferon induced transmembrane protein 3 IFlTMi Immune response I l pl 5 5 5
  • UC Gene Expression Signature derived by comparing all five UC affected to control, up- regulations suggest complement cascade activation (BF and C4A), growth regulatory (MIA) and apoptosis (A TM) changes, detoxification (NNMT) and intracellular transport (SNX26) (Table 6).
  • Down regulations in UC are seen in biosynthetic and metabolic processes (PANK3, HPGD), and in endoplasmic reticulum-, Golgi-transport/intracellular trafficking (F2RL1, GABRGl, GNGTl, SLC4A4). Table 6.
  • RNA binding motif protein 8A RBMSA Unknown I q l 2
  • Protein phosphatase 2 (formerly 2A), regulatory subunit B", alpha PPP2R3A Cell growth regulation 3q22 1
  • Solute carrier family 4 sodium bicarbonate cotransporter, member 4 SLC4A4 Ion transport 4q21
  • GABA Gamma-aminobuty ⁇ c acid
  • CD genes over-expressed in CD are overwhelmingly those of acute phase and innate immune response (involving IL-I and TNF ⁇ mediated induction of NF- ⁇ B), MHC class II mediated antigen presentation, macrophage activation and recruitment of inflammatory cells.
  • the distinctive transmural tissue damage and mesenchymal involvement in CD may be due to this major early involvement of immune and inflammatory cells.
  • Gene expression changes in UC make a strong case for loss of epithelial homeostasis as being central to UC.
  • Epithelial secretion is a process that is pivotal to maintaining intestinal mucosal integrity.
  • Intracellular trafficking and secretory functions of the endoplasmic reticulum are essential for the degradation and secretion of ingested environmental toxins by the intestinal epithelium.
  • ER endoplasmic reticulum
  • An overload of degraded, unfolded proteins has been proposed to cause ER stress as in the Irel ⁇ (Inositol requiring kinase l)-deficient mouse that develop colitis when challenged with dextran sodium sulfate.
  • IBD candidate genes apoptosis -regulating CASPlO at 2q33-34, LILRBl at 19ql3.4 (locus IBD6) and antigen- presenting genes PSME2 at 14ql l .2 (locus IBD4).
  • IBD3 locus 35 HLA-DMA, TAPl, UBD and PSMB8 (immunoproteasome for generating MHC class I binding antigenic peptides), at 6p21.3, are particularly interesting.
  • GNGTl (7q21.3) functioning in apoptosis and PRKACB (Ip36.1 , IBD7), involved in Wnt-signaling from the UC signature are also good candidates.
  • biopsies marked as "affected” manifest variable degrees of acute or chronic colitis, including one or more of the following histologic features: cryptitis, with or without accompanying crypt abscesses, crypt distortion, lamina intestinal fibrosis, crypt dropout, basal lymphoplasmacytosis, and Paneth cell metaplasia ( Figure 5, C and D). None of the biopsies indicate evidence of colitis-associated epithelial dysplasia or neoplasia.
  • CXCLl, DMBTl, ASMT, ADM, STAT3, IFI35, PSME2 and PSMB8 were selected from our microarray expression profiles for further confirmation by quantitative (q) RT-PCR ( Figure 6).
  • the qRT-PCR results show excellent agreement with the array analysis results.
  • CXCLl and DMBTl are up regulated in CD and UC affected biopsies, while ADM, STAT3, PSME2 and PSMB8 are primarily up regulated in CD affected biopsies.
  • ASMT and IFB 5 show elevated levels of transcript in CD affected and unaffected biopsies ( Figure 6).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Analytical Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Pathology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Physiology (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Other Investigation Or Analysis Of Materials By Electrical Means (AREA)

Abstract

La présente invention concerne des procédés pour déterminer le stade d'une maladie intestinale inflammatoire chez un sujet. L'invention concerne en outre des nécessaires pour déterminer le stade d'une maladie intestinale inflammatoire chez un sujet. L'invention concerne par ailleurs des procédés d'identification de biomarqueurs pour déterminer le stade d'une maladie intestinale inflammatoire chez un sujet.
PCT/US2005/044423 2004-12-06 2005-12-06 Biomarqueurs pour maladie intestinale inflammatoire WO2006063133A2 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP05853365A EP1844158A4 (fr) 2004-12-06 2005-12-06 Biomarqueurs pour maladie intestinale inflammatoire
US11/750,114 US20090258848A1 (en) 2005-12-06 2007-05-17 Biomarkers for inflammatory bowel disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63366204P 2004-12-06 2004-12-06
US60/633,662 2004-12-06

Publications (2)

Publication Number Publication Date
WO2006063133A2 true WO2006063133A2 (fr) 2006-06-15
WO2006063133A3 WO2006063133A3 (fr) 2009-04-02

Family

ID=36578568

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/044423 WO2006063133A2 (fr) 2004-12-06 2005-12-06 Biomarqueurs pour maladie intestinale inflammatoire

Country Status (2)

Country Link
EP (1) EP1844158A4 (fr)
WO (1) WO2006063133A2 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008048986A2 (fr) * 2006-10-17 2008-04-24 Children's Hospital Medical Center Technique par batterie de gènes pour prédire la réponse dans les maladies intestinales inflammatoires
WO2008079406A2 (fr) * 2006-12-19 2008-07-03 Genentech, Inc. Marqueurs d'expression génique pour une maladie intestinale inflammatoire
WO2008147900A2 (fr) * 2007-05-22 2008-12-04 Genentech, Inc. Marqueurs d'expression génique pour une affection abdominale inflammatoire
WO2009073565A3 (fr) * 2007-11-29 2009-09-24 Genentech, Inc. Marqueur d'expression génique de maladie intestinale inflammatoire
EP2160475A2 (fr) * 2007-05-22 2010-03-10 Centocor Ortho Biotech Inc. Marqueurs et procédé permettant d'évaluer et traiter la maladie de crohn et les troubles associés
WO2011011339A1 (fr) * 2009-07-20 2011-01-27 Genentech, Inc. Marqueurs de l'expression génique pour la maladie de crohn
WO2011127351A1 (fr) * 2010-04-09 2011-10-13 Exagen Diagnostics, Inc. Marqueurs biologiques pour la colite ulcéreuse et la maladie de crohn
AU2007223881B2 (en) * 2006-03-09 2014-01-09 Alan Safdi Rifaximin anti-rectal dysfunction preparation
US8663927B2 (en) 2007-09-10 2014-03-04 University Of Kentucky Research Foundation Systems and methods for diagnosis and monitoring of bacteria-related conditions
EP2944651A1 (fr) * 2007-10-02 2015-11-18 Universität zu Köln Nouveaux gènes marqueurs pour lymphocytes t de régulation du sang humain
EP2148943B1 (fr) * 2007-04-30 2016-10-05 Janssen Biotech, Inc. Procedes pour evaluer et traiter la colite ulcerative et les troubles associes a l'aide d'un panel de 19 genes
CN112941150A (zh) * 2021-03-25 2021-06-11 安徽医科大学 一种筛选溃疡性结肠炎疾病活动度指标的方法
US11299768B2 (en) 2009-01-19 2022-04-12 Biomerieux Methods for determining a patient's susceptibility of contracting a nosocomial infection and for establishing a prognosis of the progression of septic syndrome

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2774769B2 (ja) * 1993-04-26 1998-07-09 賢治 寒川 アドレノメデュリン
US6632617B1 (en) * 1997-05-13 2003-10-14 Incyte Corporation Tumor-associated antigen
US6150096A (en) * 1997-09-26 2000-11-21 Universite De Sherbrooke Molecular markers for the diagnosis of human diseases including Crohn's disease
US6228585B1 (en) * 1998-09-04 2001-05-08 Washington University Gene markers for chronic mucosal injury
WO2002059367A2 (fr) * 2000-11-30 2002-08-01 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Jeu de microechantillons de diagnostic pour ibd, maladie de crohn et colite ulcereuse
FR2822955B1 (fr) * 2001-03-27 2003-10-03 Chru Lille Methode de diagnostic de maladies inflammatoires chroniques de l'intestin
EP1534855B1 (fr) * 2002-06-25 2010-12-08 Index Diagnostics AB (publ) Procede et kit pour diagnostic de colite ulcereuse
US20060264393A1 (en) * 2002-12-30 2006-11-23 Yoram Groner Methods for evaluating genetic susceptibility and therapy for chronic inflammatory diseases
WO2004065577A2 (fr) * 2003-01-14 2004-08-05 Bristol-Myers Squibb Company Polynucleotides et polypeptides associes a la voie nf-$g(k)b

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1844158A4 *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2007223881B2 (en) * 2006-03-09 2014-01-09 Alan Safdi Rifaximin anti-rectal dysfunction preparation
US8987292B2 (en) 2006-03-09 2015-03-24 Salix Pharmaceuticals, Ltd. Rifaximin anti-rectal dysfunction preparation
US20100267575A1 (en) * 2006-10-17 2010-10-21 Childrens Hospital Medical Center Gene array technique for predicting response in inflammatory bowel diseases
WO2008048986A3 (fr) * 2006-10-17 2008-09-12 Childrens Hosp Medical Center Technique par batterie de gènes pour prédire la réponse dans les maladies intestinales inflammatoires
WO2008048986A2 (fr) * 2006-10-17 2008-04-24 Children's Hospital Medical Center Technique par batterie de gènes pour prédire la réponse dans les maladies intestinales inflammatoires
WO2008079406A2 (fr) * 2006-12-19 2008-07-03 Genentech, Inc. Marqueurs d'expression génique pour une maladie intestinale inflammatoire
WO2008079406A3 (fr) * 2006-12-19 2008-12-31 Genentech Inc Marqueurs d'expression génique pour une maladie intestinale inflammatoire
EP2148943B1 (fr) * 2007-04-30 2016-10-05 Janssen Biotech, Inc. Procedes pour evaluer et traiter la colite ulcerative et les troubles associes a l'aide d'un panel de 19 genes
WO2008147900A2 (fr) * 2007-05-22 2008-12-04 Genentech, Inc. Marqueurs d'expression génique pour une affection abdominale inflammatoire
EP2160475A2 (fr) * 2007-05-22 2010-03-10 Centocor Ortho Biotech Inc. Marqueurs et procédé permettant d'évaluer et traiter la maladie de crohn et les troubles associés
WO2008147900A3 (fr) * 2007-05-22 2009-12-03 Genentech, Inc. Marqueurs d'expression génique pour une affection abdominale inflammatoire
EP2160475A4 (fr) * 2007-05-22 2011-02-09 Centocor Ortho Biotech Inc Marqueurs et procédé permettant d'évaluer et traiter la maladie de crohn et les troubles associés
US8557745B2 (en) 2007-05-22 2013-10-15 Janssen Biotech, Inc. Markers and methods for assessing and treating Crohn's and related disorders
US8663927B2 (en) 2007-09-10 2014-03-04 University Of Kentucky Research Foundation Systems and methods for diagnosis and monitoring of bacteria-related conditions
EP2944651A1 (fr) * 2007-10-02 2015-11-18 Universität zu Köln Nouveaux gènes marqueurs pour lymphocytes t de régulation du sang humain
EP2261367A3 (fr) * 2007-11-29 2011-03-23 Genentech, Inc. Marqueurs d'expression génique pour maladies intestinales inflammatoires
CN101970689A (zh) * 2007-11-29 2011-02-09 健泰科生物技术公司 炎性肠病的基因表达标志物
WO2009073565A3 (fr) * 2007-11-29 2009-09-24 Genentech, Inc. Marqueur d'expression génique de maladie intestinale inflammatoire
US11299768B2 (en) 2009-01-19 2022-04-12 Biomerieux Methods for determining a patient's susceptibility of contracting a nosocomial infection and for establishing a prognosis of the progression of septic syndrome
CN102639710A (zh) * 2009-07-20 2012-08-15 基因泰克公司 克罗恩病的基因表达标记
EP2584049A3 (fr) * 2009-07-20 2013-08-28 Genentech, Inc. Marqueurs d'expression génétique pour la maladie de Crohn
EP2757160A3 (fr) * 2009-07-20 2014-07-30 Genentech, Inc. Marqueurs d'expression génétique pour la maladie de Crohn
WO2011011339A1 (fr) * 2009-07-20 2011-01-27 Genentech, Inc. Marqueurs de l'expression génique pour la maladie de crohn
WO2011127351A1 (fr) * 2010-04-09 2011-10-13 Exagen Diagnostics, Inc. Marqueurs biologiques pour la colite ulcéreuse et la maladie de crohn
CN112941150A (zh) * 2021-03-25 2021-06-11 安徽医科大学 一种筛选溃疡性结肠炎疾病活动度指标的方法

Also Published As

Publication number Publication date
WO2006063133A3 (fr) 2009-04-02
EP1844158A2 (fr) 2007-10-17
EP1844158A4 (fr) 2010-09-08

Similar Documents

Publication Publication Date Title
US20090258848A1 (en) Biomarkers for inflammatory bowel disease
US8299216B2 (en) Biomarkers for melanoma
WO2006063133A2 (fr) Biomarqueurs pour maladie intestinale inflammatoire
US20180106817A1 (en) Protein biomarkers and therapeutic targets for renal disorders
US7812143B2 (en) Biomarkers for cancer treatment
US20130316921A1 (en) Methods for diagnosis of kawasaki disease
EP1590469A2 (fr) Diagnostic du sepsis ou du sirs au moyen de profils de marqueurs biologiques
WO2004044555A2 (fr) Diagnostic de la septicemie ou sirs au moyen de profils de biomarqueurs
AU2012217717B2 (en) Compositions and methods for diagnosing ovarian cancer
WO2011119980A1 (fr) Biomarqueurs protéiques et génétiques pour le rejet de greffes d'organes
US9075062B2 (en) Identification of biomarkers by serum protein profiling
AU2014207321A1 (en) Anti-TNF and anti-IL17 combination therapy biomarkers for inflammatory disease
CN111876478A (zh) 肺结节诊断标志物及应用
CN113249491A (zh) 用于诊断子宫内膜癌的生物标志物及其产品和应用
WO2011044142A1 (fr) Biomarqueurs sanguins périphériques de la pneumonie interstitielle idiopathique et leurs méthodes d'utilisation
US20060246466A1 (en) Identification of biomarkers for detecting gastric carcinoma
WO2010085124A2 (fr) Marqueur pour le diagnostic du cancer du foie, la prédiction de sa récurrence et la prédiction de survie associée, trousse comprenant ledit marqueur et prédiction du pronostic chez des patients atteints d'un cancer du foie faisant appel audit marqueur
US20230118097A1 (en) Biomarkers for placenta accreta spectrum (pas) disorders
EP2607494A1 (fr) Biomarqueurs pour l'évaluation du risque de cancer des poumons
WO2017147492A1 (fr) Méthodes de détection et de traitement de maladies inflammatoires des voies aériennes

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

NENP Non-entry into the national phase in:

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005853365

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2005853365

Country of ref document: EP