US20130345322A1 - Diagnostic for colorectal cancer - Google Patents

Diagnostic for colorectal cancer Download PDF

Info

Publication number
US20130345322A1
US20130345322A1 US13/809,785 US201113809785A US2013345322A1 US 20130345322 A1 US20130345322 A1 US 20130345322A1 US 201113809785 A US201113809785 A US 201113809785A US 2013345322 A1 US2013345322 A1 US 2013345322A1
Authority
US
United States
Prior art keywords
m2pk
mac2bp
igfbp2
epcam
biomarkers
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/809,785
Inventor
Leah Jane Cosgrave
Bruce Tabor
Antony Wilks Burgess
Edouard Collins Nice
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vision Tech Bio Pty Ltd
Original Assignee
Commonwealth Scientific and Industrial Research Organization CSIRO
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
Priority claimed from AU2010903140A external-priority patent/AU2010903140A0/en
Application filed by Commonwealth Scientific and Industrial Research Organization CSIRO filed Critical Commonwealth Scientific and Industrial Research Organization CSIRO
Assigned to COMMONWEALTH SCIENTIFIC AND INDUSTRIAL RESEARCH ORGANISATION reassignment COMMONWEALTH SCIENTIFIC AND INDUSTRIAL RESEARCH ORGANISATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TABOR, BRUCE, COSGROVE, LEAH JANE, BURGESS, ANTONY WILKS, NICE, EDOUARD COLLINS
Publication of US20130345322A1 publication Critical patent/US20130345322A1/en
Assigned to VISION TECH BIO PTY. LTD. reassignment VISION TECH BIO PTY. LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: COMMONWEALTH SCIENTIFIC AND INDUSTRIAL RESEARCH ORGANISATION
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57488Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds identifable in body fluids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to determining the presence and/or level of biomarkers for detecting or diagnosing colorectal cancer.
  • the invention also relates to diagnostic kits comprising reagents for determining the presence and/or level of the biomarkers and methods of detecting or diagnosing colorectal cancer.
  • Colorectal cancer also referred to as colon cancer or bowel cancer
  • colon cancer is the second most common cause of cancer worldwide.
  • greater than 90% of patients who present while the tumour is still localised will still be alive after 5 years and can be considered cured.
  • the early detection of colorectal lesions would therefore significantly reduce the impact of colon cancer (Etzioni et al., 2003).
  • FOBT faecal occult blood test
  • Flexible sigmoidoscopy a faecal occult blood test
  • colonoscopy a faecal occult blood test
  • FOBT has relatively low specificity resulting in a high rate of false positives. All positive FOBT must therefore be followed up with colonoscopy. Sampling is done by individuals at home and requires at least two consecutive faecal samples to be analysed to achieve optimal sensitivity. Some versions of the FOBT also require dietary restrictions prior to sampling.
  • FOBT also lacks sensitivity for early stage cancerous lesions that do not bleed into the bowel and as stated above, these are the lesions for which treatment is most successful.
  • the present inventors investigated over sixty biomarkers associated with colorectal cancer, but found that none of the biomarkers alone would be suitable as a diagnostic test. Surprisingly, it was found that determining the presence and/or level of at least two biomarkers associated with colorectal cancer in a sample from a subject allowed for the detection or diagnosis of colorectal cancer at any of the stages of disease. Determining the presence and/or level of at least two biomarkers advantageously provides a diagnostic test that is at least comparable in sensitivity and specificity to the FOBT.
  • the present invention provides a method for diagnosing or detecting colorectal cancer in a subject, the method comprising:
  • the method comprises determining the presence and/or level of two biomarkers selected from M2PK, EpCam, IL-13, DKK-3, IL-8 and IGFBP2.
  • the method comprises determining the presence and/or level of expression of at least three of the biomarkers.
  • the three biomarkers are selected from M2PK, EpCam, IL-13, DKK-3, IL-8, IGFBP2, MIP1 ⁇ , TGF ⁇ 1 and MAC2BP.
  • the method comprises determining the presence and/or level of three biomarkers, wherein the three biomarkers are:
  • IL-8 IL-8, IL-13, and MAC2BP.
  • the method comprises determining the presence and/or level of expression of at least four of the biomarkers.
  • the method comprises determining the presence and/or level of four biomarkers, wherein the four biomarkers are:
  • IL-8 IL-8, MAC2BP, IGFBP2, and EpCam.
  • the method comprises determining the presence and/or level of at least five of the biomarkers.
  • the five biomarkers are IL-8, IGFBP2, MAC2BP, M2PK, and IL-13.
  • the method comprises determining the presence and/or level of at least six of the biomarkers.
  • the method comprises determining the presence and/or level of at least seven of the biomarkers.
  • the seven biomarkers are:
  • IL-8 IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, and TGF ⁇ 1;
  • IL-8 IL-8, IGFBP2, MAC2BP, M2PK, IL-13, EpCam, and MIP1 ⁇ .
  • the method comprises determining the presence and/or level of at least eight of the biomarkers.
  • the method comprises determining the presence and/or level of at least nine of the biomarkers.
  • the method comprises determining the presence and/or level of at least ten of the biomarkers.
  • the method comprises determining the presence and/or level of a combination of biomarkers as provided in any of Tables 7 to 18.
  • the method comprises detecting the presence and/or level of least one additional biomarker selected from IGF-I, IGF-II, IGF-BP2, Amphiregulin, VEGFA, VEGFD, MMP-1, MMP-2, MMP-3, MMP-7, MMP-9, TIMP-1, TIMP-2, ENA-78, MCP-1, MIP-1 ⁇ , IFN- ⁇ , IL-10, IL-13, IL-1 ⁇ , IL-4, IL-8, IL-6, MAC2BP, Tumor M2 pyruvate kinase, M65, OPN, DKK-3, EpCam, TGF ⁇ -1, and VEGFpan.
  • additional biomarker selected from IGF-I, IGF-II, IGF-BP2, Amphiregulin, VEGFA, VEGFD, MMP-1, MMP-2, MMP-3, MMP-7, MMP-9, TIMP-1, TIMP-2, ENA-78, MCP-1, MIP-1 ⁇ , IFN- ⁇ , IL
  • the method diagnoses or detects colorectal cancer with a sensitivity of at least 50%.
  • the method diagnoses or detects colorectal cancer with a sensitivity of at least 66%.
  • the method diagnoses or detects colorectal cancer with a sensitivity of at least 77%.
  • the method diagnoses or detects colorectal cancer with a specificity of at least 75%.
  • the method diagnoses or detects colorectal cancer with a specificity of at least 80%.
  • the method diagnoses or detects colorectal cancer with a specificity of at least 90%.
  • the method diagnoses or detects colorectal cancer with a specificity of at least 95%.
  • the method diagnoses or detects Dukes Stage A colorectal cancer with a sensitivity of at least 50% and a specificity of at least 95%.
  • the method diagnoses or detects Dukes Stage A colorectal cancer with a sensitivity of at least 60% and a specificity of at least 80%.
  • the method diagnoses or detects Dukes Stage A colorectal cancer with a sensitivity of at least 50% and a specificity of at least 90%.
  • the method diagnoses or detects TNM Classification T1, N0, M0 or T2, N0, M0 colorectal cancer with a sensitivity of at least 50% and a specificity of at least 95%.
  • the method diagnoses or detects TNM Classification T1, N0, M0 or T2, N0, M0 colorectal cancer with a sensitivity of at least 60% and a specificity of at least 80%.
  • the method diagnoses or detects TNM Classification T1, N0, M0 or T2, N0, M0 colorectal cancer with a sensitivity of at least 50% and a specificity of at least 90%.
  • the method comprises contacting the sample with at least one compound that binds a biomarker polypeptide.
  • the method comprises detecting the polypeptides by mass spectrometry.
  • the compound is detectably labelled.
  • the compound is an antibody.
  • the compound is bound to a solid support.
  • determining the presence and/or level of the biomarker may comprise determining the presence and/or level of a polynucleotide encoding the biomarker, such as a biomarker gene transcript.
  • the biomarkers are polynucleotides.
  • the method comprises:
  • the sample comprises blood, plasma, serum, urine, platelets, magakaryocytes or faeces.
  • the present invention provides a method of treatment comprising:
  • the present invention provides a method for monitoring the efficacy of treatment of colorectal cancer in a subject, the method comprising treating the subject for colorectal cancer and then detecting the presence and/or level of at least two biomarkers selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1 ⁇ , TGF ⁇ 1, and TIMP-1 in a sample from the subject, wherein an absence of and/or reduction in the level of expression of the polypeptides after treatment when compared to before treatment is indicative of effective treatment.
  • biomarkers selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1 ⁇ , TGF ⁇ 1, and TIMP-1
  • the present invention provides an array of at least two compounds for the diagnosis or detection of colorectal cancer, wherein each of the compounds binds a different biomarker polypeptide selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1 ⁇ , TGF ⁇ 1, and TIMP-1.
  • the present invention provides a kit for diagnosing or detecting colorectal cancer in a subject, the kit comprising two compounds that each binds a different biomarker polypeptide selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1 ⁇ , TGF ⁇ 1, and TIMP-1.
  • FIG. 1 In Study 3 an optimum combination of the 46 potential protein biomarkers was found using logistic regression modelling, resulting in a panel of seven biomarkers and is illustrated as a ROC curve (black curve). The performance of this “panel” on independent data was estimated using “leave one out” cross-validation (grey curve). The vertical lines are drawn at points of 80% and 90% specificity—operating points of interest in screening tests. Performance statistics are given in Table 5.
  • FIG. 2 Performance of a seven biomarker model identifying colorectal cancer patients from normals at each Dukes Stage illustrated by ROC curves for each stage.
  • FIG. 3 When biomarker results from Study 4 (also referred to as Study 3 remeasured) were modelled in pairs a total of 5 pairs (out of a possible 45 combinations selected from the list of 10 biomarkers above) could be shown to produce a sensitivity above 52% at a specificity of 95.
  • FIG. 4 An example of a 3 biomarker model generated from Study 4 data which had a sensitivity of at least 50% at 95% specificity. There were 968 possible 3-biomarker combinations and approximately half of those combinations showed a performance of at least 50% sensitivity at 90% specificity.
  • FIG. 6 Frequency of each biomarker in the best 485 models. These BMs represent all serum models that gave a sensitivity of at least 50% at 95% The high representation of all 10 biomarkers in the useful models demonstrates the unity of our selection of these 10 biomarkers.
  • FIG. 7 A 5 biomarker model generated from Study 4 data is illustrated as a ROC curve (black) and cross validated ROC curve (grey). This model shows a sensitivity of 68% at 95% specificity when all stages of disease are included and when cross validated gave a sensitivity of 64%. Biomarkers included are [IL-8, IGFBP2, Mac2BP, DKK-3 and M2PK].
  • FIG. 8 A 6 biomarker model generated from Study 4 data is illustrated as a ROC curve (black) and cross validated ROC curve (Grey). This model shows a sensitivity of 77% at a specificity of 95% when all stages of disease are included and when cross validated gave a sensitivity of 67%.
  • Biomarkers included are [IL-8, IGFBP2, Mac2BP, DKK-3, TGFbeta1&M2PK].
  • FIG. 9 Two alternative seven biomarker models generated from Study 3a data are shown. One was optimised for high specificity (black/new) and an alternative or model optimised for area under the curve is shown (grey/old). At 90% specificity the sensitivity was 72% for the new model and 77% for the older model. Biomarkers included were as follows:
  • FIG. 10 A seven biomarker model generated from Study 4 data is illustrated as a ROC curve (black) and cross validated ROC curve (grey). This model shows, a sensitivity of 84% at a specificity of 95%.
  • Biomarkers included are [M2PK serum, IL8.plasma, TGF beta1.serum, IGFBP2.plasma, Mac2BP.serum, TIMP1.plasma and Dkk3 plasma.
  • FIG. 11 Cross validated ROC curves showing the performance of a 3 biomarker model for each Dukes stage is illustrated.
  • This data demonstrates the validity of the choice of three biomarkers (DKK-3, M2PK and IGFBP2) for detecting cancer at different stages of the disease progression.
  • the data indicates that at Stage A if the three markers are used, the test still will achieve a significant sensitivity of 64% at 95% specificity which is comparable to the sensitivity achieved at late stage disease (79%). That is the biomarker panel of three will pick up early disease states allowing early detection.
  • Biomarkers included are Dkk3, M2PK and IGFBP2.
  • the recombinant protein, cell culture, and immunological techniques utilized in the present invention are standard procedures, well known to those skilled in the art. Such techniques are described and explained throughout the literature in sources such as, J. Perbal, A Practical Guide to Molecular Cloning, John Wiley and Sons (1984), J. Sambrook et al., Molecular Cloning: A Laboratory Manual, 3 rd edn, Cold Spring Harbour Laboratory Press (2001), R. Scopes, Protein Purification—Principals and Practice, 3 rd edn, Springer (1994), T. A. Brown (editor), Essential Molecular Biology: A Practical Approach, Volumes 1 and 2, IRL Press (1991), D. M. Glover and B. D.
  • colonal cancer also known as “colon cancer”, “bowel cancer” or “rectal cancer” refers to all forms of cancer originating from the epithelial cells lining the large intestine and/or rectum.
  • biomarker refers to any molecule, such as a gene, gene transcript (for example mRNA), peptide or protein or fragment thereof produced by a subject which is useful in differentiating subjects having colorectal cancer from normal or healthy subjects.
  • diagnosis and variants thereof such as, but not limited to, “diagnose”, “diagnosed” or “diagnosing” shall not be limited to a primary diagnosis of a clinical state, but should be taken to include diagnosis of recurrent disease.
  • the term “subject” refers to any animal that may develop colorectal cancer and includes animals such as mammals, e.g. humans, or non-human mammals such as cats and dogs, laboratory animals such as mice, rats, rabbits or guinea pigs, and livestock animals. In a preferred embodiment, the subject is a human.
  • sample may be of any suitable type and may refer, e.g., to a material in which the presence or level of biomarkers can be detected.
  • the sample is obtained from the subject so that the detection of the presence and/or level of biomarkers may be performed in vitro. Alternatively, the presence and/or level of biomarkers can be detected in vivo.
  • the sample can be used as obtained directly from the source or following at least one step of (partial) purification.
  • the sample can be prepared in any convenient medium which does not interfere with the method of the invention.
  • the sample is an aqueous solution, biological fluid, cells or tissue.
  • the sample is blood, plasma, serum, urine, platelets, megakaryocytes or faeces.
  • Pre-treatment may involve, for example, preparing plasma from blood, diluting viscous fluids, and the like.
  • Methods of treatment can involve filtration, distillation, separation, concentration, inactivation of interfering components, and the addition of reagents.
  • the selection and pre-treatment of biological samples prior to testing is well known in the art and need not be described further.
  • treating include administering a therapeutically effective amount of a compound sufficient to reduce or delay the onset or progression of colorectal cancer, or to reduce or eliminate at least one symptom of colorectal cancer.
  • the present inventors have shown that determining the presence and/or level of least two biomarkers in a sample from a subject allows for the detection or diagnosis of colorectal cancer, either early detection at Dukes Stage A or at some later stage such as Dukes Stage B or C or D, with specificity and sensitivity comparable to or greater than that achieved with the FOBT.
  • the at least two biomarkers that are useful in the methods of the present invention are selected from IL-8 (interleukin-8), IGFBP2 (insulin-like growth factor binding protein-2), MAC2BP (MAC2-binding protein; serum protein 90K), M2PK (pyruvate kinase muscle 2, pyruvate kinase 3), IL-13 (interleukin-13), DKK-3 (dickkopf homolog, 3), EpCAM (epithelial cell adhesion molecule), MIP1 ⁇ (macrophage inflammatory protein 1 ⁇ , CCL4, MIP1beta), TGF ⁇ 1 (transforming growth factor ⁇ 1 , TGFbeta1) and TIMP-1 (tissue inhibitor of metalloproteinase 1).
  • IL-8 interleukin-8
  • IGFBP2 insulin-like growth factor binding protein-2
  • MAC2BP MAC2-binding protein
  • serum protein 90K serum protein 90K
  • M2PK pyruvate kinase muscle 2, pyruv
  • references to any of these biomarkers includes reference to all polypeptide and polynucleotide variants such as isoforms and transcript variants as would be known by the person skilled in the art. NCBI accession numbers of representative sequences for each of the biomarkers are provided in Table 1.
  • the diagnostic methods of the present invention may involve a degree of quantification to determine levels biomarkers in patient samples. Such quantification is readily provided by the inclusion of appropriate control samples.
  • internal controls are included in the methods of the present invention.
  • a preferred internal control is one or more samples taken from one or more healthy individuals.
  • the term “healthy individual” shall be taken to mean an individual who is known not to suffer from colorectal cancer, such knowledge being derived from clinical data on the individual, including, but not limited to, a different diagnostic assay to that described herein.
  • control when internal controls are not included in each assay conducted, the control may be derived from an established data set.
  • Data pertaining to the control subjects are preferably selected from the group consisting of:
  • a data set comprising measurements of the presence or level of expression of biomarkers for a typical population of subjects known to have colorectal cancer
  • a data set comprising measurements of the presence or level of biomarkers for the subject being tested wherein said measurements have been made previously, such as, for example, when the subject was known to be healthy or, in the case of a subject having colorectal cancer, when the subject was diagnosed or at an earlier stage in disease progression;
  • a data set comprising measurements of the presence or level of biomarkers for a healthy individual or a population of healthy individuals
  • a data set comprising measurements of the presence or level of biomarkers for a normal individual or a population of normal individuals.
  • the term “typical population” with respect to subjects known to have colorectal cancer shall be taken to refer to a population or sample of subjects diagnosed with colorectal cancer that is representative of the spectrum of colorectal cancer patients. This is not to be taken as requiring a strict normal distribution of morphological or clinicopathological parameters in the population, since some variation in such a distribution is permissible.
  • a “typical population” will exhibit a spectrum of colorectal cancer at different stages of disease progression. It is particularly preferred that a “typical population” exhibits the expression characteristics of a cohort of subjects as described herein.
  • normal individual shall be taken to mean an individual that does not express a biomarker, or expresses a biomarker at a low level in a sample.
  • data obtained from a sufficiently large sample of the population will normalize, allowing the generation of a data set for determining the average level of a particular biomarker.
  • Compounds that bind a biomarker when used diagnostically may be linked to a diagnostic reagent such as a detectable label to allow easy detection of binding events in vitro or in vivo.
  • a diagnostic reagent such as a detectable label to allow easy detection of binding events in vitro or in vivo.
  • Suitable labels include radioisotopes, dye markers or other imaging reagents for detection and/or localisation of target molecules.
  • Compounds linked to a detectable label can be used with suitable in vivo imaging technologies such as, for example, radiology, fluoroscopy, nuclear magnetic resonance imaging (MRI), CAT-scanning, positron emission tomography (PET), computerized tomography etc.
  • the diagnostic methods of the present invention are able to diagnose or detect colorectal cancer with a sensitivity and specificity that is at least comparable to FOBT, or greater.
  • sensitivity refers to the proportion of actual positives in the diagnostic test which are correctly identified as having colorectal cancer.
  • Specificity measures the proportion of negatives which are correctly identified as not having colorectal cancer.
  • the methods of the invention are able to diagnose or detect colorectal cancer with a sensitivity of at least 50%, 60% or 66%, or at least 77%, 80%, 83%, 85%, 86%, 87%, 88%, 89%, 90%, or at least 93%.
  • the methods of the invention are able to diagnose or detect colorectal cancer with a sensitivity of at least 80%, or at least 85% or at least 90%, or at least 95%.
  • the methods of the invention are able to diagnose or detect colorectal cancer with a specificity of at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94% or at least 95%.
  • the methods of the present invention are able to detect colorectal cancer at all of the Dukes Stages with greater sensitivity than the FOBT.
  • the tumor has penetrated into, but not through, the bowel wall.
  • the tumor has penetrated through the bowel wall but there is not yet any lymph node involvement.
  • the cancer involves regional lymph nodes.
  • there is distant metastasis for example, to the liver or lung.
  • the methods of the present invention are able to diagnose or detect colorectal cancer at any Dukes Stage with a sensitivity of at least 80%.
  • TMM Malignant Tumors
  • AJCC American Joint Committee on Cancer
  • MAC Modified Astler-Coller classification
  • the Dukes Stages correspond to certain TNM Classifications.
  • Dukes Stage A corresponds to T1, T2, N0 and M0
  • Dukes Stage B corresponds to T3, T4a, T4b, N0 and M0
  • Dukes Stage C corresponds to i) T1-T2, N1/N1c, M0; ii) T1, N2a and M0; iii) T3-T4a, N1/N1c and M0; iv) T2-T3, N2a and M0; v) T1-T2, N2b and M0; vi) T4a, N2a and M0; vii) T3-T4a, N2b and M0; and viii) T4b, N1-N2 and M0.
  • reference to a Dukes Stage as used herein includes reference to the corresponding TMN classification as known in the art.
  • biomarker polypeptide is detected in a patient sample, wherein the presence and/or level of the polypeptide in the sample is indicative of colorectal cancer.
  • the method may comprise contacting a biological sample derived from the subject with a compound capable of binding to a biomarker polypeptide, and detecting the formation of complex between the compound and the biomarker polypeptide.
  • biomarker polypeptide as used herein includes fragments of biomarker polypeptides, including for example, immunogenic fragments and epitopes of the biomarker polypeptide.
  • the compound that binds the biomarker is an antibody.
  • antibody as used herein includes intact molecules as well as molecules comprising or consisting of fragments thereof, such as, for example Fab, F(ab′)2, Fv and scFv, as well as engineered variants including diabodies, triabodies, mini-bodies and single-domain antibodies which are capable of binding an epitopic determinant.
  • antibodies may exist as intact immunoglobulins, or as modifications in a variety of forms.
  • an antibody to a biomarker polypeptide is detected in a patient sample, wherein the presence and/or level of the antibody in the sample is indicative of colorectal cancer.
  • Preferred detection systems contemplated herein include any known assay for detecting proteins or antibodies in a biological sample isolated from a human subject, such as, for example, SDS/PAGE, isoelectric focussing, 2-dimensional gel electrophoresis comprising SDS/PAGE and isoelectric focussing, an immunoassay, flow cytometry e.g. fluorescence-activated cell sorting (FACS), a detection based system using an antibody or non-antibody compound, such as, for example, a small molecule (e.g. a chemical compound, agonist, antagonist, allosteric modulator, competitive inhibitor, or non-competitive inhibitor, of the protein).
  • FACS fluorescence-activated cell sorting
  • the antibody or small molecule may be used in any standard solid phase or solution phase assay format amenable to the detection of proteins.
  • Optical or fluorescent detection such as, for example, using mass spectrometry, MALDI-TOF, biosensor technology, evanescent fiber optics, or fluorescence resonance energy transfer, is clearly encompassed by the present invention.
  • Assay systems suitable for use in high throughput screening of mass samples e.g. a high throughput spectroscopy resonance method (e.g. MALDI-TOF, electrospray MS or nano-electrospray MS), are also contemplated.
  • Another suitable protein detection technique involves the use of Multiple Reaction Monitoring (MRM) in LC-MS (LC/MRM-MS) (Anderson and Hunter, 2006).
  • MRM Multiple Reaction Monitoring
  • Immunoassay formats are particularly suitable, e.g., selected from the group consisting of, an immunoblot, a Western blot, a dot blot, an enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), enzyme immunoassay.
  • Modified immunoassays utilizing fluorescence resonance energy transfer (FRET), isotope-coded affinity tags (ICAT), matrix-assisted laser desorption/ionization time of flight (MALDI-TOF), electrospray ionization (ESI), biosensor technology, evanescent fiber-optics technology or protein chip technology are also useful.
  • nucleic acid molecule or “polynucleotide” as used herein refer to an oligonucleotide, polynucleotide or any fragment thereof.
  • Comparison may be made by reference to a standard control, or to a control level that is found in healthy tissue.
  • levels of a transcribed gene can be determined by Northern blotting, and/or RT-PCR.
  • quantitative (real-time) PCR quantitative analysis of gene expression can be achieved by using appropriate primers for the gene of interest.
  • the nucleic acid may be labelled and hybridised on a gene array, in which case the gene concentration will be directly proportional to the intensity of the radioactive or fluorescent signal generated in the array.
  • PCR methods that may be used in carrying out the invention include hybridization based PCR detection systems, TaqMan assay (U.S. Pat. No. 5,962,233) and the molecular beacon assay (U.S. Pat. No. 5,925,517).
  • RNA may be isolated from a sample to be analysed using conventional procedures, such as are supplied by QIAGEN technology. This RNA is then reverse-transcribed into DNA using reverse transcriptase and the DNA molecule of interest may then be amplified by PCR techniques using specific primers.
  • Hybridisation or amplification assays such as, for example, Southern or Northern blot analysis, immunohistochemistry, single-stranded conformational polymorphism analysis (SSCP) and PCR analyses are among techniques that are useful in this respect.
  • target or probe nucleic acid may be immobilised to a solid support such as a microtitre plate, membrane, polystyrene bead, glass slide or other solid phase.
  • kits for the diagnosis or detection of colorectal cancer may be suitable for detection of nucleic acid species, or alternatively may be for detection of a polypeptide gene product, as discussed above.
  • kits For detection of polypeptides, antibodies will most typically be used as components of kits. However, any agent capable of binding specifically to a biomarker gene product will be useful in this aspect of the invention.
  • Other components of the kits will typically include labels, secondary antibodies, substrates (if the gene is an enzyme), inhibitors, co-factors and control gene product preparations to allow the user to quantitate expression levels and/or to assess whether the diagnosis experiment has worked correctly. Enzyme-linked immunosorbent assay-based (ELISA) tests and competitive ELISA tests are particularly suitable assays that can be carried out easily by the skilled person using kit components.
  • the kit further comprises means for the detection of the binding of an antibody to a biomarker polypeptide.
  • a reporter molecule such as, for example, an enzyme (such as horseradish peroxidase or alkaline phosphatase), a dye, a radionucleotide, a luminescent group, a fluorescent group, biotin or a colloidal particle, such as colloidal gold or selenium.
  • an enzyme such as horseradish peroxidase or alkaline phosphatase
  • a dye such as horseradish peroxidase or alkaline phosphatase
  • a radionucleotide such as a radionucleotide
  • a luminescent group such as a fluorescent group
  • biotin or a colloidal particle such as colloidal gold or selenium.
  • a colloidal particle such as colloidal gold or selenium.
  • a kit may additionally comprise a reference sample.
  • a reference sample comprises a polypeptide that is detected by an antibody.
  • the polypeptide is of known concentration.
  • Such a polypeptide is of particular use as a standard. Accordingly, various known concentrations of such a polypeptide may be detected using a diagnostic assay described herein.
  • kits may contain a first container such as a vial or plastic tube or a microtiter plate that contains an oligonucleotide probe.
  • the kits may optionally contain a second container that holds primers.
  • the probe may be hybridisable to DNA whose altered expression is associated with colorectal cancer and the primers are useful for amplifying this DNA.
  • Kits that contain an oligonucleotide probe immobilised on a solid support could also be developed, for example, using arrays (see supplement of issue 21(1) Nature Genetics, 1999).
  • nucleic acid primers may be included in the kit that are complementary to at least a portion of a biomarker gene as described herein.
  • the set of primers typically includes at least two oligonucleotides, preferably four oligonucleotides, that are capable of specific amplification of DNA.
  • Fluorescent-labelled oligonucleotides that will allow quantitative PCR determination may be included (e.g. TaqMan chemistry, Molecular Beacons). Suitable enzymes for amplification of the DNA, will also be included.
  • Control nucleic acid may be included for purposes of comparison or validation. Such controls could either be RNA/DNA isolated from healthy tissue, or from healthy individuals, or housekeeping genes such as ⁇ -actin or GAPDH whose mRNA levels are not affected by colorectal cancer.
  • test performance In order to develop a panel of biomarkers suitable for diagnosing or detecting colorectal cancer, the present inventors have analysed numerous biomarkers in a statistical model. Such an improvement in the performance of a test is sometimes referred to as the “in-sample” performance.
  • a fair evaluation of a test requires its assessment using out-of-sample subjects, that is, subjects not included in the construction of the initial predictive model. This is achieved by assessing the test performance using cross validation.
  • Tests for statistical significance include linear and non linear regression, including ANOVA, Kruskal-Wallis, Wilcoxon, Mann-Whitney and odds ratio, Baysian probability algorithms. As the number of biomarkers measured increases however, it can be generally more convenient to use a more sophisticated technique such as Random Forests, simple logistic, Bayes Net to name a few.
  • Bayesian probability may be adopted.
  • a 10-fold cross-validation can be used to estimate the “out-of-sample” performance of the models in question.
  • the data can be divided randomly into 10 sub-samples, each with similar proportions of healthy subject and subjects at each stage of disease.
  • each subsample can be excluded, and a logistic model built using the remaining 90% of the subjects.
  • This model can then be used to estimate the probability of cancer for the excluded sub-sample, providing an estimate of “out-of-sample” performance.
  • “out-of-sample” performance can be estimated from the study data itself.
  • These out-of sample predicted probabilities can then be compared with the actual disease status of the subjects to create a Receiver Operating Characteristic (ROC) Curve, from which the cross-validated sensitivity at 95% specificity may be estimated.
  • ROC Receiver Operating Characteristic
  • a model discriminating subjects with cancer from healthy controls can be as follows:
  • log ⁇ ( p 1 - p ) ⁇ 0 + ⁇ IL ⁇ ⁇ 8 ⁇ C IL ⁇ ⁇ 8 + ⁇ IGFBP ⁇ ⁇ 2 ⁇ C IGFBP ⁇ ⁇ 2 + ⁇ MAC ⁇ ⁇ 2 ⁇ ⁇ BP ⁇ C MAC ⁇ ⁇ 2 ⁇ ⁇ BP + ⁇ M ⁇ ⁇ 2 ⁇ ⁇ PK ⁇ C M ⁇ ⁇ 2 ⁇ PK + ⁇ DKK ⁇ ⁇ 3 ⁇ C DKK ⁇ ⁇ 3
  • Each C i is the logarithm of concentration biomarker i in the plasma (or serum) of a person.
  • Each beta ( ⁇ ) is a coefficient applying to that biomarker in the concentration units in which it is measured— ⁇ 0 is an “offset” or “intercept”. This linear logistic model is common to all results presented herein, but is far from the only way in which a combination of biomarker concentrations may be modelled to predict the probability of cancer.
  • the present invention also provides software or hardware programmed to implement an algorithm that processes data obtained by performing the method of the invention via a multivariate analysis to provide a disease score and provide or permit a diagnosis or detection of colorectal cancer and/or determine progression or status of a colorectal cancer or determine whether or not a colorectal cancer has progressed or determine whether or not a subject is responding to treatment for colorectal cancer in accordance with the results of the disease score in comparison with predetermined values.
  • a method of the invention may be used in existing knowledge-based architecture or platforms associated with pathology services. For example, results from a method described herein are transmitted via a communications network (e.g. the internet) to a processing system in which an algorithm is stored and used to generate a predicted posterior probability value which translates to the score of disease probability or risk of recurrence or metastasis or responsiveness to treatment which is then forwarded to an end user in the form of a diagnostic or predictive report.
  • a communications network e.g. the internet
  • the method of the invention may, therefore, be in the form of a kit or computer-based system which comprises the reagents necessary to detect the concentration of the biomarkers and the computer hardware and/or software to facilitate determination and transmission of reports to a clinician.
  • the assay of the present invention permits integration into existing or newly developed pathology architecture or platform systems.
  • the present invention contemplates a method of allowing a user to determine the status of a subject with respect to colorectal cancer, the method including:
  • the method for diagnosing or detecting colorectal cancer of the invention may be performed by taking a blood sample from a patient and determining the presence and/or level of any one or more of the biomarkers as described herein. If desired, the measurements may be made, for example, on a biochip so that a single analysis can be used to measure the presence and/or level of multiple biomarkers. The results of this analysis may then be inputted into into a computer program that subjects them to linear regression analysis. The computer could also contain information as to control values or expected ranges, or the clinician, nurse, medical administrator or general practitioner could input such data. This analysis wold then provide a score or likelihood of having colorectal cancer. If a second test for the patient is performed, the regression analysis may indicate a change in the score, thus indicating that the patient's disease has progressed or worsened.
  • a collection of plasma and serum samples was taken and processed from a cohort of colorectal cancer patients (Dukes Stages A-D) that were being treated at several hospitals.
  • Blood was also collected and processed from a group of about 50 healthy volunteers over the age of 65 and from a group of 15 over the age of 50.
  • Study 1 looked at 52 cancer samples and 50 controls
  • study 2 looked at 55 cancer samples and 53 controls
  • study 3 and 4 looked at 96 cancer samples and 50 controls.
  • study 2 3 and 4 the patients were age and gender matched across Dukes Stages, see Table 2 for summary statistics.
  • Biomarkers chosen to be measured in Study 1 and 2 and 3 are listed in Table 3. Biomarkers in bold were those identified as promising from each study (i.e. they were significantly different in samples from colorectal cancer patients versus control and/or they were identified in panels of combined biomarkers that distinguish colorectal cancer from controls).
  • the resulting model was then used to estimate the probability that the excluded observation is a case. This was repeated for each observation in the dataset. In this way each observation in turn acted as an independent test of the model-building algorithm.
  • the resulting dataset consisting of cases and controls each with an “independently predicted” case probability can then be compared with the original model.
  • the ability to choose from numerous biomarkers and weight them appropriately allows a search strategy which optimises performance in regions of interest on the ROC curve.
  • the cost of poor specificity is large numbers of unnecessary colonoscopies.
  • biomarkers were evaluated to select a candidate panel of colorectal cancer biomarkers, using block randomization within plates to avoid bias. From this list of 48 only 42 showed measurable levels. Individually 14 biomarkers showed significant difference between controls and CRC as assessed by t-tests; (IGFII, IGFBP2, IL-8, IL-6, MMP-1, MMP-7, s90/Mac2BP, M2PK, EpCam, TIMP-1 (serum and plasma), M65, OPN, TGF ⁇ 1, VEGFpan. As expected, none had sufficient sensitivity or specificity to be useful as a biomarker by itself (not shown). However, using a variety of modelling strategies, including use of logarithmic values, several different panels of biomarkers were found that exceeded the performance of FOBT especially for early to late stage disease.
  • FIG. 1 shows the results from a 7 biomarker panel which included IL8 (serum), IL-13 (serum), EpCAM (plasma), M2PK (plasma), IGFBP2 (serum) and Mac2BP (serum) and which was cross validated to predict its performance on independent samples.
  • log ⁇ ( p 1 - p ) ⁇ 0 + ⁇ IL ⁇ ⁇ 8 ⁇ C IL ⁇ ⁇ 8 + ⁇ IGFBP ⁇ ⁇ 2 ⁇ C IGFBP ⁇ ⁇ 2 + ⁇ MAC ⁇ ⁇ 2 ⁇ ⁇ BP ⁇ C MAC ⁇ ⁇ 2 ⁇ ⁇ BP + ⁇ M ⁇ ⁇ 2 ⁇ ⁇ PK ⁇ C M ⁇ ⁇ 2 ⁇ PK + ⁇ DKK ⁇ ⁇ 3 ⁇ C DKK ⁇ ⁇ 3
  • biomarkers were remeasured in the same cohort as Study 3. Blood was collected from 96 colorectal cancer patients and 50 normal subjects (the controls). In this study the focus was on 10 biomarkers, namely IGFBP2, IL8, IL13, Mac2BP, M2PK, Dkk3, EpCam, TGFbeta1, TIMP-1, MIP1beta. Assays were performed as described previously. Both serum and plasma levels of each of the biomarkers were measured and compared with control values.
  • the 968 combinations of between 3 and 10 biomarkers consist of the 120 combinations of 3 marker; 210 combinations of 4 markers; 252 combinations of 5 markers; 210 combinations of 6 markers; 120 combinations of 7 markers; 45 combinations of 8 markers; 10 combinations of 9 markers and the single combination that includes all 10 biomarkers.
  • the 968 combinations had a sensitivity of 50% at a specificity of 95%, see FIG. 4 which shows the results for a three biomarker combination. More than half of these combinations would have a specificity of 90% and a sensitivity of 50%.
  • FIG. 5 shows all 485 of the estimated out-of sample (10-fold cross-validated) ROC curves for tests out of a total possible 968 models based on all possible combinations of 3 to 10 of the biomarkers. Note that many individual segments of the 485 ROC curves are coincident, due to as each horizontal segment represents one control and each vertical segment one case. In this instance 50.1% of the combinations have exceeded the 50% sensitivity, 95% specificity, The best estimated “out-of-sample” performance is a sensitivity of 76% at 95% specificity. Repeating the cross-validation will select a different set of models—the sensitivity of any one combination may vary by 10% at 95% specificity due to random sampling—but result in a similar proportion of useful “useful screening tests”. Precise validation of individual models requires repeated experiments and larger sample sizes.
  • FIG. 6 shows how many of the 485 combinations with 50% sensitivity, 95% specificity, include any given biomarker.
  • 432 of the chosen “useful” combinations include M2PK.
  • At the low end 227 of the chosen “useful” combinations include MIP1beta. This high representation of all 10 biomarkers in “useful” models shows the unity and self-complementarity of the selection of these 10 biomarkers.
  • FIG. 7 to FIG. 11 demonstrate some of the results from this last study (Study 4) for combinations of 5 and 7 biomarkers, including a model where the samples are either from plasma or serum cluster.
  • FIG. 11 demonstrates the validity of the choice of three biomarkers (DKK-3, M2PK and IGFBP2) at different stages of the disease progression. The data indicates that at Stage A if the three markers are used, the test still will achieve a significant sensitivity (64%) at 95% specificity which is comparable to the sensitivity achieved at late stage disease, 79%). That is the biomarker panel of three will pick up early disease states allowing early detection.
  • Tables 8 to 16 list results from various combinations of various biomarker panel sets. Depending on the linear regression that is used, as well as the cohort control and other factors such as sample derivation and assay kit technique, there may be a variation on the actual figures or order of the markers. Regardless, many of these combinations will achieve good selectivity at high specificities so as to be useful for diagnosing or detecting colorectal cancer at any stage of the disease progression.

Abstract

The present invention provides a method for diagnosing or detecting colorectal cancer in a subject, the method comprising determining the presence and/or level of biomarkers selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1β, TGFβ1, and TIMP-1. The invention also relates to diagnostic kits comprising reagents for determining the presence and/or level of the biomarkers and methods of detecting or diagnosing colorectal cancer.

Description

    FIELD OF THE INVENTION
  • The present invention relates to determining the presence and/or level of biomarkers for detecting or diagnosing colorectal cancer. The invention also relates to diagnostic kits comprising reagents for determining the presence and/or level of the biomarkers and methods of detecting or diagnosing colorectal cancer.
  • BACKGROUND OF THE INVENTION
  • Colorectal cancer, also referred to as colon cancer or bowel cancer, is the second most common cause of cancer worldwide. There is an annual incidence of almost a million colorectal cancer cases with an annual mortality around 500,000 (Cancer in Australia: an overview, 2008). Unfortunately, 30-50% of patients have occult or overt metastases at presentation and once tumours have metastasized prognosis is very poor with a five year survival of less than 10% (Etzioni et al., 2003). By contrast, greater than 90% of patients who present while the tumour is still localised will still be alive after 5 years and can be considered cured. The early detection of colorectal lesions would therefore significantly reduce the impact of colon cancer (Etzioni et al., 2003).
  • The current screening assays in widespread use for the diagnosis of colorectal cancer are the faecal occult blood test (FOBT), flexible sigmoidoscopy, and colonoscopy (Lieberman, 2010). FOBT has relatively low specificity resulting in a high rate of false positives. All positive FOBT must therefore be followed up with colonoscopy. Sampling is done by individuals at home and requires at least two consecutive faecal samples to be analysed to achieve optimal sensitivity. Some versions of the FOBT also require dietary restrictions prior to sampling. FOBT also lacks sensitivity for early stage cancerous lesions that do not bleed into the bowel and as stated above, these are the lesions for which treatment is most successful.
  • While FOBT screening does result in reduction of mortality due to colorectal cancer it suffers from a low compliance rate (30-40%), most likely due to the unpalatable nature of the test, which limits its usefulness as a screening tool. Colonoscopy is the current gold standard and has a specificity of greater then 90% but it is intrusive and costly with a small but finite risk of complications (2.1 per 1000 procedures) (Levin, 2004). Development of a rapid, specific, cheap blood based assay would overcome compliance issues commonly seen with other screening tests (Tonus, 2006; Hundt et al., 2007) and would be more acceptable as part of a large screening assay.
  • SUMMARY OF THE INVENTION
  • The present inventors investigated over sixty biomarkers associated with colorectal cancer, but found that none of the biomarkers alone would be suitable as a diagnostic test. Surprisingly, it was found that determining the presence and/or level of at least two biomarkers associated with colorectal cancer in a sample from a subject allowed for the detection or diagnosis of colorectal cancer at any of the stages of disease. Determining the presence and/or level of at least two biomarkers advantageously provides a diagnostic test that is at least comparable in sensitivity and specificity to the FOBT.
  • Accordingly, in one aspect, the present invention provides a method for diagnosing or detecting colorectal cancer in a subject, the method comprising:
  • i) determining the presence and/or level of at least two biomarkers selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1β, TGFβ1, and TIMP-1 in a sample from the subject,
  • wherein the presence and/or level of the two biomarkers is indicative of colorectal cancer.
  • In one embodiment, the method comprises determining the presence and/or level of two biomarkers selected from M2PK, EpCam, IL-13, DKK-3, IL-8 and IGFBP2.
  • In another embodiment, the method comprises determining the presence and/or level of expression of at least three of the biomarkers.
  • In one embodiment, the three biomarkers are selected from M2PK, EpCam, IL-13, DKK-3, IL-8, IGFBP2, MIP1β, TGFβ1 and MAC2BP.
  • In one particular embodiment, the method comprises determining the presence and/or level of three biomarkers, wherein the three biomarkers are:
  • i) DKK-3, M2PK, and IGFBP2;
  • ii) M2PK, IGFBP2, and EpCAM;
  • iii) M2PK, MIP1β, and TGFβ1; or
  • iv) IL-8, IL-13, and MAC2BP.
  • In another embodiment, the method comprises determining the presence and/or level of expression of at least four of the biomarkers.
  • In one particular embodiment, the method comprises determining the presence and/or level of four biomarkers, wherein the four biomarkers are:
  • i) DKK-3, M2PK, MAC2BP, and IGFBP2;
  • ii) IL-8, IL-13, MAC2BP, and EpCam;
  • iii) DKK3, M2PK, TGFβ1, and TIMP-1;
  • iv) M2PK, MIP1β, IL-13, and TIMP-1; or
  • v) IL-8, MAC2BP, IGFBP2, and EpCam.
  • In yet another embodiment, the method comprises determining the presence and/or level of at least five of the biomarkers.
  • In one particular embodiment, the five biomarkers are IL-8, IGFBP2, MAC2BP, M2PK, and IL-13.
  • In another embodiment, the method comprises determining the presence and/or level of at least six of the biomarkers.
  • In another embodiment, the method comprises determining the presence and/or level of at least seven of the biomarkers.
  • In one particular embodiment, the seven biomarkers are:
  • i) IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, and TGF β1; or
  • ii) IL-8, IGFBP2, MAC2BP, M2PK, IL-13, EpCam, and MIP1β.
  • In yet another embodiment, the method comprises determining the presence and/or level of at least eight of the biomarkers.
  • In one embodiment, the method comprises determining the presence and/or level of at least nine of the biomarkers.
  • In yet another embodiment, the method comprises determining the presence and/or level of at least ten of the biomarkers.
  • In another embodiment, the method comprises determining the presence and/or level of a combination of biomarkers as provided in any of Tables 7 to 18.
  • In another embodiment, the method comprises detecting the presence and/or level of least one additional biomarker selected from IGF-I, IGF-II, IGF-BP2, Amphiregulin, VEGFA, VEGFD, MMP-1, MMP-2, MMP-3, MMP-7, MMP-9, TIMP-1, TIMP-2, ENA-78, MCP-1, MIP-1β, IFN-γ, IL-10, IL-13, IL-1β, IL-4, IL-8, IL-6, MAC2BP, Tumor M2 pyruvate kinase, M65, OPN, DKK-3, EpCam, TGFβ-1, and VEGFpan.
  • In one embodiment, the method diagnoses or detects colorectal cancer with a sensitivity of at least 50%.
  • In another embodiment, the method diagnoses or detects colorectal cancer with a sensitivity of at least 66%.
  • In yet another embodiment, the method diagnoses or detects colorectal cancer with a sensitivity of at least 77%.
  • In one embodiment, the method diagnoses or detects colorectal cancer with a specificity of at least 75%.
  • In one embodiment, the method diagnoses or detects colorectal cancer with a specificity of at least 80%.
  • In another embodiment, the method diagnoses or detects colorectal cancer with a specificity of at least 90%.
  • In yet another embodiment, the method diagnoses or detects colorectal cancer with a specificity of at least 95%.
  • In another embodiment, the method diagnoses or detects Dukes Stage A colorectal cancer with a sensitivity of at least 50% and a specificity of at least 95%.
  • In yet another embodiment, the method diagnoses or detects Dukes Stage A colorectal cancer with a sensitivity of at least 60% and a specificity of at least 80%.
  • In another embodiment, the method diagnoses or detects Dukes Stage A colorectal cancer with a sensitivity of at least 50% and a specificity of at least 90%.
  • The skilled person will understand that Dukes Stage A corresponds to TNM Classifications T1, N0, M0 and T2, N0, M0.
  • Thus in one embodiment, the method diagnoses or detects TNM Classification T1, N0, M0 or T2, N0, M0 colorectal cancer with a sensitivity of at least 50% and a specificity of at least 95%.
  • In yet another embodiment, the method diagnoses or detects TNM Classification T1, N0, M0 or T2, N0, M0 colorectal cancer with a sensitivity of at least 60% and a specificity of at least 80%.
  • In another embodiment, the method diagnoses or detects TNM Classification T1, N0, M0 or T2, N0, M0 colorectal cancer with a sensitivity of at least 50% and a specificity of at least 90%.
  • Any suitable technique for the detection of polypeptides may be used in the methods of the invention. In one embodiment, the method comprises contacting the sample with at least one compound that binds a biomarker polypeptide. Alternatively, the method comprises detecting the polypeptides by mass spectrometry.
  • In one particular embodiment, the compound is detectably labelled.
  • In another embodiment, the compound is an antibody.
  • In one embodiment, the compound is bound to a solid support.
  • In the methods of the invention, determining the presence and/or level of the biomarker may comprise determining the presence and/or level of a polynucleotide encoding the biomarker, such as a biomarker gene transcript. Thus, in one embodiment, the biomarkers are polynucleotides.
  • In yet another embodiment of the methods of the invention, the method comprises:
  • i) determining the presence and/or level of the biomarkers in the sample from the subject; and
  • ii) comparing the presence and/or level of the biomarkers to a control, wherein a presence and/or level in the sample that is different to the control is indicative of colorectal cancer.
  • In one embodiment, the sample comprises blood, plasma, serum, urine, platelets, magakaryocytes or faeces.
  • In another aspect, the present invention provides a method of treatment comprising:
  • (i) diagnosing or detecting colorectal cancer according to the method of the invention; and
  • (ii) administering or recommending a therapeutic for the treatment of colorectal cancer.
  • In yet another aspect, the present invention provides a method for monitoring the efficacy of treatment of colorectal cancer in a subject, the method comprising treating the subject for colorectal cancer and then detecting the presence and/or level of at least two biomarkers selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1β, TGFβ1, and TIMP-1 in a sample from the subject, wherein an absence of and/or reduction in the level of expression of the polypeptides after treatment when compared to before treatment is indicative of effective treatment.
  • In another aspect, the present invention provides an array of at least two compounds for the diagnosis or detection of colorectal cancer, wherein each of the compounds binds a different biomarker polypeptide selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1β, TGFβ1, and TIMP-1.
  • In yet another aspect, the present invention provides a kit for diagnosing or detecting colorectal cancer in a subject, the kit comprising two compounds that each binds a different biomarker polypeptide selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1β, TGFβ1, and TIMP-1.
  • Throughout this specification the word “comprise”, or variations such as “comprises” or “comprising”, will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
  • As will be apparent, preferred features and characteristics of one aspect of the invention are applicable to many other aspects of the invention.
  • The invention is hereinafter described by way of the following non-limiting Examples and with reference to the accompanying figures.
  • BRIEF DESCRIPTION OF THE ACCOMPANYING DRAWINGS
  • FIG. 1. In Study 3 an optimum combination of the 46 potential protein biomarkers was found using logistic regression modelling, resulting in a panel of seven biomarkers and is illustrated as a ROC curve (black curve). The performance of this “panel” on independent data was estimated using “leave one out” cross-validation (grey curve). The vertical lines are drawn at points of 80% and 90% specificity—operating points of interest in screening tests. Performance statistics are given in Table 5.
  • FIG. 2. Performance of a seven biomarker model identifying colorectal cancer patients from normals at each Dukes Stage illustrated by ROC curves for each stage. A (red)—Stage A, B (green)—Stage B, C (blue)—Stage C, and D (black)-Stage D from Study 3a. Performance characteristics are given in Table 6.
  • FIG. 3. When biomarker results from Study 4 (also referred to as Study 3 remeasured) were modelled in pairs a total of 5 pairs (out of a possible 45 combinations selected from the list of 10 biomarkers above) could be shown to produce a sensitivity above 52% at a specificity of 95. The performance of these pair wise biomarker combinations is illustrated as ROC curves (n=5 curves). Performance characteristics are given in Table 7.
  • FIG. 4. An example of a 3 biomarker model generated from Study 4 data which had a sensitivity of at least 50% at 95% specificity. There were 968 possible 3-biomarker combinations and approximately half of those combinations showed a performance of at least 50% sensitivity at 90% specificity.
  • FIG. 5. ROC curves are illustrated for all combinations of 3-10 biomarkers generated from Study 4 data which have a sensitivity of at least 50% at 95% specificity (n=485 cross validated curves out of a possible 968 models).
  • FIG. 6. Frequency of each biomarker in the best 485 models. These BMs represent all serum models that gave a sensitivity of at least 50% at 95% The high representation of all 10 biomarkers in the useful models demonstrates the unity of our selection of these 10 biomarkers.
  • FIG. 7. A 5 biomarker model generated from Study 4 data is illustrated as a ROC curve (black) and cross validated ROC curve (grey). This model shows a sensitivity of 68% at 95% specificity when all stages of disease are included and when cross validated gave a sensitivity of 64%. Biomarkers included are [IL-8, IGFBP2, Mac2BP, DKK-3 and M2PK].
  • FIG. 8. A 6 biomarker model generated from Study 4 data is illustrated as a ROC curve (black) and cross validated ROC curve (Grey). This model shows a sensitivity of 77% at a specificity of 95% when all stages of disease are included and when cross validated gave a sensitivity of 67%. Biomarkers included are [IL-8, IGFBP2, Mac2BP, DKK-3, TGFbeta1&M2PK].
  • FIG. 9. Two alternative seven biomarker models generated from Study 3a data are shown. One was optimised for high specificity (black/new) and an alternative or model optimised for area under the curve is shown (grey/old). At 90% specificity the sensitivity was 72% for the new model and 77% for the older model. Biomarkers included were as follows:
  • New: IL8, IGFBP2, s90MAC2BP, M2PK, DKK-3, IL-13 & TGFbeta,
  • Old: IL8, IGFBP2, s90MAC2BP, M2PK, EpCAM, IL13 & MIP-1b.
  • FIG. 10. A seven biomarker model generated from Study 4 data is illustrated as a ROC curve (black) and cross validated ROC curve (grey). This model shows, a sensitivity of 84% at a specificity of 95%. Biomarkers included are [M2PK serum, IL8.plasma, TGF beta1.serum, IGFBP2.plasma, Mac2BP.serum, TIMP1.plasma and Dkk3 plasma.
  • FIG. 11. Cross validated ROC curves showing the performance of a 3 biomarker model for each Dukes stage is illustrated. This data demonstrates the validity of the choice of three biomarkers (DKK-3, M2PK and IGFBP2) for detecting cancer at different stages of the disease progression. The data indicates that at Stage A if the three markers are used, the test still will achieve a significant sensitivity of 64% at 95% specificity which is comparable to the sensitivity achieved at late stage disease (79%). That is the biomarker panel of three will pick up early disease states allowing early detection. Biomarkers included are Dkk3, M2PK and IGFBP2.
  • KEY TO THE SEQUENCE LISTING
    • SEQ ID NO:1—amino acid sequence of IL-8
    • SEQ ID NO:2—amino acid sequence of IGFBP2
    • SEQ ID NO:3—amino acid sequence of MAC2BP
    • SEQ ID NO:4—amino acid sequence of M2PK variant 1
    • SEQ ID NO:5—amino acid sequence of M2PK variant 2
    • SEQ ID NO:6—amino acid sequence of M2PK variant 3
    • SEQ ID NO:7—amino acid sequence of IL-13
    • SEQ ID NO:8—amino acid sequence of DKK-3 variant 1
    • SEQ ID NO:9—amino acid sequence of DKK-3 variant 2
    • SEQ ID NO:10—amino acid sequence of DKK-3 variant 3
    • SEQ ID NO:11—amino acid sequence of EpCam
    • SEQ ID NO:12—amino acid sequence of MIP1β
    • SEQ ID NO:13—amino acid sequence of TGFβ1
    • SEQ ID NO:14—amino acid sequence of TIMP-1
    DETAILED DESCRIPTION General Techniques and Definitions
  • Unless specifically defined otherwise, all technical and scientific terms used herein shall be taken to have the same meaning as commonly understood by one of ordinary skill in the art (e.g., in cell culture, molecular genetics, immunology, immunohistochemistry, protein chemistry, and biochemistry).
  • Unless otherwise indicated, the recombinant protein, cell culture, and immunological techniques utilized in the present invention are standard procedures, well known to those skilled in the art. Such techniques are described and explained throughout the literature in sources such as, J. Perbal, A Practical Guide to Molecular Cloning, John Wiley and Sons (1984), J. Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd edn, Cold Spring Harbour Laboratory Press (2001), R. Scopes, Protein Purification—Principals and Practice, 3rd edn, Springer (1994), T. A. Brown (editor), Essential Molecular Biology: A Practical Approach, Volumes 1 and 2, IRL Press (1991), D. M. Glover and B. D. Hames (editors), DNA Cloning: A Practical Approach, Volumes 1-4, IRL Press (1995 and 1996), and F. M. Ausubel et al. (editors), Current Protocols in Molecular Biology, Greene Pub. Associates and Wiley-Interscience (1988, including all updates until present), Ed Harlow and David Lane (editors) Antibodies: A Laboratory Manual, Cold Spring Harbour Laboratory, (1988), and J. E. Coligan et al. (editors) Current Protocols in Immunology, John Wiley & Sons (including all updates until present).
  • As used herein, the term “colorectal cancer”, also known as “colon cancer”, “bowel cancer” or “rectal cancer”, refers to all forms of cancer originating from the epithelial cells lining the large intestine and/or rectum.
  • As used herein, “biomarker” refers to any molecule, such as a gene, gene transcript (for example mRNA), peptide or protein or fragment thereof produced by a subject which is useful in differentiating subjects having colorectal cancer from normal or healthy subjects.
  • As used herein, the term “diagnosis”, and variants thereof such as, but not limited to, “diagnose”, “diagnosed” or “diagnosing” shall not be limited to a primary diagnosis of a clinical state, but should be taken to include diagnosis of recurrent disease.
  • As used herein, the term “subject” refers to any animal that may develop colorectal cancer and includes animals such as mammals, e.g. humans, or non-human mammals such as cats and dogs, laboratory animals such as mice, rats, rabbits or guinea pigs, and livestock animals. In a preferred embodiment, the subject is a human.
  • The “sample” may be of any suitable type and may refer, e.g., to a material in which the presence or level of biomarkers can be detected. Preferably, the sample is obtained from the subject so that the detection of the presence and/or level of biomarkers may be performed in vitro. Alternatively, the presence and/or level of biomarkers can be detected in vivo. The sample can be used as obtained directly from the source or following at least one step of (partial) purification. The sample can be prepared in any convenient medium which does not interfere with the method of the invention. Typically, the sample is an aqueous solution, biological fluid, cells or tissue. Preferably, the sample is blood, plasma, serum, urine, platelets, megakaryocytes or faeces. Pre-treatment may involve, for example, preparing plasma from blood, diluting viscous fluids, and the like. Methods of treatment can involve filtration, distillation, separation, concentration, inactivation of interfering components, and the addition of reagents. The selection and pre-treatment of biological samples prior to testing is well known in the art and need not be described further.
  • As used herein the terms “treating”, “treat” or “treatment” include administering a therapeutically effective amount of a compound sufficient to reduce or delay the onset or progression of colorectal cancer, or to reduce or eliminate at least one symptom of colorectal cancer.
  • Biomarkers
  • The present inventors have shown that determining the presence and/or level of least two biomarkers in a sample from a subject allows for the detection or diagnosis of colorectal cancer, either early detection at Dukes Stage A or at some later stage such as Dukes Stage B or C or D, with specificity and sensitivity comparable to or greater than that achieved with the FOBT. The at least two biomarkers that are useful in the methods of the present invention are selected from IL-8 (interleukin-8), IGFBP2 (insulin-like growth factor binding protein-2), MAC2BP (MAC2-binding protein; serum protein 90K), M2PK (pyruvate kinase muscle 2, pyruvate kinase 3), IL-13 (interleukin-13), DKK-3 (dickkopf homolog, 3), EpCAM (epithelial cell adhesion molecule), MIP1β (macrophage inflammatory protein 1β, CCL4, MIP1beta), TGFβ1 (transforming growth factor β1 , TGFbeta1) and TIMP-1 (tissue inhibitor of metalloproteinase 1). Reference to any of these biomarkers includes reference to all polypeptide and polynucleotide variants such as isoforms and transcript variants as would be known by the person skilled in the art. NCBI accession numbers of representative sequences for each of the biomarkers are provided in Table 1.
  • TABLE 1
    NCBI accession numbers for representative biomarker sequences.
    Biomarker Representative NCBI Accession Numbers
    IL-8 NM_000584.2 (SEQ ID NO: 1)
    IGFBP2 NM_000597.2 (SEQ ID NO: 2)
    MAC2BP NM_005567.3 (SEQ ID NO: 3)
    M2PK NM_002654.3; NM_182470.1; NM_182471.1 (SEQ ID
    NOs: 4-6)
    IL-13 NM_002188.2 (SEQ ID NO: 7)
    DKK-3 NM_015881.5; NM_013253; NM_001018057.1 (SEQ ID
    NOs: 8-10)
    EpCam NM_002354.2 (SEQ ID NO: 11)
    MIP1β NM_002984.2 (SEQ ID NO: 12)
    TGFβ1 NM_000660.4 (SEQ ID NO: 13)
    TIMP-1 NM_003254.2 (SEQ ID NO: 14)
  • Detecting or Diagnosing Colorectal Cancer
  • It will be apparent from the preceding description that the diagnostic methods of the present invention may involve a degree of quantification to determine levels biomarkers in patient samples. Such quantification is readily provided by the inclusion of appropriate control samples.
  • In one embodiment, internal controls are included in the methods of the present invention. A preferred internal control is one or more samples taken from one or more healthy individuals.
  • In the present context, the term “healthy individual” shall be taken to mean an individual who is known not to suffer from colorectal cancer, such knowledge being derived from clinical data on the individual, including, but not limited to, a different diagnostic assay to that described herein.
  • As will be known to those skilled in the art, when internal controls are not included in each assay conducted, the control may be derived from an established data set.
  • Data pertaining to the control subjects are preferably selected from the group consisting of:
  • 1. a data set comprising measurements of the presence or level of expression of biomarkers for a typical population of subjects known to have colorectal cancer;
  • 2. a data set comprising measurements of the presence or level of biomarkers for the subject being tested wherein said measurements have been made previously, such as, for example, when the subject was known to be healthy or, in the case of a subject having colorectal cancer, when the subject was diagnosed or at an earlier stage in disease progression;
  • 3. a data set comprising measurements of the presence or level of biomarkers for a healthy individual or a population of healthy individuals; and
  • 4. a data set comprising measurements of the presence or level of biomarkers for a normal individual or a population of normal individuals.
  • In the present context, the term “typical population” with respect to subjects known to have colorectal cancer shall be taken to refer to a population or sample of subjects diagnosed with colorectal cancer that is representative of the spectrum of colorectal cancer patients. This is not to be taken as requiring a strict normal distribution of morphological or clinicopathological parameters in the population, since some variation in such a distribution is permissible. Preferably, a “typical population” will exhibit a spectrum of colorectal cancer at different stages of disease progression. It is particularly preferred that a “typical population” exhibits the expression characteristics of a cohort of subjects as described herein.
  • The term “normal individual” shall be taken to mean an individual that does not express a biomarker, or expresses a biomarker at a low level in a sample. As will be known to those skilled in the art, data obtained from a sufficiently large sample of the population will normalize, allowing the generation of a data set for determining the average level of a particular biomarker.
  • Those skilled in the art are readily capable of determining the baseline for comparison in any diagnostic assay of the present invention without undue experimentation, based upon the teaching provided herein.
  • Compounds that bind a biomarker when used diagnostically may be linked to a diagnostic reagent such as a detectable label to allow easy detection of binding events in vitro or in vivo. Suitable labels include radioisotopes, dye markers or other imaging reagents for detection and/or localisation of target molecules. Compounds linked to a detectable label can be used with suitable in vivo imaging technologies such as, for example, radiology, fluoroscopy, nuclear magnetic resonance imaging (MRI), CAT-scanning, positron emission tomography (PET), computerized tomography etc.
  • The diagnostic methods of the present invention are able to diagnose or detect colorectal cancer with a sensitivity and specificity that is at least comparable to FOBT, or greater. As would be understood by the person skilled in the art, sensitivity refers to the proportion of actual positives in the diagnostic test which are correctly identified as having colorectal cancer. Specificity measures the proportion of negatives which are correctly identified as not having colorectal cancer. In one embodiment, the methods of the invention are able to diagnose or detect colorectal cancer with a sensitivity of at least 50%, 60% or 66%, or at least 77%, 80%, 83%, 85%, 86%, 87%, 88%, 89%, 90%, or at least 93%. In another embodiment, the methods of the invention are able to diagnose or detect colorectal cancer with a sensitivity of at least 80%, or at least 85% or at least 90%, or at least 95%.
  • In one embodiment, the methods of the invention are able to diagnose or detect colorectal cancer with a specificity of at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94% or at least 95%.
  • Advantageously, the methods of the present invention are able to detect colorectal cancer at all of the Dukes Stages with greater sensitivity than the FOBT. In Dukes Stage A, the tumor has penetrated into, but not through, the bowel wall. In Dukes Stage B, the tumor has penetrated through the bowel wall but there is not yet any lymph node involvement. In Dukes Stage C, the cancer involves regional lymph nodes. In Dukes Stage D, there is distant metastasis, for example, to the liver or lung. In one embodiment, the methods of the present invention are able to diagnose or detect colorectal cancer at any Dukes Stage with a sensitivity of at least 80%.
  • As known to the skilled person, there are other systems for staging cancer that are know in the art. One example is the TMN Classification of Malignant Tumors (TNM) that is used by the American Joint Committee on Cancer (AJCC: Colon and rectum. in Edge et al., eds; AJCC Cancer Staging Manual, 7th ed. New York, N.Y.: Springer, 2010, pp: 143-164). Another example is the Modified Astler-Coller classification (MAC).
  • Accordingly, the skilled person will appreciate that the Dukes Stages correspond to certain TNM Classifications. For example, Dukes Stage A corresponds to T1, T2, N0 and M0; Dukes Stage B corresponds to T3, T4a, T4b, N0 and M0; and Dukes Stage C corresponds to i) T1-T2, N1/N1c, M0; ii) T1, N2a and M0; iii) T3-T4a, N1/N1c and M0; iv) T2-T3, N2a and M0; v) T1-T2, N2b and M0; vi) T4a, N2a and M0; vii) T3-T4a, N2b and M0; and viii) T4b, N1-N2 and M0. Thus, the skilled person will understand that reference to a Dukes Stage as used herein includes reference to the corresponding TMN classification as known in the art.
  • Protein Detection Techniques
  • In one embodiment, biomarker polypeptide is detected in a patient sample, wherein the presence and/or level of the polypeptide in the sample is indicative of colorectal cancer. For example, the method may comprise contacting a biological sample derived from the subject with a compound capable of binding to a biomarker polypeptide, and detecting the formation of complex between the compound and the biomarker polypeptide. The term “biomarker polypeptide” as used herein includes fragments of biomarker polypeptides, including for example, immunogenic fragments and epitopes of the biomarker polypeptide.
  • In one embodiment, the compound that binds the biomarker is an antibody.
  • The term “antibody” as used herein includes intact molecules as well as molecules comprising or consisting of fragments thereof, such as, for example Fab, F(ab′)2, Fv and scFv, as well as engineered variants including diabodies, triabodies, mini-bodies and single-domain antibodies which are capable of binding an epitopic determinant. Thus, antibodies may exist as intact immunoglobulins, or as modifications in a variety of forms.
  • In another embodiment, an antibody to a biomarker polypeptide is detected in a patient sample, wherein the presence and/or level of the antibody in the sample is indicative of colorectal cancer.
  • Preferred detection systems contemplated herein include any known assay for detecting proteins or antibodies in a biological sample isolated from a human subject, such as, for example, SDS/PAGE, isoelectric focussing, 2-dimensional gel electrophoresis comprising SDS/PAGE and isoelectric focussing, an immunoassay, flow cytometry e.g. fluorescence-activated cell sorting (FACS), a detection based system using an antibody or non-antibody compound, such as, for example, a small molecule (e.g. a chemical compound, agonist, antagonist, allosteric modulator, competitive inhibitor, or non-competitive inhibitor, of the protein). In accordance with these embodiments, the antibody or small molecule may be used in any standard solid phase or solution phase assay format amenable to the detection of proteins. Optical or fluorescent detection, such as, for example, using mass spectrometry, MALDI-TOF, biosensor technology, evanescent fiber optics, or fluorescence resonance energy transfer, is clearly encompassed by the present invention. Assay systems suitable for use in high throughput screening of mass samples, e.g. a high throughput spectroscopy resonance method (e.g. MALDI-TOF, electrospray MS or nano-electrospray MS), are also contemplated. Another suitable protein detection technique involves the use of Multiple Reaction Monitoring (MRM) in LC-MS (LC/MRM-MS) (Anderson and Hunter, 2006).
  • Immunoassay formats are particularly suitable, e.g., selected from the group consisting of, an immunoblot, a Western blot, a dot blot, an enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), enzyme immunoassay. Modified immunoassays utilizing fluorescence resonance energy transfer (FRET), isotope-coded affinity tags (ICAT), matrix-assisted laser desorption/ionization time of flight (MALDI-TOF), electrospray ionization (ESI), biosensor technology, evanescent fiber-optics technology or protein chip technology are also useful.
  • Nucleic Acid Detection Techniques
  • Any suitable technique that allows for the qualitative and/or quantitative assessment of the level of a biomarker polynucleotide in a sample may be used. The terms “nucleic acid molecule” or “polynucleotide” as used herein refer to an oligonucleotide, polynucleotide or any fragment thereof.
  • Comparison may be made by reference to a standard control, or to a control level that is found in healthy tissue. For example, levels of a transcribed gene can be determined by Northern blotting, and/or RT-PCR. With the advent of quantitative (real-time) PCR, quantitative analysis of gene expression can be achieved by using appropriate primers for the gene of interest. The nucleic acid may be labelled and hybridised on a gene array, in which case the gene concentration will be directly proportional to the intensity of the radioactive or fluorescent signal generated in the array.
  • Methods for direct sequencing of nucleotide sequences are well known to those skilled in the art and can be found for example in Ausubel et al., eds., Short Protocols in Molecular Biology, 3rd ed., Wiley, (1995) and Sambrook et al., Molecular Cloning, 3rd ed., Cold Spring Harbor Laboratory Press, (2001). Sequencing can be carried out by any suitable method, for example, dideoxy sequencing, chemical sequencing or variations thereof. Direct sequencing has the advantage of determining variation in any base pair of a particular sequence.
  • Other PCR methods that may be used in carrying out the invention include hybridization based PCR detection systems, TaqMan assay (U.S. Pat. No. 5,962,233) and the molecular beacon assay (U.S. Pat. No. 5,925,517).
  • The nucleic acid may be separated from the sample for testing. Suitable methods will be known to those of skill in the art. For example, RNA may be isolated from a sample to be analysed using conventional procedures, such as are supplied by QIAGEN technology. This RNA is then reverse-transcribed into DNA using reverse transcriptase and the DNA molecule of interest may then be amplified by PCR techniques using specific primers.
  • Diagnostic procedures may also be performed directly upon patient samples. Hybridisation or amplification assays, such as, for example, Southern or Northern blot analysis, immunohistochemistry, single-stranded conformational polymorphism analysis (SSCP) and PCR analyses are among techniques that are useful in this respect. If desired, target or probe nucleic acid may be immobilised to a solid support such as a microtitre plate, membrane, polystyrene bead, glass slide or other solid phase.
  • Kits
  • The present invention provides kits for the diagnosis or detection of colorectal cancer. Such kits may be suitable for detection of nucleic acid species, or alternatively may be for detection of a polypeptide gene product, as discussed above.
  • For detection of polypeptides, antibodies will most typically be used as components of kits. However, any agent capable of binding specifically to a biomarker gene product will be useful in this aspect of the invention. Other components of the kits will typically include labels, secondary antibodies, substrates (if the gene is an enzyme), inhibitors, co-factors and control gene product preparations to allow the user to quantitate expression levels and/or to assess whether the diagnosis experiment has worked correctly. Enzyme-linked immunosorbent assay-based (ELISA) tests and competitive ELISA tests are particularly suitable assays that can be carried out easily by the skilled person using kit components.
  • Optionally, the kit further comprises means for the detection of the binding of an antibody to a biomarker polypeptide. Such means include a reporter molecule such as, for example, an enzyme (such as horseradish peroxidase or alkaline phosphatase), a dye, a radionucleotide, a luminescent group, a fluorescent group, biotin or a colloidal particle, such as colloidal gold or selenium. Preferably such a reporter molecule is directly linked to the antibody.
  • In yet another embodiment, a kit may additionally comprise a reference sample. In one embodiment, a reference sample comprises a polypeptide that is detected by an antibody. Preferably, the polypeptide is of known concentration. Such a polypeptide is of particular use as a standard. Accordingly, various known concentrations of such a polypeptide may be detected using a diagnostic assay described herein.
  • For detection of nucleic acids, such kits may contain a first container such as a vial or plastic tube or a microtiter plate that contains an oligonucleotide probe. The kits may optionally contain a second container that holds primers. The probe may be hybridisable to DNA whose altered expression is associated with colorectal cancer and the primers are useful for amplifying this DNA. Kits that contain an oligonucleotide probe immobilised on a solid support could also be developed, for example, using arrays (see supplement of issue 21(1) Nature Genetics, 1999).
  • For PCR amplification of nucleic acid, nucleic acid primers may be included in the kit that are complementary to at least a portion of a biomarker gene as described herein. The set of primers typically includes at least two oligonucleotides, preferably four oligonucleotides, that are capable of specific amplification of DNA. Fluorescent-labelled oligonucleotides that will allow quantitative PCR determination may be included (e.g. TaqMan chemistry, Molecular Beacons). Suitable enzymes for amplification of the DNA, will also be included.
  • Control nucleic acid may be included for purposes of comparison or validation. Such controls could either be RNA/DNA isolated from healthy tissue, or from healthy individuals, or housekeeping genes such as β-actin or GAPDH whose mRNA levels are not affected by colorectal cancer.
  • Regression Algorithms and Statistics
  • In order to develop a panel of biomarkers suitable for diagnosing or detecting colorectal cancer, the present inventors have analysed numerous biomarkers in a statistical model. Such an improvement in the performance of a test is sometimes referred to as the “in-sample” performance. A fair evaluation of a test requires its assessment using out-of-sample subjects, that is, subjects not included in the construction of the initial predictive model. This is achieved by assessing the test performance using cross validation.
  • Tests for statistical significance include linear and non linear regression, including ANOVA, Kruskal-Wallis, Wilcoxon, Mann-Whitney and odds ratio, Baysian probability algorithms. As the number of biomarkers measured increases however, it can be generally more convenient to use a more sophisticated technique such as Random Forests, simple logistic, Bayes Net to name a few.
  • For example, Bayesian probability may be adopted. In this circumstance a 10-fold cross-validation can be used to estimate the “out-of-sample” performance of the models in question. For each combination of biomarkers under consideration, the data can be divided randomly into 10 sub-samples, each with similar proportions of healthy subject and subjects at each stage of disease. In turn, each subsample can be excluded, and a logistic model built using the remaining 90% of the subjects. This model can then be used to estimate the probability of cancer for the excluded sub-sample, providing an estimate of “out-of-sample” performance. By repeating this for the remaining 9 subsamples, “out-of-sample” performance can be estimated from the study data itself. These out-of sample predicted probabilities can then be compared with the actual disease status of the subjects to create a Receiver Operating Characteristic (ROC) Curve, from which the cross-validated sensitivity at 95% specificity may be estimated.
  • Each estimate of “out-of-sample” performance using cross-validation (or any other method), whilst unbiased, has an element of variability to it. Hence a ranking of models (based on biomarker combinations) can be indicative only of the relative performance of such models. However a set of biomarkers which is capable of being used in a large number of combinations to generate a diagnostic test as demonstrated via “out-of-sample” performance evaluations, almost certainly contains within itself combinations of biomarkers that will withstand repeated evaluation.
  • Many different combinations can qualify as diagnostic tests which prove useful and cost effective and have acceptable sensitivity for a given specificity. As an example, consider the five biomarkers: IL-8, IGFBP2, MAC2BP, M2PK and DKK-3. A model discriminating subjects with cancer from healthy controls can be as follows:
  • log ( p 1 - p ) = β 0 + β IL 8 C IL 8 + β IGFBP 2 C IGFBP 2 + β MAC 2 BP C MAC 2 BP + β M 2 PK C M 2 PK + β DKK 3 C DKK 3
  • Here p represents the probability that a person has colorectal cancer. Each Ci is the logarithm of concentration biomarker i in the plasma (or serum) of a person. Each beta (β) is a coefficient applying to that biomarker in the concentration units in which it is measured—β0 is an “offset” or “intercept”. This linear logistic model is common to all results presented herein, but is far from the only way in which a combination of biomarker concentrations may be modelled to predict the probability of cancer.
  • Other non linear or linear logistic algorithms that would be equally applicable include Random Forest, ANOVA, t-Test, Fisher analysis, Support Vector Machine, Linear Models for MicroArray data (LIMMA) and/or Significance Analyses of Microarray Data (SAM), Best First, Greedy Stepwise, Naive Bayes, Linear Forward Selection, Scatter Search, Linear Discriminant Analysis (LDA), Stepwise Logistic Regression, Receiver Operating Characteristic and Classification Trees (CT).
  • Thus, in light of the teachings of the present specification, the person skilled in the art will appreciate that the sensitivity and specificity of a test for diagnosing colorectal cancer may be modulated by selecting a different combination of the biomarkers as described herein
  • Knowledge-Based Systems
  • It will be apparent from the discussion herein that knowledge-based computer software and hardware for implementing an algorithm also form part of the present invention. Such computer software and/or hardware are useful for performing a method of diagnosing or detecting colorectal cancer according the invention. Thus, the present invention also provides software or hardware programmed to implement an algorithm that processes data obtained by performing the method of the invention via a multivariate analysis to provide a disease score and provide or permit a diagnosis or detection of colorectal cancer and/or determine progression or status of a colorectal cancer or determine whether or not a colorectal cancer has progressed or determine whether or not a subject is responding to treatment for colorectal cancer in accordance with the results of the disease score in comparison with predetermined values.
  • In one example, a method of the invention may be used in existing knowledge-based architecture or platforms associated with pathology services. For example, results from a method described herein are transmitted via a communications network (e.g. the internet) to a processing system in which an algorithm is stored and used to generate a predicted posterior probability value which translates to the score of disease probability or risk of recurrence or metastasis or responsiveness to treatment which is then forwarded to an end user in the form of a diagnostic or predictive report.
  • The method of the invention may, therefore, be in the form of a kit or computer-based system which comprises the reagents necessary to detect the concentration of the biomarkers and the computer hardware and/or software to facilitate determination and transmission of reports to a clinician.
  • The assay of the present invention permits integration into existing or newly developed pathology architecture or platform systems. For example, the present invention contemplates a method of allowing a user to determine the status of a subject with respect to colorectal cancer, the method including:
  • (a) receiving data in the form of levels at least two biomarkers selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1β, TGFβ1, and TIMP-1 in a readily obtained sample, optionally in combination with another marker of colorectal cancer;
    (b) processing the subject data via multivariate analysis (for example, regression analysis) to provide a disease score;
    (c) determining the status of the subject in accordance with the results of the disease score in comparison with predetermined values; and
    (d) transferring an indication of the status of the subject to the user via the communications network reference to the multivariate analysis includes an algorithm which performs the multivariate analysis function.
  • In one embodiment, the method for diagnosing or detecting colorectal cancer of the invention may be performed by taking a blood sample from a patient and determining the presence and/or level of any one or more of the biomarkers as described herein. If desired, the measurements may be made, for example, on a biochip so that a single analysis can be used to measure the presence and/or level of multiple biomarkers. The results of this analysis may then be inputted into into a computer program that subjects them to linear regression analysis. The computer could also contain information as to control values or expected ranges, or the clinician, nurse, medical administrator or general practitioner could input such data. This analysis wold then provide a score or likelihood of having colorectal cancer. If a second test for the patient is performed, the regression analysis may indicate a change in the score, thus indicating that the patient's disease has progressed or worsened.
  • EXAMPLES Materials and Methods Patient Samples
  • A collection of plasma and serum samples was taken and processed from a cohort of colorectal cancer patients (Dukes Stages A-D) that were being treated at several hospitals.
  • Blood was also collected and processed from a group of about 50 healthy volunteers over the age of 65 and from a group of 15 over the age of 50.
  • Four separate studies were undertaken with slightly different biomarkers. Study 1 looked at 52 cancer samples and 50 controls, study 2 looked at 55 cancer samples and 53 controls, study 3 and 4 looked at 96 cancer samples and 50 controls. In study 2, 3 and 4 the patients were age and gender matched across Dukes Stages, see Table 2 for summary statistics.
  • TABLE 2
    Characteristics of normal volunteers and colorectal cancer
    patients used in studies 2, 3 and 4.
    Controls Cancers
    n = 50 n = 96
    Gender
    male 25 48
    female 25 48
    Mean Age (yr) 68 68
    Dukes stage
    A 22
    B 30
    C 30
    D 14
    Tumour site
    colon
    73
    rectum 17
    unknown 6
    Proximal (includes caecum, ascending, hepatic 43
    flexure and transverse colon)
    Distal (includes splenic flexure, descending, 47
    sigmoid and rectum)
  • Biomarker Analysis
  • Analysis of biomarkers was done with commercial kits and sourced antibodies (DSL, R&D Duoset, Calbiotech, Millipore, Abnova, Genway, Peviva, Schebo, Bender) and using ELISA or Luminex assays.
  • Statistical Evaluation and Panel Biomarker Modelling
  • Results for each assay were analysed using the statistical software packages Prism and “R”. Individual performance of markers was evaluated using the non-parametric Mann-Whitney t-test and individual receiver operator characteristic (ROC) curves were generated.
  • Logistic regression and related modelling strategies were used to find combinations of biomarkers that best separated controls and colorectal cancer patients. Four separate studies were performed with the same samples/aliquots. The results of each of these is given below.
  • Results of Study 1, 2 and 3
  • Biomarkers chosen to be measured in Study 1 and 2 and 3 are listed in Table 3. Biomarkers in bold were those identified as promising from each study (i.e. they were significantly different in samples from colorectal cancer patients versus control and/or they were identified in panels of combined biomarkers that distinguish colorectal cancer from controls).
  • TABLE 3
    Biomarkers analysed in studies.
    Study 1 Study 2 Study 3
    IGF-I IGF-BP2 IGF-BP2 (DSL)
    IGF-II IGF-II IGF-II
    IGF-BP2 IGF-BP3 IFNg
    IGF-BP3 Her2 TNFa
    BTC VegFA IL-10
    Amphiregulin VegFC IL-6
    VegFA VegFD GM-CSF
    VegFC TIMP-1 IL-12
    VegFD TIMP-2 IL-13
    MMP-2 MMP-1 IL-8
    MMP-7 MMP-2 IL-4
    MMP-9 MMP-3 Il-2
    TIMP-2 MMP-7 IL-1b
    Her2 MMP-8 MMP-1
    MMP-12 MMP-2
    MMP-13 MMP-3
    ENA-78 MMP-7
    MCP-1 MMP-8
    MIP-1beta MMP-9
    GM-CSF ENA-78
    IFN-gamma MIP-1alpha
    IL-10 MIP-1beta
    IL-12 MCP-1
    IL-13 Mac-2BP
    IL-1beta TIMP-1
    IL-2 TIMP-2
    IL-4 Gro-alpha
    IL-6 Tumour M2 pyruvate
    kinase
    IL-8 M30-apoptosense
    TNF-alpha M65
    Cripto Trail-R2
    P-cadherin
    OPN
    Dkk-3
    EpCam
    TGFbeta1
    REG IV
    CEA
    DcR3
    CA19.9
    Amphiregulin
    CEACAM6
    VegFA pan
    VegFA165b
    Spondin-2
    survivin
  • Statistical Evaluation and Panel Biomarker Modelling
  • To find combinations of biomarkers that best separated controls and colorectal cancer patients, forward variable selection with Bayesian Information Criteria to penalize log-likelihood to prevent over-fitting was adopted. To estimate the likely performance of the panel of biomarkers on an independent dataset, “N-Fold” or “leave-one-out” cross validation was used. In this procedure one observation at a time was excluded whilst the entire model fitting algorithm was applied to the remaining observations.
  • The resulting model was then used to estimate the probability that the excluded observation is a case. This was repeated for each observation in the dataset. In this way each observation in turn acted as an independent test of the model-building algorithm. The resulting dataset consisting of cases and controls each with an “independently predicted” case probability can then be compared with the original model. The ability to choose from numerous biomarkers and weight them appropriately allows a search strategy which optimises performance in regions of interest on the ROC curve. The cost of poor specificity is large numbers of unnecessary colonoscopies.
  • In study 3, 48 potential biomarkers were evaluated to select a candidate panel of colorectal cancer biomarkers, using block randomization within plates to avoid bias. From this list of 48 only 42 showed measurable levels. Individually 14 biomarkers showed significant difference between controls and CRC as assessed by t-tests; (IGFII, IGFBP2, IL-8, IL-6, MMP-1, MMP-7, s90/Mac2BP, M2PK, EpCam, TIMP-1 (serum and plasma), M65, OPN, TGFβ1, VEGFpan. As expected, none had sufficient sensitivity or specificity to be useful as a biomarker by itself (not shown). However, using a variety of modelling strategies, including use of logarithmic values, several different panels of biomarkers were found that exceeded the performance of FOBT especially for early to late stage disease.
  • FIG. 1 shows the results from a 7 biomarker panel which included IL8 (serum), IL-13 (serum), EpCAM (plasma), M2PK (plasma), IGFBP2 (serum) and Mac2BP (serum) and which was cross validated to predict its performance on independent samples.
  • This 7 biomarker model, which is described at least conceptually as
  • log ( p 1 - p ) = β 0 + β IL 8 C IL 8 + β IGFBP 2 C IGFBP 2 + β MAC 2 BP C MAC 2 BP + β M 2 PK C M 2 PK + β DKK 3 C DKK 3
  • provided good performance at high specificity and was robust under cross validation. The coefficients estimated to give the best model for this biomarker combination in plasma are listed in Table 4. Performance statistics are provided in Table 5. This performance exceeds that quoted for FOBT (sensitivity 65.8%, specificity 95%) (Morikawa et al., 2005).
  • TABLE 4
    Coefficients for the biomarker combination.
    Measured Concentration
    Biomarker in Units Coefficient
    Intercept NA NA −37.74
    IL-8 serum pg/mL 1.07
    IL-13 serum pg/mL −0.28
    EpCAM plasma pg/mL −0.33
    M2PK plasma units/mL 1.40
    IGFBP2 serum ng/mL 1.99
    Mac2BP serum ng/mL 2.39
    MIP1beta Serum Pg/ml −1.19
  • TABLE 5
    Performance of the 7 biomarker model and cross-validation.
    Model
    estimate Cross validation
    Area Under the ROC Curve (AUC) 0.91 0.86
    Sensitivity at 80% specificity 0.84 0.78
    Sensitivity at 90% specificity 0.81 0.69
  • This model was also applied separately to patients from each stage of colorectal cancer (Dukes Stage A, B, C, D) and shown to perform equally well within each stage (FIG. 2). The AUCs were 0.88-0.93 and were almost equally good at discriminating all Stages of colorectal cancer. The model shows the highest sensitivity of 90% at 90% specificity for Stage C and the lowest sensitivity of 73% at 90% specificity for Stage B (Table 6).
  • TABLE 6
    Performance of the Model by Dukes Stage
    Stage A Stage B Stage C Stage D
    Area Under the ROC Curve 0.89 0.88 0.93 0.91
    (AUC)
    Sensitivity at 80% specificity 0.82 0.77 0.90 0.93
    Sensitivity at 90% specificity 0.77 0.73 0.90 0.86
  • Study 4 (Also Referred to as “Study 3 Remeasured”)
  • In study 4, 10 biomarkers were remeasured in the same cohort as Study 3. Blood was collected from 96 colorectal cancer patients and 50 normal subjects (the controls). In this study the focus was on 10 biomarkers, namely IGFBP2, IL8, IL13, Mac2BP, M2PK, Dkk3, EpCam, TGFbeta1, TIMP-1, MIP1beta. Assays were performed as described previously. Both serum and plasma levels of each of the biomarkers were measured and compared with control values.
  • When modelled in pairs (two markers), a total of 5 pairs (out of a possible 45 combinations selected from the list of 10 biomarkers above) could be shown to produce a sensitivity above 52% at a specificity of 95%. See Table 7 and FIG. 3.
  • TABLE 7
    Biomarker Pairs Producing Useful Screening Tests
    on Cross-Validation.
    Estimated Estimated
    In-Sample Out-of-Sample
    (Test) (Cross-Validated)
    Sensitivity at 95% Sensitivity at 95%
    Biomarker
    1 Biomarker 2 Specificity Specificity
    M2PK EpCAM 58.3% 58.3%
    M2PK IL13 56.3% 57.3%
    Dkk3 M2PK 55.2% 55.2%
    M2PK IL8 60.4% 54.2%
    M2PK IGFBP2 58.3% 52.1%
  • In analysing combinations of three to ten of the nominated biomarkers, there are 968 possible combinations. The 968 combinations of between 3 and 10 biomarkers consist of the 120 combinations of 3 marker; 210 combinations of 4 markers; 252 combinations of 5 markers; 210 combinations of 6 markers; 120 combinations of 7 markers; 45 combinations of 8 markers; 10 combinations of 9 markers and the single combination that includes all 10 biomarkers. When they were modelled using a linear logistic model, and then tested via 10-fold cross validation, about half of the 968 combinations had a sensitivity of 50% at a specificity of 95%, see FIG. 4 which shows the results for a three biomarker combination. More than half of these combinations would have a specificity of 90% and a sensitivity of 50%.
  • FIG. 5 shows all 485 of the estimated out-of sample (10-fold cross-validated) ROC curves for tests out of a total possible 968 models based on all possible combinations of 3 to 10 of the biomarkers. Note that many individual segments of the 485 ROC curves are coincident, due to as each horizontal segment represents one control and each vertical segment one case. In this instance 50.1% of the combinations have exceeded the 50% sensitivity, 95% specificity, The best estimated “out-of-sample” performance is a sensitivity of 76% at 95% specificity. Repeating the cross-validation will select a different set of models—the sensitivity of any one combination may vary by 10% at 95% specificity due to random sampling—but result in a similar proportion of useful “useful screening tests”. Precise validation of individual models requires repeated experiments and larger sample sizes.
  • FIG. 6 shows how many of the 485 combinations with 50% sensitivity, 95% specificity, include any given biomarker. At the high end, 432 of the chosen “useful” combinations include M2PK. At the low end 227 of the chosen “useful” combinations include MIP1beta. This high representation of all 10 biomarkers in “useful” models shows the unity and self-complementarity of the selection of these 10 biomarkers.
  • FIG. 7 to FIG. 11 demonstrate some of the results from this last study (Study 4) for combinations of 5 and 7 biomarkers, including a model where the samples are either from plasma or serum cluster. FIG. 11 demonstrates the validity of the choice of three biomarkers (DKK-3, M2PK and IGFBP2) at different stages of the disease progression. The data indicates that at Stage A if the three markers are used, the test still will achieve a significant sensitivity (64%) at 95% specificity which is comparable to the sensitivity achieved at late stage disease, 79%). That is the biomarker panel of three will pick up early disease states allowing early detection.
  • Tables 8 to 16 list results from various combinations of various biomarker panel sets. Depending on the linear regression that is used, as well as the cohort control and other factors such as sample derivation and assay kit technique, there may be a variation on the actual figures or order of the markers. Regardless, many of these combinations will achieve good selectivity at high specificities so as to be useful for diagnosing or detecting colorectal cancer at any stage of the disease progression.
  • 10
  • TABLE 8
    Combination of three biomarkers in serum that equal or exceed
    50% sensitivity at 95% specificity.
    Cross
    Test Validated
    Sensitivity Sensitivity
    at 95% at 95%
    BM1 BM2 BM3 Specificity Specificity
    Dkk3 M2PK IGFBP2 72.9% 70.8%
    Dkk3 M2PK IL8 62.5% 61.5%
    M2PK IL13 IGFBP2 65.6% 61.5%
    M2PK IGFBP2 EpCAM 63.5% 61.5%
    M2PK IL8 IGFBP2 65.6% 60.4%
    M2PK IL8 IL13 61.5% 58.3%
    M2PK MIP1beta IL13 55.2% 57.3%
    M2PK IL8 Mac2BP 59.4% 57.3%
    M2PK MIP1beta TGFbeta1 57.3% 56.3%
    M2PK IL8 EpCAM 64.6% 56.3%
    Dkk3 M2PK IL13 59.4% 55.2%
    Dkk3 M2PK EpCAM 56.3% 55.2%
    M2PK MIP1beta EpCAM 59.4% 55.2%
    M2PK IL8 TGFbeta1 58.3% 55.2%
    M2PK IL8 TIMP1 57.3% 55.2%
    TGFbeta1 Mac2BP TIMP1 54.2% 55.2%
    M2PK Mac2BP IGFBP2 58.3% 54.2%
    Dkk3 IL8 Mac2BP 55.2% 53.1%
    M2PK MIP1beta Mac2BP 52.1% 53.1%
    M2PK MIP1beta IGFBP2 57.3% 53.1%
    M2PK TIMP1 EpCAM 58.3% 53.1%
    M2PK MIP1beta IL8 56.3% 52.1%
    M2PK IL13 TIMP1 58.3% 52.1%
    Dkk3 M2PK Mac2BP 57.3% 51.0%
    Dkk3 M2PK TIMP1 55.2% 51.0%
    M2PK IL13 Mac2BP 58.3% 51.0%
    M2PK TGFbeta1 IGFBP2 52.1% 51.0%
    M2PK TGFbeta1 TIMP1 57.3% 51.0%
    M2PK TGFbeta1 Mac2BP 56.3% 50.0%
    IL8 IL13 Mac2BP 61.5% 50.0%
    IL8 TGFbeta1 Mac2BP 53.1% 50.0%
    M2PK MIP1beta TIMP1 49.0% 49.0%
    M2PK TGFbeta1 EpCAM 49.0% 47.9%
    IL8 IL13 IGFBP2 49.0% 47.9%
    IL8 Mac2BP IGFBP2 57.3% 47.9%
    Dkk3 M2PK MIP1beta 55.2% 46.9%
    TGFbeta1 Mac2BP IGFBP2 46.9% 46.9%
    Dkk3 Mac2BP IGFBP2 49.0% 45.8%
    M2PK IL13 EpCAM 50.0% 45.8%
    IL8 Mac2BP TIMP1 52.1% 45.8%
    IL13 Mac2BP IGFBP2 45.8% 44.8%
    Dkk3 IL8 TGFbeta1 41.7% 43.8%
    MIP1beta IL8 Mac2BP 42.7% 43.8%
    MIP1beta IL8 EpCAM 44.8% 43.8%
    IL8 TGFbeta1 EpCAM 46.9% 43.8%
    Dkk3 IL8 EpCAM 51.0% 42.7%
    IL8 IGFBP2 EpCAM 43.8% 42.7%
    M2PK Mac2BP TIMP1 51.0% 41.7%
  • TABLE 9
    Combination of four biomarkers including DKK-3 in serum that
    equal or exceed 50% sensitivity at 95% specificity.
    Cross
    Test Validated
    Sensitivity Sensitivity
    at 95% at 95%
    BM1 BM2 BM3 BM4 Specificity Specificity
    Dkk3 M2PK Mac2BP IGFBP2 68.8% 69.8%
    Dkk3 M2PK IL8 IL13 71.9% 68.8%
    Dkk3 M2PK IL8 EpCAM 70.8% 67.7%
    Dkk3 M2PK TGFbeta1 Mac2BP 67.7% 65.6%
    Dkk3 M2PK IL8 IGFBP2 69.8% 64.6%
    Dkk3 M2PK IL8 Mac2BP 69.8% 63.5%
    Dkk3 M2PK MIP1beta TGFbeta1 65.6% 61.5%
    Dkk3 M2PK IL8 TIMP1 68.8% 61.5%
    Dkk3 M2PK IL13 IGFBP2 63.5% 61.5%
    Dkk3 M2PK MIP1beta IL8 59.4% 60.4%
    Dkk3 M2PK IGFBP2 EpCAM 68.8% 60.4%
    Dkk3 M2PK MIP1beta IGFBP2 69.8% 59.4%
    Dkk3 M2PK TGFbeta1 IGFBP2 61.5% 59.4%
    Dkk3 M2PK IL13 Mac2BP 56.3% 58.3%
    Dkk3 M2PK TGFbeta1 TIMP1 65.6% 58.3%
    Dkk3 M2PK MIP1beta IL13 57.3% 57.3%
    Dkk3 IL8 Mac2BP IGFBP2 62.5% 56.3%
    Dkk3 M2PK IL8 TGFbeta1 65.6% 55.2%
    Dkk3 M2PK IL13 TIMP1 57.3% 55.2%
    Dkk3 M2PK Mac2BP TIMP1 57.3% 55.2%
    Dkk3 M2PK MIP1beta Mac2BP 57.3% 54.2%
    Dkk3 IL8 IL13 Mac2BP 61.5% 54.2%
    Dkk3 M2PK TGFbeta1 EpCAM 61.5% 53.1%
    Dkk3 M2PK IGFBP2 TIMP1 61.5% 53.1%
    Dkk3 M2PK IL13 TGFbeta1 63.5% 52.1%
    Dkk3 M2PK TIMP1 EpCAM 56.3% 52.1%
    Dkk3 IL8 Mac2BP TIMP1 57.3% 52.1%
    Dkk3 M2PK MIP1beta EpCAM 60.4% 51.0%
    Dkk3 TGFbeta1 Mac2BP TIMP1 59.4% 51.0%
  • TABLE 10
    Combination of four biomarkers including M2PK in serum that
    equal or exceed 50% sensitivity at 95% specificity.
    Cross
    Test Validated
    Sensitivity Sensitivity
    at 95% at 95%
    BM1 BM2 BM3 BM4 Specificity Specificity
    M2PK IL8 Mac2BP TIMP1 63.5% 65.6%
    M2PK Mac2BP IGFBP2 EpCAM 70.8% 65.6%
    M2PK IL8 IL13 Mac2BP 66.7% 64.6%
    M2PK IL8 TGFbeta1 Mac2BP 65.6% 64.6%
    M2PK MIP1beta IL13 IGFBP2 64.6% 62.5%
    M2PK IL8 IL13 TIMP1 64.6% 62.5%
    M2PK IL8 IL13 EpCAM 65.6% 62.5%
    M2PK IL13 Mac2BP IGFBP2 69.8% 62.5%
    M2PK MIP1beta IL8 IL13 63.5% 61.5%
    M2PK IL8 Mac2BP EpCAM 65.6% 61.5%
    M2PK IL13 IGFBP2 EpCAM 69.8% 61.5%
    M2PK MIP1beta IL8 TGFbeta1 58.3% 58.3%
    M2PK IL8 IL13 IGFBP2 67.7% 58.3%
    M2PK IL8 Mac2BP IGFBP2 61.5% 58.3%
    M2PK IL8 IGFBP2 EpCAM 64.6% 58.3%
    M2PK IL13 TGFbeta1 IGFBP2 64.6% 58.3%
    M2PK IL13 TGFbeta1 EpCAM 62.5% 58.3%
    M2PK IL13 IGFBP2 TIMP1 62.5% 58.3%
    M2PK TGFbeta1 Mac2BP TIMP1 62.5% 58.3%
    M2PK TGFbeta1 IGFBP2 EpCAM 61.5% 58.3%
    M2PK MIP1beta IL13 TIMP1 57.3% 57.3%
    M2PK MIP1beta TGFbeta1 TIMP1 58.3% 57.3%
    M2PK MIP1beta IGFBP2 EpCAM 65.6% 57.3%
    M2PK MIP1beta IL8 Mac2BP 54.2% 56.3%
    M2PK IL8 IL13 TGFbeta1 64.6% 56.3%
    M2PK IL8 TIMP1 EpCAM 62.5% 56.3%
    M2PK IL13 Mac2BP TIMP1 57.3% 56.3%
    M2PK TGFbeta1 Mac2BP IGFBP2 60.4% 56.3%
    M2PK IGFBP2 TIMP1 EpCAM 63.5% 56.3%
    M2PK MIP1beta IL8 TIMP1 57.3% 55.2%
    M2PK IL8 TGFbeta1 TIMP1 57.3% 55.2%
    M2PK MIP1beta IL13 Mac2BP 57.3% 54.2%
    M2PK MIP1beta IL8 EpCAM 62.5% 53.1%
    M2PK MIP1beta TIMP1 EpCAM 59.4% 52.1%
    M2PK IL13 TGFbeta1 TIMP1 64.6% 52.1%
    M2PK IL13 Mac2BP EpCAM 51.0% 52.1%
    M2PK MIP1beta IL13 TGFbeta1 57.3% 51.0%
    M2PK MIP1beta Mac2BP IGFBP2 57.3% 51.0%
    M2PK TGFbeta1 Mac2BP EpCAM 52.1% 51.0%
    M2PK TGFbeta1 TIMP1 EpCAM 59.4% 51.0%
    M2PK MIP1beta IL8 IGFBP2 52.1% 50.0%
    M2PK Mac2BP TIMP1 EpCAM 52.1% 50.0%
    M2PK MIP1beta TGFbeta1 Mac2BP 63.5% 49.0%
    M2PK MIP1beta IL13 EpCAM 50.0% 47.9%
    M2PK MIP1beta TGFbeta1 EpCAM 53.1% 47.9%
    M2PK IL13 TGFbeta1 Mac2BP 60.4% 46.9%
    M2PK MIP1beta TGFbeta1 IGFBP2 49.0% 44.8%
  • TABLE 11
    Combination of five biomarkers in serum that equal
    or exceed 50% sensitivity at 95% specificity.
    Test Cross Validated
    Sensitivity at Sensitivity at
    BM1 BM2 BM3 BM4 BM5 95% Specificity 95% Specificity
    Dkk3 M2PK IL8 IL13 Mac2BP 74.0% 70.8%
    Dkk3 M2PK IL8 IL13 TIMP1 71.9% 70.8%
    M2PK TGFbeta1 Mac2BP IGFBP2 EpCAM 69.8% 70.8%
    Dkk3 M2PK MIP1beta IL8 IL13 71.9% 69.8%
    Dkk3 M2PK MIP1beta IGFBP2 EpCAM 71.9% 69.8%
    Dkk3 M2PK IL8 IGFBP2 EpCAM 78.1% 69.8%
    Dkk3 M2PK TGFbeta1 Mac2BP IGFBP2 68.8% 69.8%
    M2PK MIP1beta Mac2BP IGFBP2 EpCAM 69.8% 68.8%
    Dkk3 M2PK IL8 TGFbeta1 Mac2BP 70.8% 67.7%
    Dkk3 M2PK Mac2BP IGFBP2 EpCAM 70.8% 67.7%
    M2PK MIP1beta IL8 IL13 Mac2BP 66.7% 67.7%
    Dkk3 M2PK IL8 IL13 IGFBP2 70.8% 66.7%
    Dkk3 M2PK IL13 TGFbeta1 IGFBP2 68.8% 66.7%
    Dkk3 M2PK IL13 IGFBP2 EpCAM 66.7% 66.7%
    M2PK IL13 Mac2BP IGFBP2 EpCAM 71.9% 66.7%
    Dkk3 M2PK IL8 IL13 TGFbeta1 69.8% 65.6%
    Dkk3 M2PK IL8 Mac2BP TIMP1 67.7% 65.6%
    Dkk3 M2PK TGFbeta1 Mac2BP EpCAM 66.7% 65.6%
    M2PK IL8 IL13 TGFbeta1 Mac2BP 71.9% 65.6%
    M2PK IL8 IL13 TGFbeta1 IGFBP2 66.7% 65.6%
    M2PK IL8 TGFbeta1 Mac2BP IGFBP2 64.6% 65.6%
    M2PK IL8 TGFbeta1 Mac2BP EpCAM 69.8% 65.6%
    Dkk3 M2PK MIP1beta IL8 IGFBP2 66.7% 64.6%
    Dkk3 M2PK MIP1beta Mac2BP IGFBP2 70.8% 64.6%
    M2PK MIP1beta IL8 IL13 TIMP1 65.6% 64.6%
    M2PK MIP1beta IL8 TGFbeta1 Mac2BP 65.6% 64.6%
    M2PK IL8 IL13 Mac2BP EpCAM 76.0% 64.6%
    M2PK IL8 IL13 IGFBP2 EpCAM 72.9% 64.6%
    M2PK IL8 TGFbeta1 Mac2BP TIMP1 64.6% 64.6%
    M2PK IL8 Mac2BP IGFBP2 EpCAM 71.9% 64.6%
    Dkk3 M2PK IL13 TGFbeta1 TIMP1 68.8% 63.5%
    Dkk3 M2PK IGFBP2 TIMP1 EpCAM 67.7% 63.5%
    M2PK MIP1beta IL13 IGFBP2 EpCAM 67.7% 63.5%
    M2PK IL13 TGFbeta1 Mac2BP IGFBP2 66.7% 63.5%
    M2PK IL13 TGFbeta1 IGFBP2 EpCAM 70.8% 63.5%
    Dkk3 M2PK MIP1beta IL13 TGFbeta1 69.8% 62.5%
    Dkk3 M2PK IL8 IL13 EpCAM 65.6% 62.5%
    Dkk3 M2PK IL8 TGFbeta1 IGFBP2 70.8% 62.5%
    Dkk3 M2PK IL8 Mac2BP IGFBP2 69.8% 62.5%
    Dkk3 M2PK IL13 TGFbeta1 Mac2BP 68.8% 62.5%
    Dkk3 M2PK TGFbeta1 Mac2BP TIMP1 68.8% 62.5%
    Dkk3 M2PK TGFbeta1 TIMP1 EpCAM 67.7% 62.5%
    M2PK IL8 IL13 Mac2BP TIMP1 64.6% 62.5%
    M2PK IL8 IL13 IGFBP2 TIMP1 66.7% 62.5%
    M2PK IL13 IGFBP2 TIMP1 EpCAM 68.8% 62.5%
    Dkk3 M2PK MIP1beta IL13 IGFBP2 63.5% 61.5%
    Dkk3 M2PK IL8 TGFbeta1 TIMP1 64.6% 61.5%
  • TABLE 12
    Combination of seven biomarkers in serum that equal or exceed 50% sensitivity at 95% specificity.
    Test Cross Validated
    Sensitivity at Sensitivity at
    BM1 BM2 BM3 BM4 BM5 BM6 BM7 95% Specificity 95% Specificity
    Dkk3 M2PK IL8 Mac2BP IGFBP2 TIMP1 EpCAM 78% 56%
    Dkk3 M2PK MIP1beta IL8 Mac2BP IGFBP2 EpCAM 76% 64%
    Dkk3 M2PK IL8 IL13 Mac2BP IGFBP2 TIMP1 73% 69%
    Dkk3 M2PK IL8 TGFbeta1 Mac2BP IGFBP2 TIMP1 72% 68%
    Dkk3 M2PK IL8 TGFbeta1 Mac2BP IGFBP2 EpCAM 71% 62%
    Dkk3 M2PK MIP1beta IL8 Mac2BP IGFBP2 TIMP1 70% 64%
    Dkk3 M2PK IL8 IL13 Mac2BP IGFBP2 EpCAM 70% 67%
    Dkk3 M2PK IL8 IL13 TGFbeta1 Mac2BP IGFBP2 69% 66%
    M2PK MIP1beta IL8 Mac2BP IGFBP2 TIMP1 EpCAM 69% 55%
    Dkk3 M2PK MIP1beta IL8 IL13 Mac2BP IGFBP2 67% 68%
    Dkk3 M2PK MIP1beta IL8 TGFbeta1 Mac2BP IGFBP2 67% 64%
    Dkk3 M2PK MIP1beta TGFbeta1 Mac2BP IGFBP2 TIMP1 65% 59%
    Dkk3 M2PK IL8 IL13 TGFbeta1 IGFBP2 TIMP1 65% 52%
    Dkk3 M2PK IL8 TGFbeta1 IGFBP2 TIMP1 EpCAM 64% 56%
    Dkk3 M2PK IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 64% 57%
    M2PK MIP1beta IL8 IL13 IGFBP2 TIMP1 EpCAM 64% 50%
    Dkk3 M2PK MIP1beta IL8 IL13 Mac2BP TIMP1 63% 55%
    Dkk3 M2PK MIP1beta IL8 TGFbeta1 IGFBP2 EpCAM 63% 58%
    Dkk3 M2PK MIP1beta IL13 TGFbeta1 Mac2BP IGFBP2 63% 53%
    Dkk3 M2PK IL8 IL13 TGFbeta1 Mac2BP TIMP1 63% 59%
    M2PK MIP1beta IL8 TGFbeta1 Mac2BP IGFBP2 EpCAM 63% 59%
    Dkk3 M2PK MIP1beta IL8 TGFbeta1 Mac2BP TIMP1 62% 51%
    Dkk3 M2PK MIP1beta IL8 IGFBP2 TIMP1 EpCAM 62% 56%
    Dkk3 M2PK MIP1beta IL13 Mac2BP IGFBP2 TIMP1 62% 39%
    Dkk3 M2PK IL8 IL13 TGFbeta1 TIMP1 EpCAM 62% 43%
    Dkk3 M2PK IL8 TGFbeta1 Mac2BP TIMP1 EpCAM 62% 50%
    Dkk3 MIP1beta IL8 IL13 Mac2BP IGFBP2 TIMP1 62% 55%
    Dkk3 MIP1beta IL8 Mac2BP IGFBP2 TIMP1 EpCAM 62% 47%
    M2PK MIP1beta IL8 IL13 Mac2BP IGFBP2 TIMP1 62% 56%
    M2PK IL8 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 62% 49%
    Dkk3 M2PK IL8 IL13 TGFbeta1 IGFBP2 EpCAM 60% 56%
    Dkk3 M2PK IL13 TGFbeta1 Mac2BP IGFBP2 EpCAM 60% 55%
    M2PK MIP1beta IL8 IL13 Mac2BP IGFBP2 EpCAM 60% 53%
  • TABLE 13
    Seven biomarker combinations with Sensitivity between 60% and 52%.
    Test Cross Validated
    Sensitivity at Sensitivity at
    BM1 BM2 BM3 BM4 BM5 BM6 BM7 95% Specificity 95% Specificity
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 EpCAM 59% 54%
    Dkk3 M2PK MIP1beta IL8 IL13 IGFBP2 EpCAM 59% 52%
    Dkk3 M2PK MIP1beta IL8 TGFbeta1 IGFBP2 TIMP1 59% 52%
    Dkk3 M2PK TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 59% 47%
    Dkk3 MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 59% 52%
    Dkk3 MIP1beta IL8 TGFbeta1 Mac2BP IGFBP2 TIMP1 59% 45%
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP 58% 41%
    Dkk3 M2PK MIP1beta IL8 TGFbeta1 TIMP1 EpCAM 58% 50%
    Dkk3 M2PK IL8 IL13 IGFBP2 TIMP1 EpCAM 58% 55%
    Dkk3 MIP1beta IL8 IL13 Mac2BP IGFBP2 EpCAM 58% 50%
    Dkk3 IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 58% 52%
    M2PK IL8 IL13 Mac2BP IGFBP2 TIMP1 EpCAM 58% 52%
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 IGFBP2 57% 49%
    Dkk3 M2PK MIP1beta IL8 IL13 TIMP1 EpCAM 57% 48%
    Dkk3 M2PK MIP1beta IL13 Mac2BP IGFBP2 EpCAM 57% 51%
    Dkk3 M2PK MIP1beta TGFbeta1 Mac2BP IGFBP2 EpCAM 57% 46%
    Dkk3 M2PK MIP1beta TGFbeta1 IGFBP2 TIMP1 EpCAM 57% 49%
    Dkk3 M2PK IL13 Mac2BP IGFBP2 TIMP1 EpCAM 57% 49%
    Dkk3 MIP1beta IL8 TGFbeta1 Mac2BP IGFBP2 EpCAM 57% 50%
    M2PK MIP1beta IL8 IL13 TGFbeta1 TIMP1 EpCAM 57% 43%
    M2PK IL8 IL13 TGFbeta1 IGFBP2 TIMP1 EpCAM 57% 52%
    Dkk3 M2PK MIP1beta Mac2BP IGFBP2 TIMP1 EpCAM 56% 47%
    Dkk3 IL8 IL13 Mac2BP IGFBP2 TIMP1 EpCAM 56% 48%
    MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 56% 54%
    Dkk3 M2PK MIP1beta IL8 TGFbeta1 Mac2BP EpCAM 55% 35%
    Dkk3 M2PK MIP1beta IL13 IGFBP2 TIMP1 EpCAM 55% 44%
    Dkk3 M2PK IL8 IL13 TGFbeta1 Mac2BP EpCAM 55% 35%
    Dkk3 IL8 IL13 TGFbeta1 Mac2BP IGFBP2 EpCAM 55% 50%
    Dkk3 IL8 IL13 TGFbeta1 IGFBP2 TIMP1 EpCAM 55% 54%
    Dkk3 M2PK MIP1beta IL8 IL13 Mac2BP EpCAM 54% 49%
    Dkk3 M2PK MIP1beta IL8 Mac2BP TIMP1 EpCAM 54% 39%
    Dkk3 M2PK IL13 TGFbeta1 IGFBP2 TIMP1 EpCAM 54% 42%
    Dkk3 IL8 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 54% 51%
    M2PK MIP1beta IL8 IL13 TGFbeta1 IGFBP2 EpCAM 54% 37%
    M2PK MIP1beta IL13 Mac2BP IGFBP2 TIMP1 EpCAM 54% 41%
    M2PK IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 54% 43%
    MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 EpCAM 54% 42%
    Dkk3 M2PK MIP1beta IL8 IL13 IGFBP2 TIMP1 53% 52%
    Dkk3 M2PK IL8 IL13 Mac2BP TIMP1 EpCAM 53% 42%
    M2PK MIP1beta IL8 TGFbeta1 IGFBP2 TIMP1 EpCAM 53% 34%
    M2PK MIP1beta IL13 TGFbeta1 Mac2BP IGFBP2 EpCAM 53% 45%
    IL8 IL13 TGFbeta1 Mac2BP IGFBP2 EpCAM 53% 51%
    MIP1beta IL8 IL13 Mac2BP IGFBP2 TIMP1 EpCAM 53% 39%
    MIP1beta IL8 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 53% 51%
  • TABLE 14
    Seven biomarker combinations with sensitivity <53%.
    Test Cross Validated
    Sensitivity at Sensitivity at
    BM1 BM2 BM3 BM4 BM5 BM6 BM7 95% Specificity 95% Specificity
    M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP EpCAM 52% 42%
    Dkk3 M2PK MIP1beta IL13 TGFbeta1 IGFBP2 EpCAM 51% 41%
    Dkk3 MIP1beta IL8 IL13 TGFbeta1 TIMP1 EpCAM 51% 35%
    M2PK MIP1beta IL8 IL13 TGFbeta1 IGFBP2 TIMP1 51% 38%
    M2PK MIP1beta IL8 TGFbeta1 Mac2BP IGFBP2 TIMP1 51% 30%
    M2PK IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 51% 37%
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 TIMP1 50% 48%
    Dkk3 MIP1beta IL8 IL13 TGFbeta1 Mac2BP TIMP1 50% 41%
    Dkk3 MIP1beta IL8 IL13 TGFbeta1 IGFBP2 TIMP1 50% 42%
    M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 50% 49%
    M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP TIMP1 50% 35%
    MIP1beta IL8 IL13 TGFbeta1 IGFBP2 TIMP1 EpCAM 50% 39%
    Dkk3 M2PK MIP1beta IL13 TGFbeta1 IGFBP2 TIMP1 49% 44%
    Dkk3 MIP1beta IL8 IL13 TGFbeta1 Mac2BP EpCAM 49% 33%
    M2PK IL8 IL13 TGFbeta1 Mac2BP TIMP1 EpCAM 49% 43%
    Dkk3 M2PK MIP1beta IL13 Mac2BP TIMP1 EpCAM 48% 43%
    Dkk3 MIP1beta IL8 IL13 Mac2BP TIMP1 EpCAM 48% 38%
    Dkk3 MIP1beta IL8 TGFbeta1 Mac2BP TIMP1 EpCAM 48% 40%
    Dkk3 MIP1beta IL13 Mac2BP IGFBP2 TIMP1 EpCAM 48% 31%
    Dkk3 IL8 IL13 TGFbeta1 Mac2BP TIMP1 EpCAM 48% 38%
    M2PK MIP1beta IL8 IL13 Mac2BP TIMP1 EpCAM 48% 33%
    M2PK MIP1beta IL13 TGFbeta1 IGFBP2 TIMP1 EpCAM 48% 45%
    M2PK MIP1beta TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 48% 37%
    M2PK MIP1beta IL13 TGFbeta1 Mac2BP TIMP1 EpCAM 47% 41%
    IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 47% 40%
    Dkk3 MIP1beta IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 46% 42%
    M2PK MIP1beta IL8 TGFbeta1 Mac2BP TIMP1 EpCAM 46% 30%
    Dkk3 M2PK MIP1beta TGFbeta1 Mac2BP TIMP1 EpCAM 45% 37%
    Dkk3 M2PK IL13 TGFbeta1 Mac2BP TIMP1 EpCAM 45% 41%
    Dkk3 MIP1beta IL8 IL13 TGFbeta1 IGFBP2 EpCAM 45% 33%
    Dkk3 MIP1beta IL8 IL13 IGFBP2 TIMP1 EpCAM 44% 40%
    Dkk3 MIP1beta IL8 TGFbeta1 IGFBP2 TIMP1 EpCAM 44% 43%
    Dkk3 MIP1beta IL13 TGFbeta1 Mac2BP TIMP1 EpCAM 44% 43%
    MIP1beta IL8 IL13 TGFbeta1 Mac2BP TIMP1 EpCAM 44% 28%
    Dkk3 M2PK MIP1beta IL13 TGFbeta1 Mac2BP TIMP1 43% 31%
    Dkk3 MIP1beta IL13 TGFbeta1 IGFBP2 TIMP1 EpCAM 42% 40%
    MIP1beta IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 42% 31%
    Dkk3 M2PK MIP1beta IL13 TGFbeta1 Mac2BP EpCAM 41% 23%
    Dkk3 M2PK MIP1beta IL13 TGFbeta1 TIMP1 EpCAM 41% 33%
    Dkk3 MIP1beta IL13 TGFbeta1 Mac2BP IGFBP2 EpCAM 41% 41%
    Dkk3 MIP1beta TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 41% 39%
    Dkk3 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 41% 37%
    M2PK MIP1beta IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 32% 27%
  • TABLE 15
    Sensitivity of nine biomarker combinations in plasma and serum samples at 95% specificity.
    BM1 BM2 BM3 BM4 BM5 BM6 BM7 BM8 BM9 Plasma Serum
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 73% 77%
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 EpCAM 73% 77%
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP TIMP1 EpCAM 54% 72%
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 IGFBP2 TIMP1 EpCAM 58% 74%
    Dkk3 M2PK MIP1beta IL8 IL13 Mac2BP IGFBP2 TIMP1 EpCAM 74% 70%
    Dkk3 M2PK MIP1beta IL8 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 71% 78%
    Dkk3 M2PK MIP1beta IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 57% 72%
    Dkk3 M2PK IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 67% 76%
    Dkk3 MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 54% 55%
    M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 58% 69%
  • TABLE 16
    Sensitivity of ten biomarker combinations in plasma and serum samples at 95% specificity.
    BM1 BM2 BM3 BM4 BM5 BM6 BM7 BM8 BM9 BM1o Plasma Serum
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 70% 55%
    Dkk3 M2PK MIP1beta IL8 IL13 TGFbeta1 Mac2BP IGFBP2 TIMP1 EpCAM 73% 68%
  • It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the invention as shown in the specific embodiments without departing from the scope of the invention as broadly described.
  • The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.
  • All publications discussed and/or referenced herein are incorporated herein in their entirety.
  • The present application claims priority from AU 2010903140, the entire contents of which are incorporated herein by reference.
  • Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed before the priority date of each claim of this application.
  • REFERENCES
    • Anderson and Hunter (2006) Mol Cell Proteomics, 5:573-588.
    • Cancer in Australia, an overview (2008) AIHW (Australian Institute of Health and Welfare) & AACR (Australian Association of Cancer Registries), Cancer series no. 46, Cat. No: CAN 42, Can berra: AIHW.
    • Etzioni et al. (2003) Nat Rev Cancer, 3:243-252.
    • Hundt et al. (2007) Cancer Epidemiol Biomarkers Prev, 16:1935-1953.
    • Kimmel (1987) Methods Enzymol, 152:507-511.
    • Kwoh et al. (1989) Proc Natl Acad Sci USA. 86:1173-1177.
    • Levin (2004) Gastroenterology, 127:1841-1844.
    • Lieberman (2010) Gastroenterology, 138:2115-2126.
    • Morikawa et al. (2005) Gastroenterology, 129:422-428.
    • Notomi et al. (2000) Nucleic Acids Res. 28:E63.
    • Tonus (2006) World J Gastroenterol, 12:7007-7011.
    • Wahl and Berger (1987) Methods Enzymol, 152:399-407.
    • Walker et al. (1992a) Proc Natl Acad Sci USA. 89:392-396.
    • Walker et al. (1992b) Nucleic Acids Res. 20:1691-1696.

Claims (31)

1. A method for diagnosing or detecting colorectal cancer in a subject, the method comprising:
i) determining the presence and/or level of at least two biomarkers selected from the group consisting of IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1β, TGFβ1, and TIMP-1 in a sample from the subject,
wherein the presence and/or level of the two biomarkers is indicative of colorectal cancer.
2. The method of claim 1, wherein the two biomarkers selected from M2PK, EpCam, IL-13, DKK-3, IL-8 and IGFBP2.
3. The method of claim 1, wherein the method comprises determining the presence and/or level of expression of at least three of the biomarkers.
4. The method of claim 3, wherein the three biomarkers are selected from the group consisting of M2PK, EpCam, IL-13, DKK-3, IL-8, IGFBP2, MIP1β, TGFβ1 and MAC2BP.
5. The method of claim 4, wherein the three biomarkers are as follows:
i) DKK-3, M2PK, and IGFBP2;
ii) M2PK, IGFBP2, and EpCAM;
iii) M2PK, MIP1β, and TGFβ1; or
iv) IL-8, IL-13, and MAC2BP.
6. The method of claim 1, wherein the method comprises determining the presence and/or level of expression of at least four of the biomarkers.
7. The method of claim 6, wherein four biomarkers are as follows:
i) DKK-3, M2PK, MAC2BP, and IGFBP2;
ii) IL-8, IL-13, MAC2BP, and EpCam;
iii) DKK3, M2PK, TGFβ1, and TIMP-1;
iv) M2PK, MIP1β, IL-13, and TIMP-1; or
v) IL-8, MAC2BP, IGFBP2, and EpCam.
8. The method of claim 1, wherein the method comprises determining the presence and/or level of at least five of the biomarkers.
9. The method of claim 1, wherein the method comprises determining the presence and/or level of at least six of the biomarkers.
10. The method of claim 1, wherein the method comprises determining the presence and/or level of at least seven of the biomarkers.
11. The method of claim 1, wherein the method comprises determining the presence and/or level of at least eight of the biomarkers.
12. The method of claim 1, wherein the method comprises determining the presence and or level of at least nine of the biomarkers.
13. The method of claim 1, wherein the method comprises determining the presence and/or level of at least ten of the biomarkers.
14. The method of claim 1, wherein the method comprises detecting the presence and/or level of least one additional biomarker selected from the group consisting of ICF-I, IGF-II, IGF-BP2, Amphiregulin, VEGFA, VEGFD, MMP-1, MMP-2, MMP-3, MMP-7, MMP-9, TIMP-1, TIMP-2, ENA-78, MCP-1, MIP-1β, IFN-γ, IL-10, IL-13, IL-1β, IL-4, IL-8, IL-6, MAC2BP, Tumor M2 pyruvate kinase, M65, OPN, DKK-3, EpCam, TGFβ-1, and VEGFpan.
15. The method of claim 1, wherein the method diagnoses or detects colorectal cancer with a sensitivity of at least 50%.
16. (canceled)
17. (canceled)
18. The method of claim 1, wherein the method diagnoses or detects Dukes Stage A colorectal cancer with a sensitivity of at least 50% and a specificity of at least 95%; a sensitivity of at least 60% and a specificity of at least 80%; or a sensitivity of at least 50% and a specificity of at least 90%.
19. (canceled)
20. (canceled)
21. (canceled)
22. (canceled)
23. (canceled)
24. (canceled)
25. (canceled)
26. The method of claim 1, wherein the method comprises:
i) determining the presence and/or level of the biomarkers in the sample from the subject; and
ii) comparing the presence and/or level of the biomarkers relative to a control, wherein a presence and/or level in the sample that is different to the control is indicative of colorectal cancer.
27. The method of claim 1, wherein the sample comprises blood, plasma, serum, urine, platelets, magakaryocytes or faeces.
28. A method of treatment comprising:
(i) diagnosing or detecting colorectal cancer according to the method of claim 1; and
(ii) administering a therapeutic suitable for the treatment of colorectal cancer.
29. A method for monitoring the efficacy of treatment of colorectal cancer in a subject, the method comprising treating the subject for colorectal cancer and then detecting the presence and/or level of at least two biomarkers selected from IL-8, IGFBP2, MAC2BP, M2PK, IL-13, DKK-3, EpCam, MIP1β, TGFβ1, and TIMP-1 in a sample from the subject, wherein an absence of and/or reduction in the level of expression of the biomarkers after treatment as compared with before treatment is indicative of an effective treatment.
30. (canceled)
31. (canceled)
US13/809,785 2010-07-14 2011-07-14 Diagnostic for colorectal cancer Abandoned US20130345322A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
AU2010903140 2010-07-14
AU2010903140A AU2010903140A0 (en) 2010-07-14 Diagnostic for colorectal cancer
PCT/AU2011/000895 WO2012006681A1 (en) 2010-07-14 2011-07-14 Diagnostic for colorectal cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2011/000895 A-371-Of-International WO2012006681A1 (en) 2010-07-14 2011-07-14 Diagnostic for colorectal cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/471,147 Continuation US10877039B2 (en) 2010-07-14 2017-03-28 Diagnostic for colorectal cancer

Publications (1)

Publication Number Publication Date
US20130345322A1 true US20130345322A1 (en) 2013-12-26

Family

ID=45468816

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/809,785 Abandoned US20130345322A1 (en) 2010-07-14 2011-07-14 Diagnostic for colorectal cancer
US15/471,147 Active US10877039B2 (en) 2010-07-14 2017-03-28 Diagnostic for colorectal cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/471,147 Active US10877039B2 (en) 2010-07-14 2017-03-28 Diagnostic for colorectal cancer

Country Status (13)

Country Link
US (2) US20130345322A1 (en)
EP (2) EP2829881B1 (en)
JP (1) JP6061344B2 (en)
CN (2) CN103140760B (en)
AU (1) AU2011279555B2 (en)
BR (1) BR112013000745B1 (en)
DK (1) DK2829881T3 (en)
ES (1) ES2653646T3 (en)
IN (1) IN2013CN01129A (en)
NO (1) NO2829881T3 (en)
PT (1) PT2829881T (en)
RU (1) RU2013103995A (en)
WO (1) WO2012006681A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150285804A1 (en) * 2012-10-12 2015-10-08 Origin Sciences Limited Diagnostic method for colorectal cancer
US10877039B2 (en) 2010-07-14 2020-12-29 Vision Tech Bio Pty. Ltd. Diagnostic for colorectal cancer
US11208461B2 (en) * 2014-07-18 2021-12-28 Sanofi Method for predicting the outcome of a treatment with aflibercept of a patient suspected to suffer from a cancer
CN114594259A (en) * 2022-04-22 2022-06-07 北京易科拜德科技有限公司 Novel model for colorectal cancer prognosis prediction and diagnosis and application thereof
WO2023015354A1 (en) * 2021-08-11 2023-02-16 Vision Tech Bio Pty Ltd Method of detecting adenoma

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9141756B1 (en) 2010-07-20 2015-09-22 University Of Southern California Multi-scale complex systems transdisciplinary analysis of response to therapy
CA2825894C (en) 2011-02-02 2021-11-30 Amgen Inc. Prognosis of cancer using a circulating biomarker
RS58553B1 (en) * 2012-08-09 2019-05-31 Inst Nat Sante Rech Med Diagnostic of heart failure
AU2015237229A1 (en) * 2014-03-28 2016-11-10 Applied Proteomics, Inc. Protein biomarker profiles for detecting colorectal tumors
CN106191215B (en) * 2015-04-29 2020-03-24 中国科学院上海生命科学研究院 Screening and application of protein molecular marker Dkk-3 related to muscular atrophy
JP7007914B2 (en) * 2015-05-12 2022-02-10 スーパーラブ ファー イースト リミテッド Methods for identifying interferons that have a direct inhibitory effect on tumors and their use
CN105219844B (en) * 2015-06-08 2018-12-14 华夏京都医疗投资管理有限公司 Gene marker combination, kit and the disease risks prediction model of a kind of a kind of disease of screening ten
CN105021828B (en) * 2015-07-20 2017-07-28 上海交通大学医学院附属新华医院 By detecting that three phase colorectal carcinoma patients serums predict tumor patient prognosis
CN105567846A (en) * 2016-02-14 2016-05-11 上海交通大学医学院附属仁济医院 Kit for detecting bacteria DNAs in faeces and application thereof in colorectal cancer diagnosis
JP6984862B2 (en) * 2017-03-15 2021-12-22 国立大学法人金沢大学 Detection of colorectal cancer using blood chemokines as markers
CN107164535A (en) * 2017-07-07 2017-09-15 沈阳宁沪科技有限公司 A kind of noninvasive high flux methylates diagnosis of colon cancer, research and treatment method
CN110540588B (en) * 2018-05-28 2022-08-23 香港中文大学 Epitope polypeptide based on tumor stem cell marker EpCAM and application thereof
CN108957014A (en) * 2018-09-27 2018-12-07 郑州大学第附属医院 Colorectal cancer blood serum designated object, expression appraisal procedure, kit and application
CN115568824A (en) * 2018-10-23 2023-01-06 布莱克索恩治疗公司 System and method for screening, diagnosing and stratifying patients
JP7442967B2 (en) 2018-11-30 2024-03-05 トヨタ紡織株式会社 center bracket
CN109504778B (en) * 2019-01-11 2021-11-09 复旦大学附属中山医院 5hmC multi-molecular marker based on apparent modification and colorectal cancer early diagnosis model
BR112021017248A2 (en) * 2019-03-01 2021-11-09 Advanced Marker Discovery S L Protein signature for the diagnosis of colorectal cancer and/or precancerous stage thereof
JP7032764B2 (en) * 2019-04-03 2022-03-09 京都府公立大学法人 How to detect colorectal cancer
JP7300660B2 (en) * 2019-04-03 2023-06-30 京都府公立大学法人 Methods for detecting colorectal cancer
EP3835789A1 (en) 2019-12-13 2021-06-16 Deutsches Krebsforschungszentrum, Stiftung des öffentlichen Rechts Biomarker panel for diagnosing colorectal cancer
WO2022130597A1 (en) * 2020-12-18 2022-06-23 国立大学法人東北大学 Inference device, inference method, inference program, generation device, and inference system
KR20240004546A (en) * 2021-04-20 2024-01-11 비전 테크 바이오 피티와이 엘티디 Biomarkers for Colorectal Cancer
WO2023128429A1 (en) * 2021-12-31 2023-07-06 주식회사 이노제닉스 Method for screening colorectal cancer and advanced adenoma, and application thereof
WO2023128419A1 (en) * 2021-12-31 2023-07-06 주식회사 이노제닉스 Method for screening colorectal cancer and colorectal polyps or advanced adenomas and application thereof
PL441319A1 (en) * 2022-05-31 2023-12-04 Uniwersytet Medyczny Im. Piastów Śląskich We Wrocławiu Set of protein biomarkers and in vitro method for diagnosing colorectal cancer

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008138522A1 (en) * 2007-05-10 2008-11-20 Roche Diagnostics Gmbh Use of timp-1 as a marker for colorectal cancer
WO2009037572A2 (en) * 2007-06-04 2009-03-26 Diagnoplex Biomarker combinations for colorectal cancer
WO2010096674A2 (en) * 2009-02-20 2010-08-26 The Regents Of The University Of California A+ biomarker assays

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5925517A (en) 1993-11-12 1999-07-20 The Public Health Research Institute Of The City Of New York, Inc. Detectably labeled dual conformation oligonucleotide probes, assays and kits
US20040157278A1 (en) * 2002-12-13 2004-08-12 Bayer Corporation Detection methods using TIMP 1
US20050014165A1 (en) * 2003-07-18 2005-01-20 California Pacific Medical Center Biomarker panel for colorectal cancer
EP1664796B1 (en) * 2003-09-15 2010-12-15 Oklahoma Medical Research Foundation Method of using cytokine assays to diagnose, treat, and evaluate ankylosing spondylitis
BRPI0401830A (en) 2004-05-25 2006-01-17 Bentonit Uniao Nordeste Sa Production Process of Dry Granulated Powders
ES2427924T3 (en) 2006-06-30 2013-11-04 Merck Sharp & Dohme Corp. IGFBP2 biomarker
WO2008073660A1 (en) 2006-11-09 2008-06-19 University Of Washington Molecules and methods for treatment and detection of cancer
EP2094719A4 (en) * 2006-12-19 2010-01-06 Genego Inc Novel methods for functional analysis of high-throughput experimental data and gene groups identified therfrom
EP2118319A4 (en) 2007-02-12 2010-08-04 Univ Johns Hopkins Early detection and prognosis of colon cancers
JP5409395B2 (en) * 2007-02-27 2014-02-05 セントクローネ インターナショナル エービー Multiplex detection of tumor cells using a panel of agents that bind to extracellular markers
WO2008116178A2 (en) * 2007-03-21 2008-09-25 Vanderbilt University Systems and methods for diagnosis and prognosis of colorectal cancer
EP2145021A2 (en) * 2007-05-17 2010-01-20 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to insulin growth factor-1 receptor modulators
US20090017463A1 (en) * 2007-07-10 2009-01-15 Vanderbilt University Methods for predicting prostate cancer recurrence
WO2009126543A1 (en) * 2008-04-08 2009-10-15 Nuclea Biomarkers, Llc Biomarker panel for prediction of recurrent colorectal cancer
US20100111969A1 (en) * 2008-10-31 2010-05-06 Apogenix Gmbh Il-4 receptor and il-13 as prognostic markers for colon and pancreas tumors
US20100173024A1 (en) * 2008-12-01 2010-07-08 LifeSpan Extension, LLC Methods and compositions for altering health, wellbeing, and lifespan
EP2370813A4 (en) * 2008-12-04 2012-05-23 Univ California Materials and methods for determining diagnosis and prognosis of prostate cancer
US20110177525A1 (en) * 2010-01-19 2011-07-21 Predictive Biosciences, Inc. Antibodies and methods of diagnosing diseases
CN103140760B (en) 2010-07-14 2016-01-27 联邦科学与工业研究组织 The diagnosis of colorectal cancer

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008138522A1 (en) * 2007-05-10 2008-11-20 Roche Diagnostics Gmbh Use of timp-1 as a marker for colorectal cancer
WO2009037572A2 (en) * 2007-06-04 2009-03-26 Diagnoplex Biomarker combinations for colorectal cancer
WO2010096674A2 (en) * 2009-02-20 2010-08-26 The Regents Of The University Of California A+ biomarker assays

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Antolovi et al, BMC Biotechology 10:35, April, 2010. *
Baier et al, Anticancer Research vol 25:3581-3584, 2005 *
Burgdorf et al, Acta Oncologica 48:1157-1164. 2009 *
Haug et al, British J Can, 96:1329-1334, 2007 *
Lacovazzi et al, 32:160-164, 2010 *
Liou et al J clin Endocrinol Metab, 95: 1717-1725, 4/2010 *
Todaro et al, Cell Stem cell, 1:389-4-2, 2007 *
Zitt et al, Disease Markers 24: 101-109, 2008 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10877039B2 (en) 2010-07-14 2020-12-29 Vision Tech Bio Pty. Ltd. Diagnostic for colorectal cancer
US20150285804A1 (en) * 2012-10-12 2015-10-08 Origin Sciences Limited Diagnostic method for colorectal cancer
US11208461B2 (en) * 2014-07-18 2021-12-28 Sanofi Method for predicting the outcome of a treatment with aflibercept of a patient suspected to suffer from a cancer
WO2023015354A1 (en) * 2021-08-11 2023-02-16 Vision Tech Bio Pty Ltd Method of detecting adenoma
CN114594259A (en) * 2022-04-22 2022-06-07 北京易科拜德科技有限公司 Novel model for colorectal cancer prognosis prediction and diagnosis and application thereof

Also Published As

Publication number Publication date
RU2013103995A (en) 2014-08-20
EP2829881A3 (en) 2015-05-13
US20170205414A1 (en) 2017-07-20
JP2013533977A (en) 2013-08-29
PT2829881T (en) 2017-11-29
ES2653646T3 (en) 2018-02-08
EP2593795A1 (en) 2013-05-22
EP2829881A2 (en) 2015-01-28
EP2829881B1 (en) 2017-08-23
IN2013CN01129A (en) 2015-07-31
CN105755112A (en) 2016-07-13
EP2593795A4 (en) 2014-01-22
NO2829881T3 (en) 2018-01-20
BR112013000745A2 (en) 2016-05-24
AU2011279555B2 (en) 2016-10-20
JP6061344B2 (en) 2017-01-18
CN105755112B (en) 2019-06-07
DK2829881T3 (en) 2017-12-04
CN103140760B (en) 2016-01-27
CN103140760A (en) 2013-06-05
US10877039B2 (en) 2020-12-29
AU2011279555A1 (en) 2013-01-31
WO2012006681A1 (en) 2012-01-19
BR112013000745B1 (en) 2021-02-02

Similar Documents

Publication Publication Date Title
US10877039B2 (en) Diagnostic for colorectal cancer
US9964542B2 (en) Bladder cancer detection composition, kit, and associated methods
CN105102631A (en) Molecular diagnostic test for cancer
AU2009240781B2 (en) An assay to detect a gynecological condition
CN113234830B (en) Product for lung cancer diagnosis and application
US20140162891A1 (en) Methods and materials for detection, diagnosis and management of ovarian cancer
CN113981098A (en) Biomarker for liver cancer diagnosis and liver cancer prognosis prediction
US20200011871A1 (en) Kits and methods for detecting markers
CN114107504A (en) Biomarker for detecting lung cancer and prognosis of lung cancer
CN113943815A (en) ALK-based biomarker panel and application thereof
CN113981097A (en) HSPA 4-based biomarker group and application thereof in liver cancer
CN112795658A (en) Biomarker-based diagnosis of early colorectal cancer
CN115087869A (en) Multiple biomarkers for lung cancer diagnosis and application thereof
US20150376723A1 (en) Prognostic biomarkers for influenza
WO2020099895A1 (en) Detection of bladder cancer
CN114015778A (en) Biomarker panel for predicting lung cancer
CN112921094A (en) Biomarker for assessing early colorectal cancer
CN112921095A (en) Biomarker for diagnosing early colorectal cancer and application thereof
KR20240004546A (en) Biomarkers for Colorectal Cancer
CN112921096A (en) Prediction of early colorectal cancer
CN113061657A (en) Product and system for diagnosing early colorectal cancer
CN113025717A (en) Indicators of early colorectal cancer
CA3228665A1 (en) Method of detecting adenoma

Legal Events

Date Code Title Description
AS Assignment

Owner name: COMMONWEALTH SCIENTIFIC AND INDUSTRIAL RESEARCH OR

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COSGROVE, LEAH JANE;TABOR, BRUCE;BURGESS, ANTONY WILKS;AND OTHERS;SIGNING DATES FROM 20130411 TO 20130722;REEL/FRAME:031113/0100

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: VISION TECH BIO PTY. LTD., AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:COMMONWEALTH SCIENTIFIC AND INDUSTRIAL RESEARCH ORGANISATION;REEL/FRAME:044339/0890

Effective date: 20171010