US20110306514A1 - Ratio based biomarkers and methods of use thereof - Google Patents

Ratio based biomarkers and methods of use thereof Download PDF

Info

Publication number
US20110306514A1
US20110306514A1 US13/144,474 US201013144474A US2011306514A1 US 20110306514 A1 US20110306514 A1 US 20110306514A1 US 201013144474 A US201013144474 A US 201013144474A US 2011306514 A1 US2011306514 A1 US 2011306514A1
Authority
US
United States
Prior art keywords
cancer
protein
sample
proteins
cancer associated
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
US13/144,474
Other languages
English (en)
Inventor
Stephen M. Hewitt
Joon-Yong Chung
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.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by US Department of Health and Human Services filed Critical US Department of Health and Human Services
Priority to US13/144,474 priority Critical patent/US20110306514A1/en
Assigned to THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHUNG, JOON-YONG, HEWITT, STEPHEN M.
Assigned to THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHUNG, JOON-YONG, HEWITT, STEPHEN M.
Publication of US20110306514A1 publication Critical patent/US20110306514A1/en
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
    • G01N33/57484Immunoassay; 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
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/582Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with fluorescent label
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • G16B25/10Gene or protein expression profiling; Expression-ratio estimation or normalisation
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B99/00Subject matter not provided for in other groups of this subclass
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/20ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for computer-aided diagnosis, e.g. based on medical expert systems
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16ZINFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS, NOT OTHERWISE PROVIDED FOR
    • G16Z99/00Subject matter not provided for in other main groups of this subclass
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/60Complex ways of combining multiple protein biomarkers for diagnosis

Definitions

  • This disclosure relates to identification of ratio-based biomarkers for the detection, progression and prognosis of disease, such as cancer, in a subject. Also provided are similar methods for determination of the etiology or risk associated with a disease or condition. This disclosure also relates to methods of predicting survival probabilities and prognosis for a subject, and to methods of stratifying patient therapeutic regimes.
  • Tumors are characterized by their extensive heterogeneity and histopathologic variability. Currently, more than 250 malignant tumors and thousands of subtypes and histologic variants have been described in humans. Nevertheless, the classic pathologic criteria, such as tumor size, grade of malignancy, and metastatic dissemination, are generally the most relevant prognostic factors in cancer. In addition to the variability of histopathologic subtypes, molecular study of tumors is even more complex. In malignant tumors, at least six genetic alterations are believed to affect the main mechanisms of cellular transformation, including growth factor and cell signaling pathways, the cell cycle, apoptosis, and mechanisms implicated in cellular invasiveness, and angiogenesis (Hanahan and Weinberg, Cell. 100(1):57-70, 2000). Overall, more than 350 genes associated with tumors have been identified, representing more than 1% of the human genome (Futreal et al., Nat Rev Cancer. 4(3):177-83, 2004).
  • the aberrant behavior of cancer in part reflects an up-regulation of certain oncogenic signaling pathways that promote proliferation, inhibit apoptosis and enable the cancer to spread and evoke angiogenesis.
  • rates of success for the treatment of tumors or responsiveness of tumors after administration with such non-cytotoxic agents have been erratic.
  • Several molecular targets have been identified as potential therapeutic targets for the treatment of solid tumors. Several of these molecular targets are proteins, found in cell signaling or growth factor pathways that are associated with the occurrence of cancer.
  • TMA analysis allows for the study of proteomics at the tissue level.
  • TMAs can be constructed from normal or diseased tissue, with a tissue section from each, being used to evaluate one or more proteins, such as the presence of absence or a disease marker protein, by immunochemical staining.
  • TMA analysis requires a solid tissue sample, and antibodies that bind to formalin-fixed, paraffin embedded samples.
  • TMA analysis over other tissue-based proteomic profiling techniques is that multiple antigens can be assayed from a single tissue section simultaneously, and that the TMA retains the pathological structure of the tissue section from which it was derived. This information is particularly important when comparing or contrasting TMA expression profile results with pathology results of the same tissue section.
  • Another commonly used technique to profile protein expression profiles is LCM. While LCM does provide the capacity to perform a directed western blot on a tissue section, the methodology is time consuming and does not provide a global expression view of a targeted protein. Immunohistochemistry while providing excellent localization, lacks quantification without sophisticated equipment such as high resolution tandem mass spectrometry, and lacks a normalization component.
  • compositions, methods and apparatus for determining prognosis for a disease or condition by identifying at least two proteins the expression (or loss of expression) of which is associated with the disease or condition in a sample from a subject with the disease or condition (or suspected to have or be susceptible to the disease or condition); quantifying the at least two disease/condition associated proteins in the sample; normalizing each associated protein; comparing the normalized value of the first disease/condition associated protein with the normalized value of the second disease/condition associated protein to obtain a biomarker indicator and; correlating the biomarker indicator with the prognosis of the subject.
  • compositions, methods and apparatus for determining cancer prognosis (e.g., survival probability) of a subject with a cancer e.g., a solid tumor, carcinoma or other classes of tumor
  • a cancer e.g., a solid tumor, carcinoma or other classes of tumor
  • identifying at least two cancer associated proteins in a sample from the subject quantifying the at least two cancer associated proteins in the sample; normalizing each cancer associated protein; comparing the normalized value of the first cancer associated protein with the normalized value of the second cancer associated protein to obtain a biomarker indicator and; correlating the biomarker indicator with survival probability of the subject.
  • compositions, methods and devices used for determining cancer survival probability of a subject are adapted for use with other diseases or conditions, as well as for examining the diagnosis, prognosis and/or prediction of response and determination of etiology or risk, of such other diseases or conditions.
  • embodiments are illustrated using cancer but it is understood that these are not to be viewed as restricted to cancer.
  • the present disclosure is further directed to methods for identifying cancer (or other disease or condition) associated proteins expressed in tissue samples, and for correlating the expression profile of the associated proteins with, for instance, various cancers, prognosis, or responses to therapies.
  • the present disclosure is further directed to methods for detecting the presence of cancer in a subject by determining levels of a first cancer associated protein and a second cancer associated protein from a biological sample from the subject; normalizing the first and second cancer associated protein contents against total cellular protein content from the biological sample; and comparing the normalized levels of the first and second cancer associated proteins with levels of the first and second cancer associated proteins in cells, tissues or bodily fluids measured in a normal control subject, wherein a change in the normalized levels of the first and second cancer associated proteins in the subject versus levels of the first and second cancer associated proteins measured in a normal control subject is associated with the presence of cancer in the subject.
  • the disclosure provides a method for detecting the presence of a cancer in a subject by detecting in a biological sample from the subject the level of a first cancer associated protein, wherein the first cancer associated protein comprises PTEN, p-AKT, p-mTOR, p-MAPK, EGFR, HER2, HER3, or a combination of two or more thereof; and comparing the level of expression of the first cancer associated protein detected in the biological sample from the subject to a predetermined statistically significant cut-off value, wherein a change (e.g., decrease or increase) in the level of expression of the first cancer associated protein in the biological sample compared to a non-cancerous (e.g., non-transformed) sample is indicative of the presence of the cancer in the subject.
  • a change e.g., decrease or increase
  • the disclosure provides a method for detecting the presence of a cancer in a subject by calculating the level of p-AKT or p-mTOR expression in a sample from the subject; normalizing the level of p-AKT or p-mTOR expression against total cellular protein content from the sample; and comparing levels of normalized expression of p-AKT or p-mTOR expression in a control non-cancerous sample (for instance, a sample from a subject without cancer, or a sample from non-cancerous tissue in the same subject, etc.), wherein a statistically significant change in the level of p-AKT or p-mTOR expression in the subject's sample as compared to the normal non-cancerous sample is an indication of the presence of cancer in the subject.
  • the cancer biomarkers involve measurement of p-MAPK, EGFR, HER2, and/or HER3.
  • Also provided is a method for identifying a survival-based cancer biomarker indicator wherein the biomarker indicator comprises at least two cancer associated proteins from a cell signaling pathway associated with the cancer, and wherein the at least two cancer associated proteins are used to obtain the survival-based cancer biomarker indicator by calculating the content (level) of the first cancer associated protein in a sample, calculating the content (level) of the second cancer associated protein in the sample, normalizing the first cancer associated protein content against the total cellular protein content in the sample, and normalizing the second cancer associated protein content against the total cellular protein content in the sample, to obtain the survival-based cancer biomarker indicator.
  • the disclosure provides a method of determining relative cancer survival rates (or more generally prognosis) for a subject with a solid tumor by obtaining a biomarker indicator, the biomarker indicator being obtained by acquiring a solid tumor sample from the subject, extracting a first cancer associated protein from the solid tumor to produce a fraction comprising the first cancer associated protein, calculating the content of the first cancer associated protein in the fraction, normalizing the first cancer associated protein content against total cellular content in the fraction, extracting a second cancer associated protein from the solid tumor sample, calculating the content of the second cancer associated protein in the fraction, normalizing the second cancer associated protein content against total cellular protein content in the fraction, and correlating the normalized first cancer associated protein content against the normalized second cancer associated protein content to obtain a biomarker indicator, and comparing the biomarker indicator with relative survival rates, thereby determining the relative cancer survival rate for the subject with the solid tumor.
  • the present disclosure is further directed to methods for predicting relative cancer survival rates for a subject with a solid tumor by detecting the presence of an antibody to a tumor antigen in the solid tumor, wherein the tumor antigen involves increased expression of p-AKT and p-mTOR or decreased expression of PTEN as compared to a normal non-cancerous sample, thereby detecting the cancer in the subject, and correlating decreased expression of the tumor antigen in the subject as compared to a normal non-cancerous sample with a lower survival rate in the subject with the solid tumor.
  • the calculated tumor antigen involves increased HER2 relative to and/or along with decreased HER3 expression.
  • the absolute value of the level of individual tumor (or other disease/condition) antigen is not necessarily determinative—rather, it is the relative amount compared to one or more other antigens that provides the predictive biomarker described herein.
  • the disclosure provides a kit comprising a membrane array and detector molecules for the detection of cancer associated proteins in a sample, the array comprises a plurality of membranes, wherein each of the plurality of membranes has substantially a same affinity for the cancer associated proteins and containers comprise detector molecules for detecting the cancer associated proteins captured on each membrane, wherein the cancer associated proteins are selected from a group of cancers consisting of solid tumors, leukemia, multiple myeloma or lymphoma.
  • the instant disclosure identifies disease or condition associated biomolecules (such as proteins and nucleic acids) that can be used to detect, diagnose, identify subjects suitable for particular treatment regimes and provides prognosis information for such subjects.
  • the subject has cancer and the biomolecules are cancer associated biomolecules.
  • the instant disclosure also identifies a method for characterizing protein expression profiles in a sample and correlating the protein expression profiles with a survival-based cancer biomarker indicator for developing cancer, confirmation of the presence of a cancer, or the relative survival rates for a subject affected by the cancer.
  • FIG. 1A is a photograph of images showing phospho-AKT (p-AKT), phospho-mTOR (p-mTOR), and PTEN expression by multiplex tissue immunoblotting (MTI).
  • FIG. 1B is photograph of images showing immunohistochemical staining of p-AKT, p-mTOR, and PTEN protein in dysplasia and extrahepatic cholangiocarcinoma (EHCC) samples.
  • EHCC extrahepatic cholangiocarcinoma
  • FIG. 2A is a Box plot of relative expression rate of p-AKT protein among normal biliary epithelia, dysplasia, and cancer cases.
  • FIG. 2A shows EHCC cases had significantly higher expression of p-AKT than normal and dysplastic epithelia cases.
  • FIG. 2B is a Box plot of relative expression rate of p-mTOR protein among normal biliary epithelia, dysplasia and cancer cases.
  • FIG. 2B shows that EHCC cases had significantly higher expression of p-mTOR than normal and dysplastic epithelia cases.
  • FIG. 2C is a linear-based correlation between p-AKT and p-mTOR protein expression.
  • FIG. 3 is a Box plot of relative expression rate of PTEN and its association with other clinicopathologic factors.
  • FIG. 3A shows that cases with T1 classification had a significantly higher relative PTEN expression than those cases with other classifications.
  • FIG. 3B is a box plot of relative expression of PTEN and its association with patients with duodenal invasion.
  • FIG. 3B shows that patients with duodenal invasion had significantly less PTEN expression than those without duodenal invasion.
  • FIG. 3C is a box plot of relative expression of PTEN and its association with patients with higher stage grouping.
  • FIG. 3C shows that patients with higher stage grouping had significantly less PTEN expression than those with lower stage grouping.
  • FIG. 3D is a box plot of relative expression of PTEN and its association with depth of tumor invasion.
  • FIG. 3D shows that patients with less tumor cell invasion had a statistically greater PTEN expression than cases with deeper tumor cell invasion, but no statistical difference with those with depth of invasion between 0.5 cm and 1.2 cm.
  • FIG. 4 is a Kaplan-Meier survival analysis of EHCC according to PTEN expression.
  • FIG. 4 shows that patients with low PTEN expression have a lower relative survival rate than patients with high PTEN expression.
  • FIG. 5 is a Kaplan-Meier survival analysis of EHCC cases according to PTEN/p-AKT or PTEN/p-mTOR expression.
  • FIG. 5A shows a Kaplan-Meier survival analysis of EHCC cases according to PTEN/pAKT expression.
  • FIG. 5A shows that low expressers of PTEN/pAKT have a significantly worse rate of survival than high expressers of PTEN/p-AKT expression.
  • FIG. 5B is a Kaplan-Meier survival analysis of EHCC cases according to PTEN/p-mTOR expression.
  • FIG. 5B shows that low expressers of PTEN/p-mTOR have a significantly worse rate of survival than high expressers of PTEN/p-mTOR expression.
  • FIG. 6 is a series of photographic images showing immunohistochemical staining of phosphorylated mammalian target of rapamycin (p-mTOR), phosphorylated protein kinase B (p-AKT; T308), phosphorylated mitogen-activated protein kinase (p-MAPK), and epidermal growth factor receptor (EGFR).
  • p-mTOR phosphorylated mammalian target of rapamycin
  • p-AKT phosphorylated protein kinase B
  • p-MAPK phosphorylated mitogen-activated protein kinase
  • EGFR epidermal growth factor receptor
  • FIG. 7 illustrates hierarchical clustering of correlation coefficients of immunohistochemical expression of p-AKT, p-MAPK, p-mTOR and EGFR, which was performed with Weight Score.
  • Four groups (Category 1 to 4) were defined. This figure demonstrates that hierarchical cluster analysis does not identify ratio based biomarkers.
  • FIG. 8 shows Kaplan-Meier survival analysis of non-small cell lung cancer patients.
  • FIG. 8A illustrates the correlation of single each antibody expression with patients' outcome.
  • FIG. 8B illustrates the correlation of the ratio p-mTOR to p-AKT (p-mTOR/p-AKT) and the ratio of p-MAPK to EGFR (p-MAPK/EGFR) with patients' outcome.
  • FIG. 8C illustrates t correlation of three groups; both of the ratios were high (+/+), either of them were high (+/ ⁇ ), both of them were low ( ⁇ / ⁇ ).
  • FIG. 8D illustrates the correlation of Double ratio with patients' outcome.
  • eIF4E eukaryotic initiation factor 4E
  • AKT protein kinase B
  • CEA carcinoembryonic antigen
  • EGFR epidermal growth factor receptor
  • EHCC extrahepatic cholangiocarcinoma
  • HER human epidermal growth factor receptor
  • HSP heat shock protein
  • MAPK mitogen-activated protein kinase
  • MTI multiplex tissue immunoblotting
  • mTOR mammalian target of rapamycin
  • mTORC mammalian target of rapamycin complex
  • p-AKT phosphorylated AKT
  • p-EGFR phosphorylated EGFR
  • PI3K phosphatidyl inositol 3 kinase
  • pMAPK phosphorylated mitogen-activated protein kinase
  • PSA prostate specific antigen
  • PTEN phosphatase and tensin homolog deleted on chromosome 10
  • TMA tissue micro array(s)
  • Array An arrangement of molecules, particularly biological macromolecules (such as proteins or polypeptides) or biological samples (such as cells or tissue sections), in addressable locations on or in a substrate.
  • the array may be regular (arranged in uniform rows and columns, for instance) or irregular (such as a tissue section).
  • the number of addressable locations on the array can vary, for example from a few (such as two) to more than 50, 100, 200, 500, 1000, 10,000, or more.
  • a “microarray” is an array that is miniaturized so as to require or be aided by microscopic examination for evaluation or analysis.
  • each arrayed sample (“feature”) is addressable, in that its location can be reliably and consistently determined within the at least two dimensions of the array.
  • the location of each sample/feature within the array is assigned to the sample at the time when it is applied to the array, and a key may be provided in order to correlate each location with the appropriate target or feature position.
  • ordered arrays are arranged in a symmetrical grid pattern, but samples could be arranged in other patterns (e.g., in radially distributed lines, spiral lines, or ordered clusters).
  • Addressable arrays usually are computer readable, in that a computer can be programmed to correlate a particular address on the array with information about the sample at that position (e.g., hybridization or binding data, including for instance signal intensity).
  • the individual features in the array are arranged regularly, for instance in a Cartesian grid pattern, which can be correlated to address information by a computer.
  • Feature(s) within an array may assume many different shapes.
  • spot refers generally to a localized placement of molecules or cells, and is not limited to a round or substantially round region.
  • substantially square regions of application can be used with arrays encompassed herein, as can be regions that are, for example substantially rectangular, triangular, oval, irregular, or another shape.
  • one or more features will occur on the array a plurality of times (e.g., twice, though more are also contemplated) to provide internal controls.
  • the array will replicate the position of features in a sample, for example, the location of markers of interest in a tissue section.
  • the array markers of interest will be transferred from the tissue section to another medium, such as a membrane, for example a nitrocellulose membrane, wherein the features are evaluated.
  • Binding or interaction An association between two substances or molecules.
  • the arrays are used to detect hybridization/binding or other interaction of a labeled molecule (termed a “probe” herein) with an immobilized target molecule in the array.
  • a probe “binds” to a target molecule in a feature on an array if, after incubation of the probe (usually in solution or suspension) with or on the array (or a slice of the array) for a period of time (usually 5 minutes or more, for instance 10 minutes, 20 minutes, 30 minutes, 60 minutes, 90 minutes, 120 minutes or more), a detectable amount of the probe associates with a feature of the array to such an extent that it is not removed when the array is washed with a relatively low stringency buffer.
  • Appropriate buffers for washing TMAs will depend on the constituents of the features of the array, and thus may be those used in washing nucleic acid hybridization systems (e.g., higher salt (such as 3 ⁇ or higher saline-sodium citrate (SSC) buffer) room temperature washes), protein interaction systems (e.g., 100 mM KCl), and so forth.
  • nucleic acid hybridization systems e.g., higher salt (such as 3 ⁇ or higher saline-sodium citrate (SSC) buffer
  • SSC saline-sodium citrate
  • Washing can be carried out, for instance, at room temperature, but other temperatures (either higher or lower) can also be used. Probes will bind target molecules to different extents, and the term “bind” encompasses both relatively weak and relatively strong interactions. Thus, some binding will persist after the array is washed in a way that is appropriate to remove the probe molecule. For instance in a lower salt buffer (such as about 0.5 to about 1.5 ⁇ SSC), 55-65° C. washes can be used for nucleic acid probes, or a higher salt buffer (e.g., 500 mM or 1000 mM KCl, tris-buffered saline with Tween® 20 (TBST)) for protein probes, and so forth.
  • a lower salt buffer such as about 0.5 to about 1.5 ⁇ SSC
  • 55-65° C. washes can be used for nucleic acid probes
  • a higher salt buffer e.g., 500 mM or 1000 mM KCl, tris-buffered s
  • probe and target molecules are nucleic acids
  • binding of the probe to a target can be discussed in terms of the specific complementarity between base sequences of the probe and the target nucleic acid.
  • probe or the target is a protein
  • specificity of binding and binding affinity can be discussed.
  • binding characteristics of an array for a particular probe refers to the specific binding pattern (and optionally the specific relative signal intensities) that forms between the probe and the array after excess (unbound or not specifically bound) probe is washed away.
  • This pattern (which may contain no positive signals, some or all positive signals, and will likely have signals of differing intensity) conveys information about the binding affinity of that probe for molecules within the spots or tissue sections of the array, and can be decoded by reference to the key of the array (which lists the addresses of the spots on the array surface or identifies the probe's potential binding partner).
  • the relative intensity of the binding signal from individual features in many instances is indicative of the relative level in a particular feature on the array of the target that binds to or interacts with the probe.
  • Quantification of the binding pattern of an array/probe combination can be carried out using any of several existing techniques, including scanning the signals' intensities into a computer for calculation of relative density of each spot.
  • Biomarker Indicator A molecular-biology based diagnostic and/or prognostic indication that disease may be present, may develop, and the like.
  • the biomarker indicator is a prognostic and/or diagnostic indicator of the development of a disease such as cancer, and associated rate of survival (or other prognosis) for a subject with the cancer.
  • Biomarker indicators are determined by calculating the content/level of at least two disease/cancer associated proteins in a sample, and normalizing the content of the two or more associated proteins relative to total cellular protein content in the sample.
  • the biomarker indicator includes the ratio (quotient) of the level of one protein to another, the ratio of two proteins to one or one protein to two, the sum of the levels of two proteins, or the sum of the two or more ratios of protein levels, the difference between the levels of two (or more) proteins or the ratios of proteins, the mathematical product (that is, result of multiplying together) of the levels or two or more proteins or ratios thereof, and so forth.
  • Cancer Associated Protein A substance produced in tumor cells that trigger an immune response in the host.
  • Cancer Associated Protein is used interchangeably with Tumor Antigen.
  • the substance may be broadly categorized based on the substance's expression pattern and/or location of expression. For example, Tumor-Specific Antigens are present only in tumor cells and are not found in normal/healthy cells. Tumor-Associated Antigens are present on some tumor cells and also present on some normal/healthy cells.
  • the above definition also encompasses the terms “Cancer-Specific Markers” and “Tissue-Specific Markers”. Cancer-Specific markers are related to the presence of a certain cancerous tissue.
  • Tissue-Specific Markers are carcinoembryonic antigen (CEA), a blood-borne protein first noted to be produced by tumors of the gastrointestinal system.
  • CEA carcinoembryonic antigen
  • Tissue-Specific Markers are related to specific tissues which have developed cancer. Generally speaking, these substances are not specifically related to the tumor, and may be present at elevated levels when no cancer is present.
  • elevated levels of Tissue-Specific Markers point to a specific tissue being at fault. For example, highly elevated levels of PSA (Prostate Specific Antigen) are often associated with the development of prostate cancer.
  • PSA Prostate Specific Antigen
  • Freezing refers to the solidification of a liquid sample, to a point of solidity (rigidity) sufficient that it can be sectioned or sliced. Freezing usually occurs at a temperature at or below the freezing temperature of water, but where the sample contains constituents other than water, the “freezing” (solidification) point may be substantially different from 0° C.
  • a liquid biological sample such as sera, may be frozen in an embedding compound so that the sample can be sectioned, sliced, stained and evaluated.
  • Fluorophore A chemical compound, which when excited by exposure to a particular wavelength of light, emits light (i.e., fluoresces), for example at a different wavelength. Fluorophores can be described in terms of their emission profile, or “color.” Green fluorophores, for example Cy3, FITC, and Oregon Green, are characterized by their emission at wavelengths generally in the range of 515-540 ⁇ . Red fluorophores, for example Texas Red, Cy5 and tetramethylrhodamine, are characterized by their emission at wavelengths generally in the range of 590-690 ⁇ .
  • fluorophores examples include for instance: 4-acetamido-4′-isothiocyanatostilbene-2,2′ disulfonic acid, acridine and derivatives such as acridine and acridine isothiocyanate, 5-(2′-aminoethyl)aminonaphthalene-1-sulfonic acid (EDANS), 4-amino-N-[3-vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate (Lucifer Yellow VS), N-(4-anilino-1-naphthyl)maleimide, anthranilamide, Brilliant Yellow, coumarin and derivatives such as coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), 7-amino-4-trifluoromethylcouluarin (Coumaran
  • fluorophores include CFP (green fluorescent protein) and variants and derivatives thereof, LissamineTM, diethylaminocoumarin, fluorescein chlorotriazinyl, naphthofluorescein, 4,7-dichlororhodamine and xanthene and derivatives thereof.
  • CFP green fluorescent protein
  • LissamineTM diethylaminocoumarin
  • fluorescein chlorotriazinyl 1,4-dichlororhodamine
  • xanthene 1,7-dichlororhodamine
  • High-throughput genomics or proteomics Application of genetic data such as genes or proteins with various techniques such as microarrays or other genomic technologies to rapidly identify large numbers of genes or proteins, or distinguish their structure, expression or function from normal or abnormal cells or tissues.
  • Isolated An “isolated” biological component (such as a nucleic acid molecule, protein or organelle) has been substantially separated or purified away from other biological components in the cell of the organism in which the component naturally occurs, i.e., other chromosomal and extra-chromosomal DNA and RNA, proteins and organelles, or from other components in the reaction mixture used to generate the molecule (if it is synthesized in vitro).
  • Nucleic acids and proteins that have been “isolated” include nucleic acids and proteins purified by standard purification methods. The term embraces nucleic acids and proteins prepared by recombinant expression in a host cell as well as chemically synthesized molecules.
  • Label Detectable marker or reporter molecules, which can be attached to nucleic acids or proteins, for example probe molecules.
  • Typical labels include fluorophores, radioactive isotopes, ligands, chemiluminescent agents, metal sols and colloids, and enzymes. Methods for labeling and guidance in the choice of labels useful for various purposes are discussed, e.g., in Sambrook et al., in Molecular Cloning: A Laboratory Manual , Cold Spring Harbor Laboratory Press (1989) and Ausubel et al., in Current Protocols in Molecular Biology , Greene Publishing Associates and Wiley-Intersciences (1987).
  • Malignant A term describing cells that have the properties of dysplasia, anaplasia, invasion and metastasis.
  • Membrane a term describing a thin sheet of natural or synthetic material that is porous or otherwise at least partially permeable to biomolecules.
  • Neoplasia Abnormal growth of cells, including benign and malignant neoplasms.
  • Probe A molecule that may bind to or interact with one or more targets (e.g., biological macromolecules or cells).
  • a probe can be any molecule that is used to challenge (“probe,” “assay,” “interrogate” or “screen”) a TMA, MTI, LCM, or other assay, in order to determine the binding, activity, or interaction characteristics of the arrayed target(s) with that probe molecule.
  • probes may be from different and varied molecular classes.
  • Such classes are, for instance, nucleic acids (such as single or double stranded DNA or RNA), oligo- or polypeptides (such as proteins, for instance antibodies, protein fragments including domains or sub-domains, and mutants or variants of naturally occurring proteins), or various types of other potential polypeptide-binding molecules.
  • ligands such as drugs, toxins, venoms, hormones, co-factors, substrates or reaction products of enzymatic reactions or analogs thereof, transition state analogs, minerals, salts, and so forth).
  • probe also encompasses substrates and/or assays systems used to assess the activity of a target within a feature of the array.
  • TMA sections can be assayed for the activity of a protein in one or more features using a probe that is a substrate of that protein (which substrate may contain a label, as discussed herein), or a probe that is a reporter system that interacts with the target protein to produce a detectable signal.
  • a probe molecule for use in probing a TMA is detectable or produces a detectable product.
  • Probes can be detectable based on their inherent characteristics (e.g., immunogenicity, color, fluorescence) or can be rendered detectable by being labeled with an independently detectable tag or label.
  • the tag may be any recognizable feature that is, for example, microscopically distinguishable in shape, size, color, optical density, etc.; differently absorbing or emitting of light; chemically reactive; magnetically or electronically encoded; or in some other way detectable.
  • Specific examples of tags are fluorescent or luminescent molecules that are attached to the probe, or radioactive monomers or molecules that can be added during or after synthesis of the probe molecule.
  • Other tags may be immunogenic sequences (such as epitope tags) or molecules of known binding pairs (such as members of the strept/avidin:biotin system). Additional tags and detection systems are known to those of skill in the art, and can be used in the disclosed methods.
  • probe molecules for instance one protein
  • mixtures of probes will be used, for example, mixtures of two proteins or two nucleic acid molecules.
  • co-applied probes may be labeled with different tags, such that they can be simultaneously detected as different signals (e.g., two fluorophores that emit at different wavelengths or two gold particles of different sizes).
  • one of these co-applied probes will be a control probe (or probe standard), which is designed to hybridize to a known and expected sequence in one or more of the spots on the array.
  • the probe is a heterogeneous mixture, for instance a heterogeneous mixture of nucleic acid molecules or proteins.
  • a probe may be a pool of proteins (for instance, a protein preparation from a cell sample) that can be used as a probe to assay a TMA that contains known proteins (e.g., known antibodies or other proteins), and a signal at a locus on the array interpreted as an indication that the pool contains one or more proteins that interact with the target in that locus (e.g., contains an antigen the target antibody at that locus has affinity for).
  • Probe standard A probe molecule for use as a control in analyzing an array.
  • Positive probe standards include any probes that are known to interact with at least one of the targets of the array.
  • Negative probe standards include any probes that are known not to specifically interact with at least one target of the array.
  • Probe standards that may be used in any one system include molecules of the same class as the test probe that will be used to assay the array. For instance, if the array will be used to examine the interaction of a protein with polypeptides in the array, the probe standard can be a protein or oligo- or polypeptide.
  • a control probe sequence can be designed to hybridize with a so-called “housekeeping” gene.
  • the housekeeping gene is one which is known or suspected to maintain a relatively constant expression level (or at least known to be positively expressed) in a plurality of cells, tissues, or conditions.
  • Many of such “housekeeping” genes are well known in the art; specific examples include histones, ⁇ -actin, or ribosomal subunits (either mRNA encoding for ribosomal proteins or rRNAs). Housekeeping genes can be specific for the cell type being assayed, or the species or Kingdom from which the sample being tested in the array has been produced.
  • a probe standard will be supplied that is unlabeled.
  • Such unlabeled probe standards can be used in a labeling reaction as a standard for comparing labeling efficiency of the test probe that is being studied.
  • labeled probe standards will be provided in the kits.
  • Probing refers to incubating an array with a probe molecule (usually in solution) in order to determine whether the probe molecule will bind to, hybridize or otherwise interact with molecules immobilized on the array. Synonyms include “interrogating,” “challenging,” “screening” and “assaying” an array. Thus, a TMA is said to be “probed” or “assayed” or “challenged” when it is incubated with a probe molecule (such as a labeled or otherwise detectable polypeptide, nucleic acid molecule, or ligand, or a positive, single-stranded and detectable nucleic acid molecule that corresponds to a feature of interest).
  • a probe molecule such as a labeled or otherwise detectable polypeptide, nucleic acid molecule, or ligand, or a positive, single-stranded and detectable nucleic acid molecule that corresponds to a feature of interest.
  • Protein/Polypeptide A biological molecule expressed by a gene or other encoding nucleic acid, and comprised of amino acids. More generally, a polypeptide is any linear chain of amino acids, usually about 50 or more amino acid residues in length, regardless of post-translational modification (e.g., glycosylation or phosphorylation).
  • TMA examples include a plurality of polypeptide samples (targets) placed at addressable locations within an array substrate (e.g., a block of embedding material).
  • the polypeptide at each location can be referred to as a target polypeptide, or target polypeptide sample.
  • polypeptides are deposited into the array in a substantially native configuration, such that at least a portion of the individual polypeptides within the locus is in a native configuration.
  • Such native configuration-polypeptides are capable of binding to or interacting with molecules in solution that are applied to the surface of the array section in a manner that approximates natural intra- or intermolecular interactions.
  • binding of a molecule in solution (for instance, a probe) to a target polypeptide immobilized in an array will be indicative of the likelihood of such interactions in the natural situation (i.e., within a cell).
  • the polypeptides in features of a Tissue Micro Array retain function and therefore can be assayed for an activity.
  • TMA protein analysis using TMA
  • block or section such as formalin fixed, paraffin embedded tissue
  • the features can be quantified as a direct replicate of the block or section.
  • Protein purification Polypeptides for use in the present disclosure can be purified by any of the means known in the art. See, e.g., Guide to Protein Purification , ed. Guide to Protein Purification , ed. Academic Press, San Diego, 1990; and Scopes, Protein Purification: Principles and Practice , Springer Verlag, New York, 1982.
  • Proteomics Global, whole-cell analysis of gene expression at the protein level, yielding a protein profile for a given cell or tissue.
  • the comparison of two protein profiles (proteomes) from cells that have been differently treated (or that are otherwise different, for instance genetically) provides information on the effects the treatment or condition (or other difference) has on protein expression and modification.
  • Subproteomics is analysis of the protein profile of a portion a cell, for instance of an organelle or a protein complex.
  • a mitochondrial proteome is the profile of the protein expression content of a mitochondrion under certain conditions.
  • Proteomic analysis is increasingly being performed using peptide and protein arrays; such arrays are reviewed in Emili and Cagney ( Nat. Biotech. 18:393-397, 2000).
  • a purified nucleic acid preparation is one in which the specified nucleic acid is more enriched than the nucleic acid is in its generative environment, for instance within a cell or in a biochemical reaction chamber.
  • a preparation of substantially pure nucleic acid may be purified such that the desired nucleic acid represents at least 50% of the total nucleic acid content of the preparation.
  • a substantially pure nucleic acid will represent at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, or at least 95% or more of the total nucleic acid content of the preparation.
  • a preparation of substantially pure protein may be purified such that the desired protein represents at least 50% of the total protein content of the preparation.
  • a substantially pure protein will represent at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, or at least 95% or more of the total protein content of the preparation.
  • Bound probe molecules can be stripped from an array, for instance a protein Tissue Micro Array, in order to use the same array for another probe interaction analysis (e.g., to determine the level of a different protein in the arrayed samples, particularly where the arrayed samples contain mixtures of proteins). Any process that will remove substantially all of the first probe molecule from the array, without also significantly removing the immobilized nucleic acid mixtures of the array, can be used.
  • one method for stripping a protein array is by washing it in stripping buffer (e.g., 1 M (NH 4 ) 2 SO 4 and 1 M urea), for instance at room temperature for about 30-60 minutes.
  • one method for stripping an array containing nucleic acids is by boiling it in stripping buffer (e.g., very low or no salt with 0.1% SDS), for instance for about an hour or more.
  • stripping buffer e.g., very low or no salt with 0.1% SDS
  • the stripped array will be equilibrated, for instance in a low stringency wash buffer, prior to incubation with another probe molecule.
  • a sample such as a biological sample, is a sample obtained for example, from a subject.
  • biological samples include all clinical samples useful for detection of cancer in subjects, including, but not limited to, cells, tissues, and bodily fluids, such as: blood; derivatives and fractions of blood, such as serum; biopsied or surgically removed tissue, including tissues that are, for example, unfixed, frozen, fixed in formalin and/or embedded in paraffin; swabs; skin scrapes; urine; sputum; cerebrospinal fluid; prostate fluid; pus; or bone marrow aspirates.
  • a sample includes a solid tumor biopsy obtained from a human subject, such as an EHCC biopsy.
  • a sample includes cells, for example a group of cells collected or archived as part of a tissue section.
  • Samples may come from human or non-human animals, as well as in vitro grown cell lines or xenografts.
  • Subject Living, multicellular vertebrate organisms, a group that includes both human and veterinary subjects, for example, mammals, birds, and primates.
  • Target As used herein, individual molecules, cells, tissue sections or mixtures that are placed onto a TMA, TMI, LCM, or other platform for analysis, are referred to as targets. Targets on a single array can be derived from one to several thousand different samples, such as cell or tissue types (more generally, from a plurality of specimens). In certain embodiments of the arrays and methods described herein, the target feature on the array contains a heterogeneous mixture of molecules that proportionately reflects the levels of the starting (source) material from which the molecules are derived; such arrays can be used to comparatively examine the level of constituents in an array feature.
  • the features of the array contain mRNA or mRNA-derived molecules (e.g., aRNA, cRNA or cDNA) that are present in proportionate amounts to the nucleic acids they represent in the starting sample (e.g., tissue) from which the mRNA was extracted to generate the feature.
  • some arrays will include features that contain heterogeneous mixtures of proteins that reflect the levels (e.g., proportionate levels) of those proteins in a starting material, such as a tissue sample.
  • a target on the array is discrete, in that signals from that target can be distinguished from signals of neighboring targets, either by the naked eye (macroarrays) or by scanning or reading by a piece of equipment or with the assistance of a microscope (microarrays).
  • Tissue Microarrays An array of samples, such as biological samples, placed into a block of substrate (such as embedding compound), which loaded block is then sliced (sectioned) to produce a cross-section of the biological sample, each containing a portion of the sample in the block. The samples “freeze” into the block of substrate, such that the loaded block can be sectioned and will maintain the portions of sample in addressable locations that correlate to the locations of the samples in the loaded block.
  • TMA include protein Tissue Micro Arrays (in which the samples contain one or more known or unknown proteins), and nucleic acid Tissue Micro Arrays (in which the samples contain one or more known or unknown nucleic acids). Additional examples of Tissue Microarrays are discussed herein.
  • Tissue Microarrays are constructed as a block containing substantially columnar samples contained in wells in the block. Once one or more samples are loaded into wells in the block, it can be sliced (sectioned) to provide a plurality of identical or substantially identical individual arrays. The individual arrays can be used for parallel analysis of the same set of features, for instance with different probes or under different conditions.
  • the wells in the block may be formed perpendicular to the surface from which sections are removed.
  • the columns may be non-parallel but will vary in a predictable relationship to one another, such that the position at which each column intersects a section can be predicted.
  • Tissue Micro Array substrate itself is essentially immaterial, though it is usually substantially flat on at least one side and may be rectangular or square in general shape.
  • Tissue Micro Arrays are constructed as blocks that contain a biological sample, for example a tissue sample, in which the biological sample in the block is transferred to a stack of replicate membranes, which can be probed using standard immunohistochemistry techniques.
  • the block provides a level of histomorphological correlation with the original biological sample in the block.
  • Tumor A neoplasm that may be either malignant or non-malignant.
  • Tumors of the same tissue type refers to primary tumors originating in a particular organ (such as breast, prostate, bladder or lung). Tumors of the same tissue type may be divided into tumors of different sub-types (a classic example being bronchogenic carcinomas (lung tumors), which can be an adenocarcinoma, small cell, squamous cell, or large cell tumor). Recurrent and metastatic tumors are also contemplated.
  • TNM Malignant Tumors
  • UICC International Union against Cancer
  • AJCC American Joint Committee on Cancer
  • a watchful-waiting protocol is a wait-and-see clinical approach for the treatment of disease, for example, prostate cancer.
  • the subject may get better (or not get worse) without treatment; if the condition worsens, the physician managing the subjects' health will decide what to do next, for example a radical prostatectomy.
  • the methods disclosed herein require the detection of at least one cancer associated protein.
  • the methods disclosed herein require the detection of at least two, three, four, five, or more, cancer associated proteins.
  • the cancer associated protein comprises a cell signaling or growth factor pathway protein, or another cancer associated molecule such as a cytokeratin, a protein associated with cytoskelatin or a protein associated with localization of another protein (such as adaptors and so forth), have utility in cancer as well.
  • the methods include detecting the presence of at least one cancer associated protein in a biological sample, such a tissue section or biopsy. In some embodiments, identifying the presence of the at least one cancer associated proteins in a sample determines if a particular therapeutic regime is successful as a means to increase the relative survival rate (and more generally, improve the prognosis) of a subject with a cancer.
  • identifying the presence of at least one cancer associated protein associated with a cell signaling pathway or growth factor pathway in a sample can determine if a particular therapeutic regime is successful as a means to increase the prognosis or relative survival rate of a subject with a cancer.
  • the methods disclosed herein can be used to determine if preventative treatment should be administered to a subject at risk for developing cancer, or if a treatment should be administered to a subject to prevent the progression of existing pathological structures, such as from early stage to a more advanced stage of cancer.
  • the methods disclosed herein can also be used to confirm a diagnosis of cancer in the subject.
  • Methods for determining cancer survival probability of a subject with cancer are provided herein.
  • methods for predicting relative survival rate for a subject with a solid tumor, such as a carcinoma are disclosed.
  • the methods disclosed can also be used to detection cancer in a subject such as, but not limited to, solid tumors, such as carcinomas of breast, lung, prostate, colon, gastric, liver, thyroid, kidney, bile duct and renal tissue.
  • the methods of the instant disclosure can be used to detect the presence of EHCC in a subject.
  • the methods disclosed herein can be used to detect the presence of a hematopoietic cancer such as lymphoma, leukemia or multiple myeloma in a biological sample.
  • a hematopoietic cancer such as lymphoma, leukemia or multiple myeloma in a biological sample.
  • methods for determining relative survival rates for a subject with a hematopoietic cancer such as lymphoma, leukemia or multiple myeloma.
  • the methods disclosed can also be used to determine the risk of developing cancer, such as, but not limited to carcinoma.
  • the methods are also useful as a prognostic tool to evaluate subjects with cancer prior to treatment and as a means for determining a therapeutic regimen for a subject that is anticipated to increase the relative survival rate of the subject with cancer.
  • the methods are useful not only in determining risk, but for pathological confirmation of cancer associated proteins.
  • the detection of cancer associated proteins can be correlated with a histomorphological structure in the original tissue block sample.
  • the methods disclosed herein can also be used to detect a cancer or determine the risk of developing a cancer based on the protein expression profile of the biological sample tested.
  • the methods include obtaining a sample from a subject, identifying at least two cancer associated proteins in the sample; quantifying the content of the two cancer associated proteins; normalizing the content of the two cancer associated proteins to obtain a normalized value for each cancer associated protein and comparing the normalized value of the first cancer associated protein with the normalized value of the second cancer associated protein to obtain a biomarker indicator, and correlating the biomarker indicator with survival probability of the subject with the cancer.
  • the instant disclosure identifies a survival-based cancer biomarker comprising at least two cancer associated proteins, wherein the at least two cancer associated proteins are proteins from a cell signaling pathway associated with the cancer, and wherein the two cancer associated proteins are used to obtain a biomarker indicator that can be used to determine relative survival rates of a subject with cancer.
  • the instant application discloses a method of detecting the presence of a cancer in a subject, the methods generally comprise determining the level of a first and second cancer associated protein and normalizing the presence of the cancer associate protein to obtain a biomarker indicator that correlates with the present of cancer.
  • the methods as disclosed herein include selecting a subject in need of detecting the presence of the cancer associated protein, and obtaining a sample including the cancer associated protein from this subject.
  • a subject can be selected who is suspected to have a cancer, such as breast, colon, stomach (gastric), cervical, brain, head and neck, prostate, biliary tract or lung cancer.
  • a subject can be selected that is symptomatic with a cancer.
  • the subject can be a subject who has been diagnosed with a carcinoma, such as, but not limited to bile duct carcinoma, EHCC, lung cancer (such as non-small cell lung cancer; NSCLC), and gastric cancer.
  • the subject's risk for progressing to another stage of cancer can be determined.
  • a subject with cancer can also be evaluated to determine if a therapeutic regimen is appropriate for the subject using methods disclosed herein.
  • a subject of interest can also be selected to determine if preventative treatment such as, watchful-waiting protocols, should be undertaken.
  • this disclosure provides a method of determining survival probability for a subject with cancer.
  • the method is directed to calculating the survival probability for a subject with a solid tumor, using whole tissue sections, tissue microarrays, and arrays of minute tissue sections.
  • the method is directed to determining survival probability for a subject using solidified cell samples, such as leukemia cells frozen for example, in an embedding compound.
  • lung cancer such as non-small cell lung cancer.
  • “old fashion” immunohistochemistry and automated image analyses were applied to generate a continuous variable to reflect the staining for the analyte of interest.
  • four selected individual analytes p-mTOR, p-Akt, p-MAPK, and EGFR
  • FIG. 8B the ratio-metric approach was applied,
  • biomarkers described herein are predictive/diagnostic/prognostic because one (or more) of the component antigens increases or decreases. However, provided biomarkers illustrate that the relative amounts of the component antigens are what is most relevant (and significant), in that one or another of the component antigens can be altered (up or down) without the other antigen(s) altering, and the data still reflects that the biomarker is predictive. As clearly illustrated herein with the HER2/HER3 system, the balance of the components in the calculated biomarker is key. In this example, either an increase in HER2 or a decrease in HER3 alters the biological status/disease state/prognosis of the subject with the same outcome.
  • the stoichiometry of the component antigens strongly influences the calculated biomarker—and the biological outcome.
  • This system can be viewed as a flow moving through or in a pathway—it is important both that the elements of the pathway are present and also that there is sufficient signaling capacity within the entire pathway for the signal (the biological effect) to be felt.
  • One signal in excess in the pathway may not matter, where the pathway is at or beyond capacity.
  • the methods and other embodiments provided herein, though exemplified in the context of various cancers, are applicable to other diseases and conditions.
  • the illustrated cancer approach can be applied to any tissue-based disease where a cell/tissue-based analysis is feasible. Although the analysis does not require a cell-by-cell analysis, in many embodiments it is applied to a cell-type, within a tissue. Examples of measurements of non-malignant (that is, not linked to cancer) processes include cirrhosis of the liver, renal disease (glomerular or tubular) and other processes that will be recognizable by one of ordinary skill
  • a cancer associated protein is a protein known to be associated with cancer or a protein that can be determined (via methods known or routinely developed in the art) to be associated with a specific type or form of cancer.
  • high levels of expression (upregulation) of CA 125 in ovarian tissue samples are commonly associated with an increased risk for the development of ovarian cancer.
  • cancer-specific markers or “tissue-specific markers” are often linked with a specific form of cancer, or location of cancer, and are therefore considered cancer associated proteins as defined herein.
  • tumor antigen refers to a substance (e.g., a protein) produced by a tumor cell that is not typically produced (and therefore associated) with a normal, non-cancerous cell. More distantly “associated” proteins are also considered, including for instance cytokeratins, other structural proteins, proteins that interact therewith, and so forth. The success of ratio based markers, as illustrated herein, is detection of aberrant expression of normally expressed proteins.
  • proteins that were not previously characterized with a cancer may be detected through various techniques known or developed in the art, to be associated with a specific type or form of cancer, these types of cancer associated proteins are often termed “tumor associated antigens” and include mutated or aberrant proteins that are produced as a result of the presence of the cancer.
  • a previously characterized cellular protein may be found to be directly (or indirectly) impacted by a cell signaling pathway that in turn, activates or positively influences the development or progression of cancer, such as proteins found in cellular survival, apoptosis and growth factor pathways (e.g., 4E-BP1) and is therefore a cancer associated protein as defined herein. Specific examples of cancer associated proteins are discussed in more detail below.
  • two or more cancer associated proteins are identified and the relative contents (protein expression levels) are compared to provide a biomarker indicator of disease.
  • overexpression of HER2 in normal breast tissue may be considered a risk factor for the development of breast cancer.
  • upregulation of a second protein in the same breast tissue sample, such as survivin (BIRC5), a protein associated with inhibition of apoptosis may provide significant accumulative evidence in conjunction with the first cancer associated protein to indicate that there is an elevated risk for uncontrolled cellular proliferation and the development of a malignant neoplasm in the sample.
  • BIRC5 survivin
  • the identification and quantitation of two or more cancer associated proteins in a sample using the methods disclosed herein can be used to determine the presence of cancer in a subject and, additionally, the relative survival rate of a subject with cancer.
  • the two or more cancer associated proteins may or may not be directly linked to one another, for example, by protein structure or function.
  • the first cancer associated protein may be a protein from a growth factor pathway
  • the second cancer associated protein may be directed to the expression of a gene product related to nutrient metabolism.
  • the two or more cancer associated proteins may be directly linked to one another.
  • the first cancer associated protein may be related to overexpression of a cell signaling protein, such as mTOR, while the second cancer associated protein may be a protein from the same cell signaling pathway, such as AKT.
  • the two or more cancer associated proteins may be indirectly linked to one another.
  • the first cancer associated protein may be a cell signaling protein, such as AKT
  • the second cancer associated protein is a cellular proliferation marker, such as Ki-67.
  • the first cancer associated protein may be remotely upstream or remotely downstream from the second cancer associated protein. In another embodiment, the first cancer associated protein may be directly upstream or directly downstream from the second cancer associated protein.
  • the two or more cancer associated proteins may be related in terms of protein function.
  • the first cancer associated protein may be directed to the expression of a phosphorylated (activated) protein
  • the second cancer associated protein is also directed to expression of a phosphorylated protein, such as pERK1/2.
  • the two or more cancer associated proteins are proteins that are known, or can be determined by methods known to one of ordinary skill in the art, to be inter-connected.
  • the two or more cancer associated proteins are found within the same cell signaling or growth factor pathway.
  • the two or more cancer associated proteins are known or can be determined by one of ordinary skill in the art to “cross-talk”.
  • cross-talk refers to the phenomenon that signal components in signal transduction can be shared between different signal pathways and responses to a signal inducing condition (e.g., stress) can activate multiple responses in a cell, tissue or organism.
  • the term “cross-talk” refers to the mechanism by which activated signal molecules in a primary signal transduction pathway can regulate or influence signaling molecules in another primary signal transduction pathway.
  • the cancer associated protein comprises a tumor antigen or tumor associated antigen.
  • the cancer associated protein is present in a solid tumor.
  • the cancer associated protein is present in a carcinoma selected from the group consisting of breast, lung, prostate, colon, liver, thyroid, kidney, and bile duct carcinoma.
  • the cancer associated protein is observed in a group of cancers consisting of, but not limited to adrenal tumor, bile duct cancer, bladder cancer, bone cancer, brain tumor, breast cancer, cardiac sarcoma, cervical cancer, colorectal cancer, endometrial cancer, esophageal cancer, germ cell cancer, gynecologic cancer, head and neck cancer, hepatoblastoma, renal cancer, laryngeal cancer, leukemia, liver cancer, lung cancer; lymphoma, melanoma, multiple myeloma, neuroblastoma, oral cancer, ovarian cancer, pancreatic cancer, parathyroid cancer, pituitary tumor, prostate cancer, retinoblastoma, rhabdomtosarcoma, skin cancer (non-melanoma), small bowel, stomach (gastric) cancer, testicular cancer, thyroid cancer, uterine cancer, vaginal cancer, vulvar cancer, and Wilms' tumor.
  • the cancer associated protein is observed in a
  • the cancer associated protein is used to determine cancer survival probability for a subject with a cancer by identifying at least two cancer associated proteins in a sample, quantifying the at least two cancer associate proteins in the sample, normalizing the content of the two cancer associated proteins and comparing the normalized levels of the first and second cancer associated proteins to obtain a biomarker indicator and correlating the biomarker indicator with survival probability of the subject with the cancer.
  • the cancer associated protein is used to detect the presence of a cancer in a subject by determining the levels of at least two cancer associated proteins in a sample, normalizing the content of the two cancer associated proteins and comparing the normalized levels of the first and second cancer associated proteins with the level of the first and second cancer associated proteins in a normal non-cancerous subject, in order to identify the presence of the cancer in the subject.
  • the cancer associated proteins can be used to detect the presence of a solid tumor, such as a carcinoma in a subject.
  • the two cancer associated proteins can be used to detect the presence of a carcinoma in a subject, such as a carcinoma selected from the group consisting of, but not limited to, breast, lung, prostate, colon, stomach (gastric), ovarian, cervical, brain, skin, esophageal, biliary tract, and extrahepatic cholangiocarinoma (EHCC).
  • a carcinoma selected from the group consisting of, but not limited to, breast, lung, prostate, colon, stomach (gastric), ovarian, cervical, brain, skin, esophageal, biliary tract, and extrahepatic cholangiocarinoma (EHCC).
  • EHCC extrahepatic cholangiocarinoma
  • the two or more cancer associated proteins are correlated with the presence of a blood-borne cancer.
  • the two or more cancer associated proteins are specific for the detection of leukemia, multiple myeloma or lymphoma.
  • the two or more cancer associated proteins are specific for the identification or detection of leukemia in a subject.
  • the two or more cancer associated proteins are tumor antigens selected from the group consisting of, but not limited to, AKT; p-AKT; Blood Group Tn Antigen, CA150; CA19-9; CA50; CAB39L; CD22; CD24; CD63; CD66a+CD66c+CD66d+CD66e; CTAG1B; CTAG2; Carcino Embryonic Antigen (CEA); EBAG9; EGFR; FLJ14868; FMNL1; GAGE1; GPA33; Ganglioside OAcGD3; Heparanase 1; HER2; HER3; JAKMIP2; LRIG3; Lung carcinoma Cluster 2; M2A Oncofetal Antigen, MAGE 1; MAGEA10; MAGEA11; MAGEA12; MAGEA2; MAGEA4; MAGEB1; MAGEB2; MAGEB3; MAGEB4; MAGEB6; MAGEC1; MAGEE1; MAGEH1;
  • the cancer associated proteins are tumor associated antigens selected from the group consisting of, but not limited to, 5T4; AKT; p-AKT; ACRBP; Blood Group Tn Antigen; CD164; CD20; CTHRC1; ErbB 2; FATE1; HER2; HER3; GPNMB; Galectin 8; HORMAD1; LYK5; MAGEA6; MAGEA8; MAGEA9; MAGEB18; MAGED2; MAPK; p-MAPK; mTOR; p-mTOR; MUC1; MUC2; MelanA; Melanoma gp100; NYS48; PARP9; PATE; Prostein; PTEN; SDCCAG8; SEPT1; SLC45A2; TBC1D2; TRP1; XAGE1; and tumor associated antigens of epithelial origin.
  • 5T4 AKT
  • p-AKT ACRBP
  • ACRBP Blood Group Tn Antigen
  • CD164 CD20
  • the two or more cancer associated proteins comprise two or more proteins from the PI3K, AKT or ERK1/2 signaling pathway.
  • the two or more cancer associated proteins are selected from the group consisting of, but not limited to, 4E-BP1, phosphorylated 4e-BP1 (p-4E-BP1), eIF-4E, phosphorylated eIF-4E (p-eIF-4E), AKT, phosphorylated AKT (pAKT), Erk1/2, Hsp27, Hsp 90, Tc11, Grb10, Ft1, Jip1, Posh, mTOR, phosphorylated mTOR(p-mTOR), periostin, and PTEN.
  • the cancer associated protein as defined herein refers to a tumor antigen or a tumor associated antigen.
  • the tumor antigen or tumor associated antigen is specific for the identification or detection of a carcinoma selected from the group consisting of, but not limited to, breast, lung, prostate, colon, ovarian, cervical, brain, skin, esophageal, biliary tract and extrahepatic cholangiocarinomas.
  • the cancer associated protein is specific for the detection or identification of EHCC.
  • the cancer associated protein is p-AKT, p-mTOR p-MAPK, EGFR, Her2, Her3 or PTEN.
  • the cancer associated protein comprises a cell signaling or growth factor pathway protein.
  • the cancer associated protein is selected from the group of signaling pathways consisting of, but not limited to, PI3K, AKT, PTEN, ERK1/2, Wnt, and TGF- ⁇ , as well documented in the art, see for example, Strimpakos et al. Cancer Treat Rev; Nov. 13, 2008; or Katoh and Katoh, Int J Mol. Med. 2008; 14:4042-5.
  • the cancer associated protein comprises a protein of the p53 pathway.
  • the cancer associated protein of the p53 pathway includes but is not limited to, 53BP1, ALDH11, BRCC45, BNIP3L, CDKN2A, DRAM, DBC1, DDB2, ING1, JAB, MDM2, OVCA1, PARC, PBK, PIG3, PRMT4, and protein products of p21, p53, p63 and p73.
  • the cancer associated protein comprises a growth factor or growth factor receptor protein.
  • the cancer associated protein is selected from the group consisting of, but not limited to, ALK, EGFR, Erb 3, GCSF receptors, Kit (c), PDGF receptors, Pan Trk, Raf 1, Ret, TIE, Trk A, Trk B, Trk C, VEGF receptor 1, VEGF receptor 2, VEGF receptor 3, and Xmrk.
  • the cancer associated protein comprises an EGF protein, selected from the group consisting of, but not limited to, ACK1, EGF, EPS8; Erb 2, Erb 3, Erb 4, TMEFF2, and Xmrk.
  • the two or more cancer associated proteins comprise a FGF, PDGF, TGF, THF or VEGF protein, as well known in the art (Cao et al., J. Mol. Med; 2008; (7) 785-9; Katoh and Katoh, Clinical Cancer Research 2007; 14: 4042-5; Jiang and Hui, Developmental Cell 2008; 6:801-12; and Antonescu, Modern Pathology 2008; 21:suppl. 2:S31-36).
  • the cancer associated protein is a signal transducer protein.
  • the signal transducer protein is selected from the group consisting of, but not limited to, A RAF, ASPP1, ax1, B Raf, CBLB, CD45R, ELMO, ERAS, FES, JAK1, JAK2, JAK3, JNK1, JNK2, KAT13A, NDRG1, PI3K, PIM, Ras (p21), SRC1, Styk1, and c Ab1.
  • phospho-specific antibodies can be used to detect phosphorylated proteins in a sample (i.e., the detection of pAKT, p-mTOR, p-MAPK, or pERK1/2). Signal intensity from detection of the phosphorylated proteins can be quantified to obtain intensity values for each detected phosphorylated protein.
  • the cancer associated protein comprises a phosphorylated cancer associated protein.
  • the phosphorylated cancer associated protein is selected from the group consisting of, but not limited to, pAKT, p-mTOR, p-4E-BP1, p-eIF-4E (eukaryotic initiation factor 4E), pERK1/2, pHER2 (human epidermal growth factor receptor), p70S6K1 (phosphorylated ribosomal protein S6 kinase 1), phosphorylated ribosomal protein S6 (S6), p-glycogen synthase kinase 3(3, pMAPK, and pEGFR.
  • the cancer associated protein comprises a protein associated with the AKT signaling pathway.
  • the cancer associate protein associate with the AKT signaling pathway includes, but is not limited to, 4E-BP1, phosphorylated 4e-BP1 (p-4E-BP1), eIF-4E, phosphorylated eIF-4E (p-eIF-4E), AKT, phosphorylated AKT (pAKT), Erk1/2, Hsp27, Hsp 90, Tc11, Grb10, Ft1, Jip1, Posh, mTOR, phosphorylated mTOR(p-mTOR), periostin and PTEN.
  • the phosphorylated cancer associated protein is detected by phosphor-specific antibodies including, but not limited to, pAKT antibodies, p-mTOR antibodies, p-4E-BP1 antibodies, p-eIF-4E antibodies, pERK1/2 antibodies, p-glycogen synthase kinase 3 ⁇ antibodies, pMAPK antibodies, and pEGFR antibodies.
  • phosphor-specific antibodies including, but not limited to, pAKT antibodies, p-mTOR antibodies, p-4E-BP1 antibodies, p-eIF-4E antibodies, pERK1/2 antibodies, p-glycogen synthase kinase 3 ⁇ antibodies, pMAPK antibodies, and pEGFR antibodies.
  • the cancer associated protein is a tumor antigen or a tumor associated antigen selected from the group of factors consisting of, but not limited to, EGF, FGF, PDGF, TGF, TNF, and VEGF.
  • the cancer associated protein comprises a protein from a cellular apoptosis pathway.
  • the cancer associated protein is a protein of the p53 pathway.
  • FIGS. 6A-6D that were presented in the provisional application from which this filing claims priority (and which are incorporated herein by reference) are schematic drawings of a EGF, AKT, p53 and cellular apoptosis pathway as contemplated by the instant disclosure and identify several cancer associated proteins that can be utilized by the methods disclosed herein.
  • the cancer associated protein includes a cancer associated protein from the group of proteins consisting of, but not limited to, AKT, p-AKT, PTEN, PI3K, PIP2, PIP3 and Ras.
  • the cancer associated protein comprises a tyrosine kinase receptor, such as AKT.
  • the cancer associate protein is a protein selected from the signal transduction pathways selected from AKT, HER2, and EGFR.
  • the cancer associated protein is selected from the group consisting of stathmin, total prostate specific antigen (tPSA), human kallikrein 2 (hk2), type 1 insulin-like growth factor receptor, NF-KB, hypoxia inducible factor-1, protein kinase A type I, vascular endothelial growth factor, 5-lipoxygenase, 12-lipoxygenase, angiotensin II receptor type 1, bradykinin receptor type 1, interleukin-6, ras, MDM2, bcl-2/bclxL, vitamin D receptor, estrogen receptor- ⁇ and peroxisome proliferator-activated receptors (PPARs).
  • stathmin total prostate specific antigen
  • hk2 human kallikrein 2
  • type 1 insulin-like growth factor receptor NF-KB
  • hypoxia inducible factor-1 protein kinase A type I
  • vascular endothelial growth factor 5-lipoxygenase
  • 12-lipoxygenase angiotensin II receptor
  • the cancer associated protein expression profiles may be used to check how a subject is responding to treatment. For example, a decrease or return to normal level of protein expression by the cancer associated protein may indicate that the cancer is responding to therapy (wherein a decrease in the cancer associated protein expression is linked to a decreased incidence of cancer), whereas an increase in protein expression of the cancer associated protein may indicate (wherein an increase in cancer associated protein expression is linked to an increased incidence of cancer) that the subject is not responding to treatment.
  • cancer associated proteins may also be used to check for recurrence. If a cancer associated protein is used to determine whether a treatment is working or if there is recurrence, the cancer associated protein levels can be measured over a period of time to see if the levels are steady-state, increasing or decreasing.
  • cancer associated proteins levels may be checked or monitored at the time of diagnosis; before, during, and after therapy; and then periodically to monitor for recurrence.
  • cancer associated proteins that are currently unknown might reasonably be incorporated into the above lists of cancer associated proteins without undue experimentation.
  • a suspected cancer associated protein can be tested through the use of various signal modulating agents, in concentrations that can feasibly be achieved and maintained clinically, on human cancer cell lines.
  • the suppression, reversal or inhibition of tested cancer associated proteins, especially those associated with cell signaling pathways, which appear promising, can then be tested in animal models, and ultimately tested in a clinical environment.
  • the cancer associated protein is detected in a sample using an antibody specific for the detection of the cancer associated protein and a porous membrane to separate the cancer associated protein from the remainder of the sample.
  • the cancer associated protein is detected in a sample comprising a formal fixed, paraffin embedded tissue block, fresh frozen tissue biopsy, solidified cells, serum or other biological fluid. Specific examples of the types of samples that can be tested using the methods disclosed herein will be discussed in detail below. Additionally, the types of probes or detector molecules that can be used by the methods disclosed herein to detect or identify two or more cancer associated proteins of the instant application will also be discussed in more detail below.
  • the probe to be used in the methods disclosed herein is specific for the detection of a cancer associated protein.
  • the probe is an antibody with an affinity for the cancer associated protein.
  • the antibody is specific for the detection of a cancer associated protein from a solid tumor, such as a carcinoma.
  • the probe is an antibody with an affinity for the detection of the cancer associated protein
  • the cancer associated protein is selected from the group of cancers consisting of, but not limited to, adrenal tumor, bile duct cancer, bladder cancer, bone cancer, brain tumor, breast cancer, cardiac sarcoma, cervical cancer, colorectal cancer, endometrial cancer, esophageal cancer, germ cell cancer, gynecologic cancer, head and neck cancer, hepatoblastoma, renal cancer, laryngeal cancer, leukemia, liver cancer, lung cancer; lymphoma, melanoma, multiple myeloma, neuroblastoma, oral cancer, ovarian cancer, pancreatic cancer, parathyroid cancer, pituitary tumor, prostate cancer, retinoblastoma, rhabdomtosarcoma, skin cancer (non-melanoma), small bowel cancer, stomach (gastric) cancer, testicular cancer, thyroid cancer, uterine cancer, vaginal
  • the probe is specific for the identification of a cancer associated protein associated with a carcinoma, selected from the group consisting of, but not limited to, breast, lung, prostate, colon, stomach (gastric), ovarian, cervical, brain, skin, esophageal, biliary tract, and EHCC.
  • the probe is specific for the detection of a cancer associated protein associated with EHCC, lung cancer, or gastric cancer.
  • the probe is specific for the identification of a cancer associated protein of a blood-borne cancer. In a further embodiment, the probe is specific for the detection of a cancer associated protein of leukemia, multiple myeloma or lymphoma. In a particular embodiment, the probe is specific for the detection of a leukemia cancer associated protein.
  • the probe is specific for the detection of an individual cancer associated protein, wherein the individual cancer associated protein is a tumor antigen selected from the group consisting of, but is not limited to, AKT; p-AKT; Blood Group Tn Antigen, CA150; CA19-9; CA50; CAB39L; CD22; CD24; CD63; CD66a+CD66c+CD66d+CD66c; CTAG1B; CTAG2; Carcino Embryonic Antigen (CEA); EBAG9; EGFR; FLJ14868; FMNL1; GAGE1; GPA33; Ganglioside OAcGD3; Heparanase 1; HER2; HER3; JAKMIP2; LRIG3; Lung carcinoma Cluster 2; M2A Oncofetal Antigen, MAGE 1; MAGEA10; MAGEA11; MAGEA12; MAGEA2; MAGEA4; MAGEB1 MAGEB2; MAGEB3; MAGEB4; MAGEB6;
  • the probe is specific for the detection of an individual cancer associated protein, wherein the individual cancer associated protein is a tumor associated antigen selected from the group consisting of, but not limited to, 5T4; AKT; p-AKT; ACRBP; Blood Group Tn Antigen; CD164; CD20; CTHRC1; ErbB 2; FATE1; HER2; HER3; GPNMB; Galectin 8; HORMAD1; LYK5; MAGEA6; MAGEA8; MAGEA9; MAGEB18; MAGED2; MAPK; p-MAPK; mTOR; p-mTOR; MUC1; MUC2; MelanA; Melanoma gp100; NYS48; PARP9; PATE; Prostein; PTEN; SDCCAG8; SEPT1; SLC45A2; TBC1D2; TRP1; XAGE1; and tumor associated antigens of epithelial origin.
  • the individual cancer associated protein is a tumor associated antigen selected from
  • the probe is specific for the detection of a cancer associated protein, wherein the cancer associated protein is a protein of the PI3K, AKT or ERK1/2 signaling pathway.
  • the probe is specific for the identification of a cancer associated protein from a cell signaling or growth factor pathway.
  • the probe is specific for the identification of a cancer associated protein from a cell signaling pathway selected from the group consisting of, but not limited to, PI3K, AKT, PTEN, ERK1/2, Wnt, and TGF- ⁇ .
  • the AKT signaling pathway is known to play an important role in the signaling pathways in response to growth factors and serves to regulate several cellular functions including nutrient metabolism, cell growth, apoptosis and survival.
  • AKT is a serine/threonine kinase that belongs to a much larger (AGC) family of super protein kinases. Deregulation of AKT has been frequently associated with human disease including cancer. For all AGC family kinases, phosphorylation of the serine and threonine residue is necessary for full activation of the kinase.
  • the probe is an antibody that is specific for the detection of a cancer associated protein of the AKT signaling pathway.
  • identification of a cancer associated protein by the probe can be used as an indicator of activation of the cancer associate protein in the AKT signaling pathway.
  • the probe is specific for the detection of phosphorylated AKT, which in turn is a measure of activated AKT. Because deregulations of the AKT signaling pathway are associated with disease states such as cancer, the detection of an activated AKT cancer associate protein may be an indicator of a cancerous disease state.
  • the probe is specific for the detection and identification of a cancer associated protein selected from the group consisting of, but not limited to, 4E-BP1, phosphorylated 4e-BP1 (p-4E-BP1), eIF-4E, phosphorylated eIF-4E (p-eIF-4E), AKT, phosphorylated AKT (pAKT), Erk1/2, Hsp27, Hsp 90, Tc11, Grb10, Ft1, Jip1, Posh, mTOR, phosphorylated mTOR(p-mTOR), periostin, and PTEN.
  • 4E-BP1 phosphorylated 4e-BP1
  • eIF-4E phosphorylated eIF-4E
  • p-eIF-4E phosphorylated eIF-4E
  • AKT phosphorylated AKT
  • pAKT phosphorylated AKT
  • the probe used to detect the cancer associated protein is a tumor antigen antibody or a tumor associated antigen antibody.
  • the probe is a tumor antigen or a tumor associated antigen antibody associated with a carcinoma, including, but not limited to, breast, lung, prostate, colon, stomach (gastric), ovarian, cervical, brain, skin, esophageal, biliary tract and extrahepatic cholangiocarinomas.
  • the probe is a tumor antigen or tumor associated antigen antibody associated with EHCC.
  • the probe is an antibody specific for the detection of AKT, p-AKT, MAPK, p-MAPK, EGFR, Her2, Her3, mTOR, p-mTOR, or PTEN.
  • the probe used to detect the cancer associate protein is a tumor antigen antibody including, but not limited to, AKT antibodies, p-AKT antibodies, CA150 antibodies, CA19-9 antibodies, CA50 antibodies, CAB39L antibodies, CD22 antibodies, CD24 antibodies, CD63 antibodies, CD66 antibodies; CTAG1B antibodies, CTAG2 antibodies, Carcino Embryonic Antigen (CEA) antibodies, EBAG9 antibodies, FLJ14868 antibodies, FMNL1 antibodies, GAGE1 antibodies, GPA33 antibodies, Ganglioside OAcGD3 antibodies, Heparanase 1 antibodies, HER2 antibodies, HER 3 antibodies, JAKMIP2 antibodies, LRIG3 antibodies, Lung carcinoma cluster 2 antibodies, MAGE 1 antibodies, MAGEA10 antibodies, MAGEA11 antibodies, MAGEA12 antibodies, MAGEA2 antibodies, MAGEA4 antibodies, MAGEB1 antibodies, MAGEB2 antibodies, MAGEB3 antibodies, MAGEB4 antibodies, MAGEB6 antibodies, MAGEC1 antibodies, MAGEE1 antibodies, MAGEH
  • the cancer associated proteins are tumor associated antigens selected from the group consisting of, but not limited to, 5T4 antibodies, ACRBP antibodies, CD164 antibodies, CD20 antibodies, CTHRC1 antibodies, ErbB 2 antibodies, FATE1 antibodies, GPNMB antibodies, Galectin 8 antibodies, HORMAD1 antibodies, LYK5 antibodies, MAGEA6 antibodies, MAGEA8 antibodies, MAGEA9 antibodies, MAGEB18 antibodies, MAGED2 antibodies, MUC1 antibodies, MUC2 antibodies, MelanA antibodies, Melanoma gp100 antibodies, NYS48 antibodies, PARP9 antibodies, PATE antibodies, Prostein antibodies, SDCCAG8 antibodies, SEPT1 antibodies, SLC45A2 antibodies, TBC1D2 antibodies, TRP1 antibodies, XAGE1 antibodies, antibodies to tumor associated antigens of epithelial origin, and antibodies to any of the other (cancer) antigens referenced herein or recognized in the art.
  • tumor associated antigens selected from the group consisting of, but not limited to, 5T4 antibodies,
  • the probe in the methods discloses herein is an antibody with an affinity for the cancer associated protein.
  • the probe used to detect the cancer associated protein comprises an antibody of a cell signaling pathway.
  • the probe comprises an antibody of the AKT cell signaling pathway, including, but not limited to, AKT antibodies, phospho-AKT antibodies (e.g., antibodies specific for Phospho T308-AKT or Phospho-S473-AKT), mTOR antibodies and phospho-mTOR antibodies.
  • the probe used is specific for the detection of a cancer associated protein of the P′I′EN cell signaling pathway, including, but not limited to, PTEN antibodies and TPTE antibodies.
  • the probe used is an antibody specific for the detection of a cancer associated protein of the Wnt cell signaling pathway, including, but not limited to, APC antibodies, LEF1 antibodies, PTCH antibodies, Sonic Hedgehog antibodies, WISP2 antibodies; WNT2 antibodies; WNT2B antibodies; WNT4 antibodies; Wnt1 antibodies; Wnt10a antibodies; Wnt5a antibodies; Wnt6 antibodies; and Wnt8a antibodies.
  • the probe used is specific for the detection of a cancer associated protein of the p53 pathway.
  • the probe is an antibody of the p53 pathway, including, but not limited to, 53BP1 antibodies, ALDH11 antibodies, BRCC45 antibodies, BNIP3L antibodies, CDKN2A antibodies, DRAM antibodies, DBC1 antibodies, DDB2 antibodies, ING1 antibodies, JAB antibodies, MDM2 antibodies, OVCA1 antibodies, PARC antibodies, PBK antibodies, PIG3 antibodies, PRMT4 antibodies, p21 antibodies, p53 antibodies, p63 antibodies, and p73 antibodies.
  • the antibody used in the methods disclosed herein to detect the cancer associated protein comprises a growth factor or growth factor receptor antibody from a growth factor pathway.
  • the antibody is a growth factor receptor antibody including, but not limited to, ALK antibodies.
  • the antibody used to detect the cancer associated protein is an EGF antibody.
  • the antibody used to detect the cancer associated protein is a EGF antibody selected from the group, but not limited to, ACK1 antibodies, EGF antibodies, EPS8 antibodies; Erb 2 antibodies, Erb 3 antibodies, Erb 4 antibodies, TMEFF2 antibodies, and Xmrk antibodies.
  • each probe used in the methods disclosed herein to detect a cancer associated protein is specific for the detection of a single cancer associated protein.
  • each probe is an antibody with a specific affinity for a single cancer associated protein.
  • the probe used in the methods disclosed herein to detect the cancer associated protein is a signal transducer antibody.
  • the signal transducer antibody is selected from the group consisting of, but not limited to, A RAF antibodies, ASPP1 antibodies, ax1 antibodies, B Raf antibodies, CBLB antibodies; CD45R antibodies, ELMO antibodies, ERAS antibodies, FES antibodies, JAK1 antibodies, JAK2 antibodies, JAK3 antibodies, JNK1 antibodies, JNK2 antibodies, KAT13A antibodies, NDRG1 antibodies, PI3K antibodies, PIM antibodies, Ras (p21) antibodies, SRC1 antibodies, Styk1 antibodies, and c Ab1 antibodies.
  • phospho-specific antibodies can be used to detect phosphorylated cancer associated proteins in a sample (i.e., the detection of pAKT, p-mTOR, p-MAPK or pERK1/2). The phospho-specific antibodies are incubated with the sample for a sufficient amount of time to bind with the phosphorylated cancer associated proteins.
  • the sample is washed and then incubated with a secondary antibody comprising a detectable label, as routinely used in the art, resulting in the detection of phosphorylated cancer associated proteins in the sample.
  • a secondary antibody comprising a detectable label, as routinely used in the art.
  • Signal intensity from detection of the phosphorylated cancer associated proteins can be quantified to obtain intensity values for each detected phosphorylated cancer associated protein.
  • the instant disclosure contemplates, a probe specific for the detection of a cancer associated protein, wherein the probe is specific for an activated cancer associated protein.
  • the activated cancer associated protein comprises a phosphorylated cancer associated protein.
  • the phosphorylated cancer associated protein detected by the probe includes, but is not limited to, pAKT, p-mTOR, p-4E-BP1, p-eIF-4E (eukaryotic initiation factor 4E), pERK1/2, pHER2 (human epidermal growth factor receptor), p70S6K1 (phosphorylated ribosomal protein S6 kinase 1), phosphorylated ribosomal protein S6 (S6), p-glycogen synthase kinase 3 ⁇ , pMAPK, and pEGFR.
  • a phospho-specific antibody is used to detect the cancer associated protein in the sample.
  • the phosphorylated cancer associated protein is detected by phospho-specific antibodies including, but not limited to, pAKT antibodies, p-mTOR antibodies, p-4E-BP1 antibodies, p-eIF-4E antibodies, pERK1/2 antibodies, p-glycogen synthase kinase 3 ⁇ antibodies, pMAPK antibodies, and pEGFR antibodies.
  • the probe used to detect the cancer associated protein binds to the cancer associated protein in a manner that allows secondary antibodies comprising a tag or detectable label, such as biotin, to bind to the primary antibody, and thereby elicit a detectable label or tag as commonly known in the art.
  • a tag or detectable label such as biotin
  • a cancer associated protein is detected in a sample using a probe that has specificity for the (cancer) associated protein.
  • the probe used to detect the cancer associated protein can be an antibody.
  • the antibody can be a phospho-specific antibody that has specificity for a phosphorylated (activated) cancer associated protein.
  • other protein modifications that can be detected using, for instance, antibodies; such modifications include methylation, acetylation and ubiquitination. For instance, the utility of acetylation (with reference to drug response) is recognized (e.g. Chen et al., Anal Chem. 80(16):6390-6306, 2008).
  • total cellular protein content is measured for each membrane in the TMA, for example, by incubation with biotin. After biotinylation, the membranes are incubated with antibodies against the cancer associated protein to be detected. All primary antibodies are incubated overnight and incubated with secondary antibodies, such as streptavidin-linked Cy5 and FITC conjugated anti-rabbit IgG or anti-mouse IgG. The membranes (or blots) are dried, mounted and scanned. Regions of interest are selected, and the signal intensity quantified.
  • inter-array normalization The expression level of each cancer associated protein present on the membrane is normalized against the intensity of total cellular protein content of the same membrane, herein referred to as inter-array normalization.
  • the inter-array normalization step accounts for, and eliminates, background variations between membranes within a set. Thus, allowing the accurate determination of content for each cancer associated protein on each membrane within a set.
  • a second normalization step occurs.
  • each cancer associated protein detected in each individual sample is normalized to account for variations between individually tested samples.
  • each detected cancer associated protein is normalized against the expression level of known variable in the sample, such as normal epithelium or stroma.
  • the normal epithelia or stroma intensity data is compiled and awarded a value of 1.00.
  • the cancer associated proteins detected in the same sample are normalized against the value awarded to the normal epithelia resulting in a relative increase or decrease in expression of the cancer associated protein detected in the same sample.
  • the second normalization process accounts for, and eliminates, variations that occur between testing's of samples, such as differences that arise out of human error for example loading differences or differences that occur when two users are asked to perform the same assay.
  • each detected cancer associated protein is normalized against the expression level of known “housekeeping” protein in the sample, such as actin.
  • the intensity data for actin is compiled and awarded a value of 1.00.
  • the cancer associated proteins detected in the same sample are normalized against the value awarded to actin resulting in a relative increase or decrease in expression of the cancer associated protein detected in the same sample to accommodate for variations in testing between samples or batches.
  • the signal (or intensity) generated by the cancer associated protein can be normalized for background variation within a sample.
  • a cancer associated protein signal is normalized against a known “housekeeping” protein in the same sample.
  • this normalized procedure accounts for any non-linear transfer of proteins from one membrane to the next, and so on.
  • intra-array normalization can occur.
  • multiple samples representing discreet tissue samples or sources of sample may undergo independent testing to detect a cancer associated protein, e.g. a sample from a normal tissue section, an advanced stage disease tissue section, and an unknown disease state sample are concurrently evaluated.
  • the three independent samples can be normalized to account for discrepancies obtained within each sample.
  • the median expression of a “housekeeping” protein known to be present in each sample is used to normalize each sample to the next sample, and so on.
  • the median expression of a protein known to be expressed in each sample can be used to normalize each sample to the next sample (e.g. tubulin).
  • one sample can be transferred to multiple membranes and the stack of membranes is probed with multiple antibodies, in this case, each antibody preferably possesses an affinity for one cancer associated protein.
  • one antibody can be specific for the detection of PTEN
  • another antibody can be specific for the detection of phospho-AKT, or for a specific phosphorylated residue of AKT (e.g., Phospho T308-AKT or Phospho-S473-AKT)
  • a further antibody can possess affinity for the detection of phospho-mTOR.
  • the three cancer associated proteins migrate through the stack of membranes based on a variety of properties, including charge or mass, and can migrate to (for instance, be captured by) different membranes in the stack.
  • all three cancer associated proteins migrate to a different membrane in the stack, wherein each cancer associated protein is detected by an antibody specific for each cancer associated protein.
  • the detection levels observed or detected in each membrane can be normalized to obtain a normalized intensity value for each cancer associated protein detected in each membrane and within the sample.
  • the methodology disclosed herein can therefore accurately quantitate the content, level, or intensity of protein expression for one or more cancer associated proteins in a sample. Using this information, the expression level is normalized against a standard, such as total cellular protein content in the membrane, to account for variations in the background between membranes within one sample, and finally and additional normalization step can be used to account for variations between individual test samples.
  • a biomarker indicator as defined herein is a ratio-based determination of one or more cancer associated proteins in conjunction with an internal standard (such as, a housekeeping protein) or a normalization step (such as direct comparison of the cancer associated protein level against total cellular protein content) that is indicative of cancer.
  • an internal standard such as, a housekeeping protein
  • a normalization step such as direct comparison of the cancer associated protein level against total cellular protein content
  • the biomarker indicator comprises a ratio between at least one or more different cancer associated proteins.
  • the biomarker indicator can be used to detect the presence of cancer in a subject.
  • the biomarker indicator is a predictor of relative rates of survival for a subject with cancer.
  • the biomarker indicator can be used to monitor progression of disease.
  • an subject diagnosed with early stage cancer can be routinely monitor for the detection of specific cancer associated proteins, for examples levels of pAKT or p-mTOR (or one or more of p-mTOR, p-Akt, p-MAPK, EGFR, Her2 and Her3) can be measured in a subject to define an early stage biomarker indicator.
  • biomarker indicator the ratio of the one cancer associated protein as compared to total cellular content in the sample.
  • a significant decrease in biomarker indicator levels after measuring the same pAKT and p-mTOR cancer associated proteins may be interpreted as a development of the cancer (wherein a significant decrease in pAKT or p-mTOR levels are linked to increased risk of cancer), and consequently linked with a decreased rate of survival.
  • no change in the biomarker indicator may mean that a patient is not responding well to therapy and that a new treatment regime should be initiated.
  • a significant increase in the biomarker indicator may mean that treatment of the subject appears to be successful and should be continued. Additional specific biomarkers (using additional cancer associated proteins) are described herein.
  • the biomarker indicator is broadly applicable in various uses because the biomarker indicator provides the user with a starting point from which additional testing can be performed, and the results of the additional testing can be correlated with the first round of result so that a prognosis or adjustment of therapeutic regime can be made.
  • the biomarker indicator because of the inherent normalization steps involved means that the biomarker indicator is not vulnerable to discrepancies that exist between individual membranes, the particular membrane probed, or the type of probe used to obtain the biomarker indicator ratio. It will be apparent to one of ordinary skill in the art that the biomarker indicator can be made more or less stringent by using cancer associated proteins that are more strongly or less strongly correlated to one another.
  • a biomarker indicator formulated using cancer associated proteins from the AKT cell signaling pathway and the selection of two proteins associated with a cancer linked to the AKT cell signaling pathway will be considered a strong biomarker indicator.
  • the selection of two cancer associated proteins that are not currently known to be directly associated may also be found to be a strong biomarker indicator dependent upon the level of expression observed and the resulting biomarker indicator.
  • a multitude of equivalent areas such as circular or rectangular areas (for example, two, three, four, five, or more), from each region of a membrane can be defined and the mean value of fluorescence for each region determined and compared to the total area and thus, protein expression for each membrane.
  • Specific antibody signals can be calculated, as already discussed, and the sample normalized based on expression levels of total cellular protein content in the membrane.
  • relative expression intensities of each area within a sample can be normalized to normal epithelium or stroma. Thus, allowing for a relative comparison to be drawn between the cancer associated proteins detected in the sample and background expression.
  • analysis of tissue sections from patients with divergent clinical courses can be used to identify novel prognostic cancer associated proteins that better diagnose cancer, the stage of cancer in the subject, and furthermore can be used to correlate the expressed cancer associated protein levels against relative survival rates to predict patient survival, for example, post-surgery.
  • survival probability determination is performed by conducting statistical analysis of the one or more cancer associated proteins in conjunction with total cellular protein content of the sample to obtain a biomarker indicator. In another embodiment, survival probability determination is performed by conducting univariate statistical analysis of one or more cancer associated proteins in conjunction with total cellular protein content of the sample to obtain a biomarker indicator. In a further embodiment, survival probability determination is performed by conducting multivariate statistical analysis of one or more cancer associated proteins in conjunction with total cellular protein content of the sample to obtain a biomarker indicator. In some examples, the biomarker indicator is a measure of relative survival rate based on normalized expression of one or more cancer associated proteins in the sample.
  • statistical analysis of the one or more cancer associated proteins can be performed as a means to monitor progression of a cancer from a normal (disease free) sample or precancerous condition to an advanced stage of disease, wherein the normal or precancerous sample are correlated with a relatively high survival rate and a statistically significant worse patients' survival rate is associated with an advanced stage of disease sample.
  • the biomarker indicator can be used to determine prognosis of a subject with a cancer and thereby stratify patient treatment regimes based on responsiveness of the subject with cancer to different forms of treatment. For example, a subject with cancer who is currently undergoing treatment for the disease, and who demonstrates a statistically significant decrease in expression of two or more cancer associated proteins may be concluded (wherein a statistically significant decrease in the two cancer associated proteins correlates with decreased risk) as being responsive to the current form of treatment. Similarly, a subject with cancer who is undergoing treatment and continues to demonstrate a statistically significant increase in the one or more cancer associated proteins (wherein a statistically significant increase in the two cancer associated proteins correlates with increased risk) may be constructed as failing to respond positively to the current cancer treatment regime.
  • calculating survival analysis information comprises categorizing samples (cases) as high or low expressers of the cancer associated proteins under investigation by statistical analysis.
  • differential expression of p-AKT, p-mTOR and total P′I′EN in normal epithelia, dysplasia, and extrahepatic cholangiocarcinoma cases can be compared by Annova and Duncan's tests after normalization of expression.
  • expression of p-mTOR, p-Akt, p-MAPK, and/or EGFR are determined.
  • associations between categorical variables can be examined using Pearsons X 2 and Fisher's exact tests.
  • a recursive partition technique coupled with log-rank statistics can be employed to identify cut off points that discriminate outcome of patients based on the expression of the cancer associated proteins.
  • survival curves can be calculated using the Kaplan-Meier method. Statistical significance can be examined for example, by log-rank test and Cox proportional hazards regression model. In one embodiment, a P value of ⁇ 0.20 is considered statistically significant. In another embodiment, a P value of ⁇ 0.10 is considered statistically significant. In a further embodiment, a P value of ⁇ 0.05 is considered statistically significant.
  • a biomarker indicator for a subject with cancer can be obtained by determining the ratio of one or more cancer associated proteins in conjunction with a determination of total cellular protein content in the sample.
  • a biomarker indicator for a subject with cancer can be obtained by determining the ratio of PTEN/p-AKT in a sample. In yet another embodiment, a biomarker indicator for a subject with cancer can be obtained by determining the ratio of PTEN/p-mTOR in a sample.
  • the biomarker indicator is directed to a subject with a carcinoma, such as bile duct carcinoma.
  • the biomarker indicator is obtained by determining the ratio of one or more cancer associated proteins against total cellular protein content for a subject diagnosed with EHCC.
  • the biomarker indicator is obtained by determining the ratio of cancer associated proteins against total cellular protein content for a subject diagnosed with lung cancer or gastric cancer, or another cancer.
  • One of the advantages of the disclosed methods is that it allows multiple antigens to be assayed from a single tissue section. This approach permits simultaneously quantifying multiple cancer associated proteins with preservation of the morphologic structure of the tissue.
  • a further benefit of the disclosed methods is incorporation of a normalization step that allows for the accurate assessment and comparison of inter- and intra-array samples.
  • the methods disclosed herein allows for confirmation of the protein expression profiles observed, by standard immunohistochemistry techniques.
  • any ratio-based biomarker offers utility, and the combination of the two offers greater utility for the question addressed.
  • BM2 and BM4 can be thought of as normalizing biomarkers.
  • the denominator is downstream from the numerator in the relevant signalling pathway.
  • the biomarker indicator for a subject with cancer can be obtained by determining the ratio of p-mTOR/p-Akt and p-MAPK/EGFR in a sample.
  • the simple addition of these ratios provides an even more statistically significant biomarker indicator.
  • the two ratios used to generate this additive biomarker indicator that is, p-mTOR/p-Akt and p-MAPK/EGFR are themselves statistically significant predictive ratios.
  • the disclosure is directed to a method of making and using a platform to perform the disclosed methods, such as a Tissue MicroArray (TMA) or Multiplex Tissue Immunoblotting (MTI) array to detect the presence of a cancer in a subject.
  • TMA Tissue MicroArray
  • MMI Multiplex Tissue Immunoblotting
  • Particular embodiments are especially useful in connection with archival tissue samples that have been fixed and embedded, for instance in paraffin (FFPE).
  • FFPE paraffin
  • the method can involve providing a substrate (e.g., a gel, such as an embedding compound) to which a tissue section or tissue block is placed, then freezing and archiving of samples.
  • the block can then be sectioned into a plurality of sections such that the samples are at addressable locations in the sections.
  • Blocks or sections are deparaffinized and treated with pre-digestion enzymes for a brief time.
  • Slides' comprising a tissue block or tissue section is protease inhibited and transferred to a multi-membrane stack, such as a nitrocellulose membrane. Each membrane is incubated with primary antibodies against a specific protein marker of interest, for example a cancer associated protein.
  • total cellular proteins are measured by biotinylation of proteins present in the membrane, followed by incubation of the membranes with a secondary probe, such as streptavidin-Cy5.
  • a secondary probe such as streptavidin-Cy5.
  • signal intensity is quantified, and a ratio of the specific protein marker of interest/total protein content is obtained.
  • the obtained ratio is a biomarker indicator of survival that is used to predict or determine a subject's survival rate and thereby, can also be correlated with prognosis.
  • total protein detected by biotinylation can be used to generate the ratio.
  • biomolecules on a TMA are transferred to one or more membranes and can be visualized using detector molecules (“probes”), for example antibodies, lectins, or DNA hybridization probes, having specific affinity for the biomolecule(s) of interest.
  • probes for example antibodies, lectins, or DNA hybridization probes, having specific affinity for the biomolecule(s) of interest.
  • Specific embodiments provided herein include direct layered expression scanning techniques, which utilize a stack of “blank” membranes that are not specific for any particular target molecule. Instead, all (or a subset, e.g. proteins or nucleic acids) biomolecules in the sample ubiquitously bind to such membranes so as to give the user the flexibility of detecting a wide range of biomolecules in an open format.
  • the substrate is maintained at or below freezing while the samples are placed in the sample wells and frozen.
  • the samples are bonded to the substrate when the samples are frozen.
  • TMAs that are either loaded with sample or “blank” blocks, containing sample wells but no samples or an incomplete sample set) made using the described methods, and individual sections cut from such tissue blocks.
  • Other embodiments provide methods of parallel analysis of samples, such as biological samples (e.g., a protein, a mixture of proteins, a nucleic acid, a mixture of nucleic acids, a cell, or a biological fluid). Examples of these methods involve obtaining a plurality of (biological) samples, and placing each in an addressable location in a recipient array (for instance, a blank TMA) to produce a loaded array.
  • a recipient array for instance, a blank TMA
  • the recipient array is kept at or below freezing while the samples are being placed in the array. Sections can be cut (for instance, using a microtome or other device) from loaded arrays (arrays into which samples have been placed).
  • sections are cut from the arrays in a manner such that each section contains a plurality of portions of the samples placed in the array, which each maintain their assigned location. Sections from the provided TMA can be used to perform one or more biological analyses of samples in the arrays.
  • the biological samples are placed into recipient array as liquids (for instance, suspensions), and frozen after being placed in the array.
  • Biological samples include all clinical samples useful for detection of cancer in subjects, including, but not limited to, cells, tissues, and bodily fluids, such as: blood; derivatives and fractions of blood, such as serum; biopsied or surgically removed tissue, including tissues that are, for example, unfixed, frozen, fixed in formalin and/or embedded in paraffin; urine; sputum; cerebrospinal fluid; prostate secretions, pus; or bone marrow aspirates.
  • a sample includes a tissue biopsy obtained from a human subject, such as a fixed tissue section.
  • the sample includes cells from a liquid biological sample that are cancerous and that are frozen in an embedding material (such as paraffin) to become a solid sample suitable for manipulation by, for example, TMA.
  • the method includes analyzing tissue sections archived and previously obtained from the subject of interest.
  • a recipient array substrate may include an embedding compound that is solid at 0° C.
  • recipient array contains a plurality of wells to receive the biological samples. Examples of such wells have a substantially circular cross section having a diameter of less than about 2 mm.
  • more that one biological analysis for instance, an immunological binding assay, protein binding assay, activity assay, amplification reaction, or nucleic acid hybridization
  • the results of such analyses can be compared for the more than one biological analysis in corresponding assigned locations of different sections from the array to determine if there is a correlation between the results of the different biological analyses at different assigned locations.
  • the results of the different biological analyses performed on sections of a TMA are used to evaluate a reagent for disease diagnosis or treatment (e.g., evaluating a reagent selected from the group of antibodies, genetic probes, and antisense molecules, or a reagent selected from the group of biological inhibitors, biological enhancers, or other biological modulators); identify a prognostic marker for cancer; identify a prognostic marker for a non-cancerous disease; select targets for anti-cancer drug development; prioritize targets for anti-cancer drug development; assess or select therapy for a subject; and/or find a biochemical target for medical therapy.
  • a reagent for disease diagnosis or treatment e.g., evaluating a reagent selected from the group of antibodies, genetic probes, and antisense molecules, or a reagent selected from the group of biological inhibitors, biological enhancers, or other biological modulators.
  • identifying a prognostic marker for cancer or identifying a prognostic marker for a non-cancerous disease involves selecting a marker associated with a poor clinical outcome.
  • selecting therapy for the subject involves selecting an antineoplastic therapy that is associated with a particular biological analysis outcome.
  • Also provided are methods of analyzing a TMA which methods involve providing a plurality of elongated biological samples at addressable locations in a block of embedding substrate, such that when the block is frozen and cut into predetermined array sections, a two dimensional array of portions of the biological samples is presented at a surface of each section, with each portion of the biological samples at an addressable location in the array sections, and wherein each biological sample in the block has a third dimension so that when sequential sections of the block are cut, the biological samples maintain a predetermined relationship in the array sections; and exposing a plurality of the array sections to a probe that interacts with one or more of the biological samples of the array, to identify those biological samples that share or differ in a biological property.
  • the common biological property is a molecular characteristic, such as a presence or absence, or altered level of expression of a protein or gene, alteration of copy number, structure or function of a protein or gene, genetic locus, chromosomal region or chromosome.
  • the common biological property is correlated with at least one other characteristic of the samples, for instance clinical information (one or more of clinical course, tumor stage, oncogene status, and age of the subject from whom each sample was taken) about a subject from whom each sample was taken.
  • thin membranes in a stacked or layered configuration are applied to the sample, such as a tissue section, or protein or nucleic acid gel, and reagents and reaction conditions are provided so that at least a portion of the biomolecules (such as proteins) are eluted from the sample and transferred onto a plurality of the stacked membranes.
  • the resultant loaded (treated) membranes (or layers) are then separated.
  • Each membrane may be incubated with one or more different detectors (for example antibodies) specific for a particular biomolecule (such as a protein) of interest.
  • the detectors employed are labeled or otherwise detected using any of a variety of techniques, for instance chemiluminescence.
  • each membrane has essentially the same pattern of proteins bound to it, but different combinations of proteins are made visible (detectable) on each membrane due to the particular detectors (e.g., antibodies) selected to be applied.
  • detectors e.g., antibodies
  • one membrane layer may display proteins involved in programmed cell death (apoptosis) while an adjacent layer may display proteins involved in cell division such as tyrosine kinases.
  • nucleic acids may be targeted by using labeled DNA probes as detectors in lieu of antibodies.
  • different types of target biomolecules may be detected in different layers.
  • protein-specific detectors e.g., antibodies
  • nucleic acid detectors e.g., hybridization probes
  • a sample from which biological molecules are to be transferred e.g., a tissue section or gel
  • a sample and stack both the sample and stack (an assembled “contact transfer stack”) are placed inside a fluid impervious enclosure such as a plastic bag or the like.
  • the sample is supported by a substantially fluid impervious support, such as a glass slide; in these embodiments, the stack of membranes is placed on the other side of the sample.
  • the sample from which biomolecules are to be transferred is not supported by an impervious support, and the sample is placed between members of the membrane stack, such that one or more membranes is placed adjacent to each of two faces of the sample.
  • a liquid transfer reagent Also within the enclosure is a liquid transfer reagent. Heat and/or pressure are applied to the contents of the enclosure (from one or both sides) so as to permit proteins and other molecules to be transferred from the sample to the membrane stack. This produces multiple copies or replicas of the biomolecular content of the tissue sample.
  • the processed membranes (or layers) then may be separated and incubated with one or more different probes (e.g., nucleic acid hybridization probes or antibodies) specific for particular targets of interest.
  • the probes employed are labeled or otherwise detectable using any of a variety of techniques such as chemiluminescence.
  • each membrane has essentially the same pattern of biomolecules (including proteins and/or nucleic acids) bound to it, different combinations of such biomolecules are made visible on each membrane due to the particular probes or antibodies selected to be applied.
  • one membrane layer may be used to detect proteins associated with disease, for example breast cancer, while an adjacent layer may be used in detecting proteins associated with normal breast epithelium.
  • one membrane layer may be used to detect proteins associated with pancreatic cancer, while an adjacent layer may be used in detecting proteins associated with thyroid cancer.
  • the disclosed methods may be used for a side-by-side comparison of the protein expression patterns in different archival tissue samples, for instance from patients with different diseases, disease outcomes, or responses to therapies.
  • a particular drug can be correlated to a specific protein expression pattern from the diseased organ this provides a useful tool for predicting whether future patients likely will benefit or be harmed by that drug.
  • the disclosed methods may be used for a side-by-side comparison of disease state tissue, such as advanced stage prostate cancer, with a normal prostate tissue section.
  • disease state tissue such as advanced stage prostate cancer
  • the expression profile of normal prostate tissue can be used as a baseline for monitoring progression of prostate disease in a subject by contrasting the expression profile of a normal prostate section with a dysplasia section or with an advanced stage prostate section.
  • the protein expression profile of a normal tissue can act as a template to detect prostate cancer in a sample by contrasting the protein expression profile or expression of nucleic acid markers of interest in the normal prostate section against an unknown disease state section, wherein observations of new or significantly different protein or nucleic acid expression profiles may be an indicator of advanced disease state.
  • an advanced disease sample and normal tissue sample can be compared against a sample of unknown disease state.
  • the expression profile of the unknown disease state sample can be compared against both the diseased and disease free sample to identify where, in the transitional process the unknown sample is.
  • the provided methods may be used to screen archival tissue, which is usually formalin fixed and paraffin embedded.
  • Provided methods may also be used for examination of proteins that cannot be detected with antibodies in situ but can be detected after the protein has been transferred onto a membrane.
  • provided methods enable the quantitative analysis of targets in tissue, for example, the quantification of cell surface receptor density on malignant cells.
  • the methods, device, arrays, and kits provided herein can be used with laser capture micro dissected samples, permitting molecular analysis of tissue without protein or nucleic acid purification as a prerequisite.
  • These embodiments retain the two-dimensional relationship of distinct cell populations within the same tissue section so as to preserve the spatial relationships between the dissected cells and permit different cell types to be processed and analyzed in parallel.
  • methods are provided for detecting biomolecules in a sample collected by LCM, by eluting the biomolecules away from the microdissected sample and binding them to one or more membranes in a layered or stacked configuration, then visualizing the biomolecules on the membranes.
  • cellular samples embedded in/on an LCM transfer film are positioned adjacent to a stack of one or more membranes, and reagents and reaction conditions are provided so that the biomolecules are eluted from the cellular sample and transferred onto the membrane(s). Biomolecules on the membrane then can be detected and visualized using detector molecules (e.g., antibodies or DNA probes) having specific affinity for the biomolecule(s) of interest.
  • detector molecules e.g., antibodies or DNA probes
  • biomolecules e.g., proteins or nucleic acids
  • these membranes are constructed and/or chemically treated to have a high affinity but low capacity for the biomolecules. This allows the creation multiple replicates of the molecular content of the gel.
  • the membranes are separated and each is incubated with a one or a unique mixture (also referred to as a “cocktail”) of detectors (e.g., antibodies specific for a particular subset of proteins, nucleic acid probes, etc).
  • the detector cocktail is an antibody cocktail that has been carefully formulated so that no two antibodies in a cocktail bind overlapping or adjacent protein spots. Thus, protein spots that are too close together to be discriminated on a single membrane are detected on separate membranes according to the inventive method herein.
  • proteins that have been separated are transferred from the gel/support onto the membrane stack to allow the creation of multiple replicates or imprints of the protein content of the gel/support.
  • the amount of protein loaded into the wells is greater than the amount conventionally loaded so as to permit a more even and uniform distribution of the proteins throughout the stack.
  • antibodies can be used to detect many post-translational protein modification (e.g. phosphorylation), certain examples of disclosed methods can be employed to identify or analyze protein function as well as structure.
  • phospho-specific antibodies can be used in the disclosed methods to detect the presence of phosphorylated proteins in the sample.
  • the inability to detect phospho-specific binding in the method may be construed as no appreciable level of phosphorylated proteins being present in the sample.
  • described methods can be used for one-dimensional gels such as the identification of transcription factors separated by a gel-shift assay.
  • one specific embodiment is a method of analyzing the proteome of a biological sample.
  • Such a method involves separating the protein of interest from another protein present in the sample; transferring a portion of the separated protein to a plurality of membranes (for instance, 2, 10, 20 or more) in a stacked configuration; incubating each of the membranes in the presence of one or more species of predetermined ligand molecules (e.g., 2, 10, 20 or more) under conditions sufficient to permit binding between the separated protein and a ligand capable of binding to such protein; and analyzing the proteome by determining the occurrence of binding between the protein and any of the species of predetermined ligand molecules.
  • predetermined ligand molecules e.g., 2, 10, 20 or more
  • Another embodiment is a method for analyzing the extent of similarity between the proteomes of two or more samples.
  • Such a method involves, for each such sample, separating a protein of such sample from another protein present in the sample; transferring a portion of the separated protein to a plurality of membranes (e.g., 2, 10, 20 or more) in a stacked configuration; incubating two or more of the membranes in the presence of one or more species of predetermined ligand molecules (e.g., 2, 10, 20 or more) under conditions sufficient to permit binding between the separated protein and a ligand capable of binding to such protein; and analyzing the extent of similarity between the proteomes by comparing the separated proteins of each such sample with the separated proteins of another such sample for the occurrence of binding between the separated protein and any of the species of predetermined ligand molecules.
  • Another embodiment is a method for uniquely visualizing a desired predetermined protein if present in a biological sample.
  • This method involves separating the proteins present in the sample from one another; transferring a portion of the separated proteins of the sample to a plurality of membranes (for instance, 2, 10, 20 or more) in a stacked configuration; incubating two or more of the membranes in the presence of one or more species of predetermined detector/ligand molecules (e.g., 2, 10, 20 or more) under conditions sufficient to permit binding between desired predetermined protein and a ligand capable of binding to such protein; and visualizing any binding between the protein and any of the species of predetermined ligand molecules.
  • a plurality of membranes for instance, 2, 10, 20 or more
  • predetermined detector/ligand molecules e.g., 2, 10, 20 or more
  • electrophoresis for instance, 2-dimensional (2-D) gel electrophoresis
  • sample is obtained from mammalian cells or tissue, and particularly from human cells or tissue
  • mammalian cells or tissue are human cells or tissue and the separated protein is a product of a human gene.
  • the detector/ligand species can be any of a variety of molecule types.
  • the species of detector/ligand is an antibody, an antibody fragment, a single chain antibody, a receptor protein, a solubilized receptor derivative, a receptor ligands, a metal ion, a virus, a viral protein, an enzyme substrate, a toxin, a toxin candidate, a pharmacological agent, a pharmacological agent candidate, a hybridization probe, a oligonucleotide, and others as discussed herein.
  • inventions include all such methods wherein the binding of at least one of the species of detector/ligand is dependent upon the structure of the separated biomolecule (e.g., protein or nucleic acid). It still further provides the embodiments of all such methods wherein the binding of at least one of the species of detector/ligand is dependent upon the function of the separated biomolecule (e.g., a phosphorylated protein versus a non-phosphorylated protein).
  • the binding of at least one of the species of detector/ligand is dependent upon the structure of the separated biomolecule (e.g., protein or nucleic acid). It still further provides the embodiments of all such methods wherein the binding of at least one of the species of detector/ligand is dependent upon the function of the separated biomolecule (e.g., a phosphorylated protein versus a non-phosphorylated protein).
  • the disclosure also provides all such methods wherein at least one of the membranes is incubated with more than one species of ligand or detector molecule. Also provided are embodiments of all such methods wherein at least two membranes are employed, at least 10 membranes are employed, or at least 20 membranes are employed.
  • Additional embodiments are membranes that have a high affinity but a low capacity for proteins and/or other biomolecules so as to allow the creation of multiple replicates or imprints of the proteins eluted from a gel. Examples of these membranes are substantially thinner than those conventionally used for blotting.
  • the membranes are optionally provided with (or within) a frame, so that they may be easily handled and manipulated when separated from that stack.
  • the frame optionally defines a channel to permit release of air and fluid trapped between adjacent membranes.
  • Removable tabs or the like also may be provided on each frame to permit the stack to be held together, for instance when it is applied to the gel.
  • Loaded membranes may be scanned or otherwise digitally imaged using one of several commercially available scientific imaging instruments (for example, Image Quant). Imaging instrumentation and software, such as those described herein, may be employed to permit viewing, analysis, and/or interpretation of the expression patterns from the sample (e.g., a tissue sample or other two-dimensional source, such as a gel). Software may be provided with template images corresponding to each of the membrane images. This allows the identity of the biomolecule in each defined locus (e.g., a spot on a 2-D gel, a band on a 1-D gel, or a localized molecular deposit in a tissue sample) to be confirmed based on its vertical and horizontal position.
  • image Quant for example, Image Quant
  • Imaging instrumentation and software such as those described herein, may be employed to permit viewing, analysis, and/or interpretation of the expression patterns from the sample (e.g., a tissue sample or other two-dimensional source, such as a gel).
  • Software may be provided with template images corresponding to each of the membrane images. This
  • the software also can allow the density of each locus to be calculated so as to provide a quantitative read-out.
  • the software may also have links to a database of images generated from other gels to allow comparisons to be made between different diseased and normal samples.
  • the source sample e.g., actual tissue sections or other substantially two-dimensional source
  • a substantially similar sample e.g., an adjacent tissue slice
  • conventional techniques e.g., histochemical techniques
  • kits for uniquely visualizing a desired predetermined protein such as, a cancer associated protein, if present in a biological sample includes a plurality of membranes, each having a specific affinity for at least one cancer associated protein, and a plurality of detector/ligand species (e.g., species such as an antibody, an antibody fragment, a single chain antibody, a receptor protein, a solubilized receptor derivative, a receptor ligand, a metal ion, a virus, a viral protein, an enzyme substrate, a pharmacological agent, and a pharmacological agent candidate), each adapted to detect the desired cancer associated protein if bound to the membranes.
  • detector/ligand species e.g., species such as an antibody, an antibody fragment, a single chain antibody, a receptor protein, a solubilized receptor derivative, a receptor ligand, a metal ion, a virus, a viral protein, an enzyme substrate, a pharmacological agent, and a pharmacological agent candidate
  • the membranes described above include a porous substrate having a thickness of less than about 30 microns.
  • a kit include membranes that are polycarbonate membranes, especially polycarbonate membranes coated with a material for increasing the affinity of the membrane to biomolecules, for instance nitrocellulose, poly-L-lysine, or mixtures thereof.
  • cancer associated proteins from a sample may also be used as techniques to evaluate and quantify cancer associated proteins (or the corresponding nucleic acid expression profiles).
  • HPLC coupled with mass spectrometry to detect, isolate, and substantially purify cancer associated proteins from a sample and that the identified/detected cancer associated proteins can be used to obtain a biomarker indicator indicative of the presence of cancer or a particular disease state (e.g., early or advanced stage).
  • any two-dimensional sample material that contains releasable biomolecules can be used as a source of biomolecules in the provided transfer processes.
  • two-dimensional it is meant that the material is, or can be formulated so that it is, substantially flat and relatively thin.
  • substantially two-dimensional samples include tissue samples such as thin section slices (e.g., archival or frozen tissue samples), tissue arrays, cDNA or other nucleic acid microarrays, protein microarrays, 1-D protein gels, 1-D nucleic acid gels, 2-D protein gels, and so forth.
  • the described transfer methods, arrays, and devices can be used in forensic procedures to detect and study biological material such as bodily fluids; and so forth.
  • substances may be suspended in a liquid or gas, then run through and optionally affixed to a filter such as a sheet of filter paper, with the filter then used as the transfer sample.
  • a filter such as a sheet of filter paper
  • these samples can be referred to as structurally transformed samples, because their format is altered to render them substantially two dimensional prior to transfer onto a membrane stack.
  • modified single/fluid based cell samples e.g., leukemic cell samples
  • Embodiments provided herein may be used to identify biomolecules (e.g., proteins or nucleic acids) in any biological sample including bodily fluids (e.g. blood, plasma, serum, urine, bile, cerebrospinal fluid, aqueous or vitreous humor, or any bodily secretion), a transudate, an exudate (e.g. fluid obtained from an abscess or any other site of infection or inflammation), fluid obtained from a joint, and so forth.
  • bodily fluids e.g. blood, plasma, serum, urine, bile, cerebrospinal fluid, aqueous or vitreous humor, or any bodily secretion
  • a transudate e.g. blood, plasma, serum, urine, bile, cerebrospinal fluid, aqueous or vitreous humor, or any bodily secretion
  • an exudate e.g. fluid obtained from an abscess or any other site of infection or inflammation
  • fluid obtained from a joint e
  • the membranes comprise a material that non-specifically increases the affinity of the membrane to the biological molecules, or class of biomolecules (such as proteins or nucleic acids), that are moved through the membranes.
  • the membranes may be dipped in, coated with, or impregnated with nitrocellulose, poly-L-lysine, or mixtures thereof.
  • the membranes are sufficiently thin to allow the biomolecules to move through the plurality of membranes (for example 10, 50 100 or more) in the stack.
  • the membranes may be made of a material that does not substantially impede movement of the biomolecules through the membranes, such as polycarbonate, cellulose acetate or mixtures thereof.
  • the material of the membranes may maintain a relative relationship of biomolecules as they traverse through the membranes, so that the same biomolecules move through the plurality of membranes at corresponding positions. In such examples, the relative relationship allows the different membranes to be substantial “copies” of one another.
  • the membranes will be present as a stack of membranes that will include at least 2, at least 5, at least 10, at least 20, at least 50, or even more individual membranes.
  • Representative membranes for use in methods that utilize a membrane to detect and/or quantify biomolecules of interest include having a high affinity for protein and/or other biomolecules, but that have a low capacity for retaining such molecules. This binding profile permits biomolecules to pass through the membrane stack with only a limited number being trapped on each successive layer, thereby allowing multiple “copies” of the biomolecules in the sample to be generated. In other words, the low capacity allows the creation of multiple replicates as only a limited quantity of biomolecules is trapped on each layer.
  • the thickness of the substrate is for example, less than about 30 microns, and in particular embodiments is between 4-20 microns, for example between about 8-10 microns.
  • the pore size of the substrate is, for example between 0.1 to 5.0 microns, such as about 0.4-0.6 microns, and more specifically 4.0 microns.
  • the user has the option of analyzing a large number of different samples in parallel, thereby permitting direct comparisons between different patient samples (e.g., different patient samples, or patient samples and a reference standard, or samples of different tissues, etc.).
  • different patient samples e.g., different patient samples, or patient samples and a reference standard, or samples of different tissues, etc.
  • different samples from the same patient at different stages of diseases can be compared in a side-by-side arrangement, as can samples from different patients with the same disease, e.g., lung or another cancer.
  • each of the membranes comprises a ligand coating (e.g., a unique ligand coating, in that it is different from the other ligands in the membrane stack) that selectively binds to proteins in the biological sample based on a particular characteristic of the protein chemistry (e.g., hydrophobicity, carbohydrate content, etc).
  • the unique ligand coating may bind to proteins in the biological sample based on a particular functionality of the protein (e.g., phosphorylated, methylated).
  • membranes that are applicable for use with the methods described herein, for example, with a two-dimensional transfer assay of biomolecules.
  • Several different types of membranes are contemplated. Some of these membrane types overlap, in that a first membrane comprising a protein chemistry based ligand coating and a second membrane comprising a protein functionality based ligand coating can be used simultaneously in a membrane stack to transfer biomolecules from a sample. Even though not explicitly enumerated, all variations and combinations of the described methods are encompassed herein.
  • types of membranes and analysis of membranes disclosed in U.S. Pat. No. 6,969,615 and U.S. Pat. No. 6,951,761 are incorporated herein by reference in their entirety.
  • the processed membranes can be separated and each incubated with one or more different detector molecules (such as nucleic acid hybridization probes, lectins, or antibodies) specific for particular targets of interest.
  • detector molecules such as nucleic acid hybridization probes, lectins, or antibodies
  • the detectors/probes employed are labeled or otherwise detectable using any of a variety of techniques such as chemiluminescence.
  • chemiluminescence chemiluminescence
  • one membrane layer may display proteins involved in a disease state, such as cancer, while an adjacent layer may display proteins involved in normal tissue such as a “housekeeping” protein.
  • nucleic acids may be targeted and detected by using labeled DNA hybridization probes rather than antibodies.
  • both protein and nucleic acid targets can be detected in parallel by applying both antibodies and nucleic acid probes to different layers of the membrane stack.
  • Digital images of membranes may be created using a variety of instruments including the Image Station® CCD instrument available from Kodak Scientific Imaging (New Haven, Conn.). Alternatively, images may be captured on film (such as X-ray film) and digitalized by flat bed scanners. Software is preferably provided to align the images and perform densitometry functions. In examples using densitometry, the user can select the region of interest for analysis and the signal intensities are recorded and normalized. The numerical intensity values are then compared.
  • the membranes are separated from the stack and each is incubated in a separate solution of primary antibody specific for a desired protein. Only the area of the membrane containing the desired protein binds the antibody, forming a layer of antibody molecules. After incubation for about 1-8 hours, the membranes are usually washed in buffer to remove unbound antibody.
  • the loaded membranes are incubated with a secondary antibody that binds to the primary antibody.
  • the secondary antibody may be covalently linked to an enzyme such as horseradish peroxidase (HRP) or alkaline phosphatase (AP) that catalyzes substrate and the protein/antibody complex can be visualized using a number of techniques such as ECL, direct fluorescence, or colorimetric reactions.
  • HRP horseradish peroxidase
  • AP alkaline phosphatase
  • Ligands can be antibody fragments, receptors, receptor ligands, enzymes, viruses or viral particles, enzyme substrates or other small molecules that bind to specific proteins.
  • kits can also be employed to identify the functional state of the cancer associated proteins. One way to do so is to use phospho-specific antibodies to determine the phosphorylative state of a protein of interest.
  • Another approach to identify protein function is to first renature the proteins on the membranes by any of a number of techniques known in the art such as incubating the membrane in Triton-X® (octylphenol polymerized with ethylene oxide). Once renatured, proteins will regain their enzymatic activity and one of several substrate degradation assays known in the art can be used. With this approach the activity of kinases, phosphates and metalloproteinases can be determined.
  • Panels of proteins of interest for scientific research may be grouped by the proteins being involved in a particular cellular phenomenon such as apoptosis, cell cycle, signal transduction, etc.
  • Panels of proteins for clinical diagnostics may be grouped by proteins associated with a particular disease such as prostate cancer or breast cancer, etc.
  • the detectors/ligands employed are labeled or otherwise made detectable using any of several techniques, such as enhanced chemiluminescence (ECL), fluorescence, counter-ligand staining, radioactivity, paramagnetism, enzymatic activity, differential staining, protein assays involving nucleic acid amplification, etc.
  • ECL enhanced chemiluminescence
  • the membrane blots are preferably scanned, and more preferably, digitally imaged, to permit their storage, transmission, and reference.
  • Such scanning and/or digitalization may be accomplished using any of several commercially available scientific imaging instruments (see, e.g., Patton et al., Electrophoresis 14:650-658, 1993; Tietz et al., Electrophoresis 12:46-54, 1991; Spragg et al., Anal Biochem. 129:255-268, 1983; Garrison et al., J. Biol. Chem. 257:13144-13149, 1982; all herein incorporated by reference).
  • scientific imaging instruments see, e.g., Patton et al., Electrophoresis 14:650-658, 1993; Tietz et al., Electrophoresis 12:46-54, 1991; Spragg et al., Anal Biochem. 129:255-268, 1983; Garrison et al., J. Biol. Chem. 257:13144-13149, 1982; all herein incorporated by reference).
  • probes such as antibodies
  • detector cocktails that are useful in the analysis, detection and/or quantification of a cancer are described in Section V herein.
  • a panel of cancer associated proteins of interest is selected, such as PTEN, p-AKT, p-mTOR, p-MAPK, EGFR, HER2, and HER3 (or some subset thereof). These can be randomly selected proteins and/or proteins that are not directly related to one another, or may be groups of known proteins previously implicated to play a role in one or more particular cellular phenomena (e.g. apoptosis or growth factor pathways) or a particular disease (e.g. prostate cancer specific antigen, PSA).
  • apoptosis or growth factor pathways e.g. apoptosis or growth factor pathways
  • PSA prostate cancer specific antigen
  • these will be cancer associated proteins that have been characterized by sequence or coordinates on 2-D gels or for which ligands have been or could be generated.
  • Databases of annotated 2-D gels include the Quest Protein Database Center (on-line at //siva.cshl.org), the Swiss 2-D PAGE database (on-line at //expasy.cbr.nrc.ca/ch2d), Appel et al. Electrophoresis. 14(11):1232-1238, 1993; the Danish Centre for Human Genome Research (on-line at //biobase.dk/cgi-bin/celis), Celis et al., FEBS Lett. 398(2-3):129-134, 1996, etc.
  • Antibodies may be obtained from a variety of sources such as BD Transduction Laboratories (Lexington, Ky.) or Santa Cruz Biotechnology (Santa Cruz. CA).
  • Immunological identification of the cancer associated proteins on the membranes thus preferably involves the selection of antibodies having a high affinity and specificity for their protein targets.
  • antibodies monoclonal or polyclonal
  • This cross-reactivity phenomenon becomes increasingly apparent as the concentration of antibody increases relative to that of the sample proteins.
  • the first step in the antibody selection process preferably involves choosing antibodies (and their working concentrations) that consistently visualize preferably one but no more than five proteins on the same membrane.
  • the undesired proteins can be eliminated based on their X-Y positions on the membranes. Since the molecular weight and charge (pI) of a given protein is generally constant, it should appear at about the same coordinates on the gel each time it is run.
  • examples of the methods disclosed herein avoid this problem by designing an antibody cocktail to detect adjacent or overlapping cancer associated proteins on different membranes.
  • the antibody cocktails will be additionally tested for their specificity by control tests. For example, in a first test, membranes made from the transfer of a single gel (or from several gels that contain the same sample and were prepared in the same manner) will be probed with cocktails that differ in only one antibody component (each cocktail will lack one of the antibodies). As a result of this procedure, immunoblotted membranes should differ from each other in only one spot.
  • Each cocktail can also include one or more antibodies against “housekeeping” proteins (i.e., abundant structural proteins found in all eukaryotic cells such as actin, tubulin, etc.).
  • the antibodies employed will contain an antibody to actin, which will result in the production of a spot.
  • These antibodies serve as internal landmarks to normalize samples for loading differences and to compensate for any distortion caused by the gel running process.
  • the present disclosure allows not only the simultaneous characterization of a large number of different cancer associated proteins but also permits the characterization of a large number of characteristics of a single protein based on the number of different characteristics.
  • the protein p70 S6 kinase required for cell growth and cell cycle progression, is activated by phosphate group attachments (phosphorylation) to threonine on position 229 and/or 389 of the protein. Identification of this kinase would provide not only a determination of its presence or absence but also a demonstration of its activity.
  • a kit containing at least a four-membrane stack four copies can be made of a 2-D gel.
  • the first membrane would be incubated in antibody specific for the whole protein to determine if this protein is present in the sample or not.
  • the second membrane can be used in a kinase assay to determine if the protein is active or not.
  • the third membrane can be probed with phospho-p70 S6 kinase (Thr229) antibody to determine if activity of the enzyme is due to activation of this site.
  • the fourth membrane can be probed with phospho-p70 S6 Kinase (Thr389) antibody to determine if the activity of the enzyme is due to activation of this site. And since all of these tests are done on the single sample (rather than different batches of the same sample) the information obtained is more reliable.
  • Antibody cocktails are preferably stored in vials, preferably made of plastic or glass, and are optionally combined in a kit to create a “panel” of protein targets of interests.
  • Panels for scientific research may be grouped by the proteins involved in a particular cellular phenomenon such as apoptosis, cell cycle, signal transduction, etc.
  • Panels for clinical diagnostics may be grouped by proteins associated with a particular disease such as prostate cancer or breast cancer, etc.
  • breast cancer is the most common form of cancer in women and is the second leading cause of cancer-related deaths in women living in the United States, with more than 40,000 women dying from the disease each year.
  • Identification of breast cancer specific molecular targets has enormous potential to enhance detection, treatment, and prognosis of breast cancer disease.
  • understanding the role of breast cancer specific molecular targets in the process of transformation could reveal additional opportunities to therapeutically target other breast cancer specific targets, such as cancer associated proteins in the same or related cell signaling or growth factor pathways.
  • the instant disclosure contemplates methods for detecting breast cancer specific cancer associated proteins in a sample comprising a TMA that incorporates a probe that is specific for the detection of the breast cancer specific cancer associated protein; identifying the breast cancer specific cancer associated protein in the sample; and thereby correlating the presence of the breast cancer specific cancer associated protein with the presence of, or increased risk of, developing breast cancer.
  • the methods disclosed herein identify a simplified molecular signature for the identification of tumors and for the determination of relative survival rates of such tumors.
  • an advanced disease breast cancer sample can be probed using antibodies raised specifically against proteins known to be expressed in a disease state tissue.
  • an antibody cocktail comprising epidermal growth factor receptor (EGFR), HER2/neu, as well as the downstream activation factors, extracellular signal-regulated kinase 1/2 (ERK1/2), AKT, initiation factor 4E-binding protein 1 (4E-BP1), phosphorylated ribosomal protein S6 kinase 1 (p70S6K1), and ribosomal protein S6 (S6), all elements of the signaling pathway in breast cancer, can be applied to the one or more membranes of a TMA. Subsequent immunodetection and secondary fluorescent antibodies can be applied to the membrane(s) of the TMA and signal intensities of the cancer associated proteins can be quantified by molecular scanning densitometry software.
  • the intensities of the cancer associated proteins can be compared to the total cellular content of the sample to obtain a biomarker indicator.
  • the biomarker indicator can be correlated with relative cancer rates of survival.
  • the sample is studied by immunohistochemistry using phosphorylation-specific antibodies for the detection of activated (phosphorylated) breast cancer specific cancer associated proteins. Identification of breast cancer specific cancer associated proteins in the sample may correlate with the presence of breast cancer, an increased risk of developing breast cancer, or a decrease in overall survival rate for a subject with breast cancer.
  • expression of breast cancer specific cancer associated proteins measured in a breast tumor sample can be correlated with pathologic grade, patient survival, and tumor recurrence to determine which, if any, of the cancer associated proteins can be used as a molecular signature for breast cancer.
  • a protein such as 4E-BP1
  • the protein 4E-BP1 can be proposed as a molecular signature in the cell signaling of breast cancers and therefore applied as a breast cancer specific cancer associated protein.
  • Activation of the Src pathway is thought to cause resistance to standard medical treatment in some patients with breast cancer.
  • inhibiting the Src signaling pathway while providing standard of care treatment might overcome some aspects of drug resistance in affected patients. Understanding which parts and proteins of the Src pathway to measure in human tumors is therefore important when developing a molecular diagnostic tool that will allow oncologist's to select an appropriate signal transduction inhibitor in the clinic.
  • C35 is a protein abundantly expressed in breast cancer cells (C17orf37).
  • Anti-C35 antibodies can be utilized by routine immunohistochemistry techniques to confirm expression of the gene product of C35 in human tumors and normal tissues.
  • C35 is found to be highly expressed in breast carcinoma compared with normal breast epithelium and other normal tissues. Accordingly, C35 may be used in the disclosed methods as a potential molecular target for the therapeutic treatment of breast cancers or alternatively, as a molecular signature for the positive identification and detection of breast cancer in a sample.
  • candidate cancer associated proteins specific for prostate cancer include, but are not limited to hepsin, pim-1 kinase, AMACR, AIPC, e-cadherin (ECAD), ⁇ -methyl: acyl-coenzyme A racemase and EZH2 can be evaluated for use in a method to detect or monitor progression of prostate cancer.
  • EZH2 coupled with moderate expression of ECAD as detected using the methods disclosed herein may be found to be most strongly associated with the recurrence of prostate cancer, and thus, useful in defining a cohort of high-risk patients that can be offered adjuvant therapy.
  • EZH2-ECAD status is highly statistically significant with disease recurrence after radical prostatectomy, suggesting that EZH2-ECAD positive tumors require more aggressive treatment. Accordingly, EZH2-ECAD negative tumors may be considered a valuable selection tool for the development of watchful-waiting protocols aiding in the definition of low-risk disease for prostate cancer.
  • the identification of cancer associated proteins that are indicative of prostate cancer further provides cancer associated proteins that are useful for the diagnosis, detection, characterization and prognosis of prostate cancer.
  • the present disclosure also provides methods for characterizing prostate tissue in a subject comprising, providing a prostate tissue sample, detecting protein expression levels of at least two cancer associated proteins, comparing the protein expression levels of the at least two cancer associated proteins to a non-cancerous control sample, wherein a change in the protein expression level of the two cancer associated proteins as compared to the non-cancerous sample is associated with an increased risk for developing prostate cancer.
  • the two or more cancer associated proteins are selected from the group consisting of HEPSIN, FKBP5, FASN, FOLH1, TNFSF10, PCM1, S100A11, IGFBP3, SLUG, GSTM3, IL1R2, ITGB4, CCND2, EDNRB, APP, THROMBOSPONDIN 1, ANNEXIN A1, EPHA1, NCK1, MAPK6, SGK, HEVIN, MEIS2, MYLK, FZD7, CAVEOLIN2, TACC1, ARHB, PSG9, GSTM1, KERATIN 5, TIMP2, GELSOLIN, ITM2C, GSTM5, VINCULIN, FHL1, GSTP1, MEIS1, ETS2, PPP2CB, CATHEPSIN B.
  • COL1A2 RIG, VIMENTIN, MOESIN, MCAM, FIBRONECTIN 1, NBL1, ANNEXIN A4, ANEXIN A11, IL1R1, IGFBP5, CYSTATIN C, COL15A1, ADAMTS1, SKI, EGR1, FOSB, CFLAR, JUN, YWHAB, NRAS, C7, SCYA2, ITGA1, LUMICAN, C1S, C4BPA, COL3A1, FAT, MMECD10, CLUSTERIN, and PLA2G2A.
  • the disclosure additionally provides a method for detecting prostate cancer in a subject, comprising: providing a sample from a subject, calculating the protein expression level of at least two cancer associated proteins relative to the protein expression levels of a non-cancerous prostate tissue sample, wherein the two or more cancer associated proteins are selected from the group consisting of IGFBP5, MADH4, NBL1, SEPP1, RAB2, FAT, PP1CB, MPDZ, PRKCL2, ATF2, RAB5A, and Cathepsin H, wherein decreased expression of the cancer associated proteins in comparison to a normal non-cancerous sample is diagnostic of metastatic prostate cancer.
  • the method further provides a method for characterizing prostate cancer in a subject, comprising providing a tumor sample from a subject diagnosed with prostate cancer, detecting increased expression of at least two cancer associated proteins relative to a non-cancerous prostate tissue of two or more cancer associated proteins selected from the group consisting of CTBP1, MAP3K10, TBXA2R, MTA1, RAP2, TRAP1, TFCP2, E2-EPF, UBCH10, TASTIN, EZH2, FLS353, MYBL2, LIMK1, TRAF4, wherein increased expression of the two or more cancer associated proteins is diagnostic of metastatic prostate cancer.
  • Surgery is typically the first line of therapy for primary lung cancer, which is then followed up by radiation and/or chemotherapy. After removal of the primary tumor, a significant proportion of patients undergoing resection manifest evidence of non-detectable metastatic disease and show low survival rates.
  • a user can evaluate capabilities for discovering cancer associated proteins of metastatic lung cancer directly from a sample, for example, a formalin-fixed archival lung cancer tissue section.
  • lung cancer specific cancer associated proteins such as carcinoembryonic antigen (CEA), CYFRA21-1, plasma kallikrein B1 (KLKB1), Annexin A3, CKs, Prx I, II, III, fatty acid binding protein, and Neuron-Specific Enolase (NSE).
  • CEA carcinoembryonic antigen
  • CYFRA21-1 plasma kallikrein B1
  • KLKB1 plasma kallikrein B1
  • Annexin A3 Annexin A3
  • Prx I, II, III fatty acid binding protein
  • NSE Neuron-Specific Enolase
  • the instant disclosure contemplates methods for detecting lung cancer specific cancer associated proteins in a sample comprising a TMA that incorporates a probe that is specific for the detection of the lung cancer specific cancer associated protein by identifying the lung cancer specific cancer associated protein in the sample; and thereby correlating the presence of the lung cancer specific cancer associated protein with the presence of, or increased risk of, developing lung cancer.
  • the methods disclosed herein identify a simplified molecular signature for the identification of lung cancers and for the determination of relative survival rates of such tumors.
  • an advanced disease lung cancer sample can be probed using antibodies raised specifically against proteins known to be expressed in a disease state tissue.
  • an antibody cocktail comprising CEA, CYFRA21-1, KLKB1, or NSE, all proteins previously identified in lung cancer samples, can be applied to the one or more membranes of a TMA.
  • Subsequent immunodetection and secondary fluorescent antibodies can be applied to the membrane(s) of the TMA and signal intensities of the lung cancer specific cancer associated proteins can be quantified by molecular scanning densitometry software.
  • the intensities of the lung cancer associated proteins can be compared to the total cellular content of the sample to obtain a biomarker indicator.
  • the biomarker indicator can be correlated with relative cancer survival rates.
  • SCLC small cell lung cancer
  • the reference values for these proteins in human serum in healthy subjects vary from pg/mL (ProGRP) to ng/mL (NSE and CEA) levels and thus the detection of these SCLS specific cancer associated proteins can be evaluated through the analysis of a patients' serum using the methods disclosed herein, and is not therefore limited to an analysis by tissue biopsy or tissue block section.
  • ProGRP ProGRP
  • NSE and CEA ng/mL
  • the concentrations of the SCLC specific cancer associated proteins an ELISA and RIA can be used.
  • the intensity profiles of the SCLC specific proteins can be normalized using the methods disclosed herein to obtain a biomarker indicator for SCLC.
  • the biomarker indicator can then be used to obtain relative cancer survival rates using the methods discussed herein.
  • a panel of cancer associated proteins can be developed for the use in identification, detection and/or quantification of pancreatic cancer associated proteins in a sample.
  • a panel of five pancreatic cancer associated proteins comprising LCN2, REG1A, REG3, TIMP1, and IGFBP4 may be used to identify precancerous growths (pancreatic intraepithelial neoplasia), that are not observed in cancer patients or healthy control subjects.
  • the five-cancer associated protein panel is screened against blood samples from subjects to detect the presence of any, or all five proteins.
  • Positive detection of all five proteins in the protein panel is indicative or precancerous lesions in the subject, while detection of one or two proteins from the protein panel at low concentrations may be construed as a subject with currently a low-risk for pancreatic cancer. Accordingly, the five protein panel is potentially useful for the detection of pancreatic cancer, and may also be useful in identifying a cohort of subjects that are at an advanced stage of disease. Moreover, the development of specific cancer or multi-type cancer associated protein panels, such as the exemplary five protein panel discussed above in reference to pancreatic cancer, can be used to monitor, for example, the progression of disease in a subject from precancerous lesions to an advance stage of disease.
  • TG2 tissue type transglutaminase
  • the methods disclosed herein contemplate a method to detect the presence of ovarian cancer in a sample by calculating the content of at least one ovarian cancer specific cancer associated protein (e.g., TG2) in the sample, normalizing the content of the at least one ovarian cancer specific cancer associated protein, comparing the normalized value of the at least one cancer associated protein against normalized values for a normal non-cancerous sample, and correlating the change in normalized value of the cancer associated protein in the sample versus a non-cancerous sample to detect the presence of ovarian cancer in the sample.
  • TG2 as defined herein is a cancer associated protein with potential to detect or identify ovarian cancer in a sample using the methods disclosed herein.
  • TG2 is also a potential therapeutic target for the treatment of ovarian cancer and in particular, can be used to monitor the progression of disease or responsiveness of a subject to therapy, especially, chemotherapy-resistant tumors using the methods disclosed herein.
  • Extrahepatic cholangiocarcinoma is a malignant neoplasm of biliary tract epithelia arising from hepatic hilum to distal bile duct, and constitutes approximately 80-90% of all cholangiocarcinomas (Malhi and Gores, J. Hepatol 2006; 45:856-67).
  • EHCC Extrahepatic cholangiocarcinoma
  • Asia including Korea
  • surgical resection is the mainstay of treatment; however it is curative only in a limited number of patients, primarily those without advanced stage disease (Seyama and Makuuchi, World J.
  • PI3K phosphatidyl inositol 3 kinase
  • AKT signaling pathway is known to play an important role in regulating tumor cellular survival, apoptosis, and protein translation.
  • PI3K is activated by receptor tyrosine kinases (RTKs), and activation of RTKs leads to allosteric joining to the cellular membrane and subsequent tyrosine phosphorylation of the regulatory subunit of PI3K.
  • RTKs receptor tyrosine kinases
  • PI3K converts phosphatidyl inositol 2 phosphate (PIP2) to phosphatidyl inositol 3 phosphate (PIP3) (Cromwell et al., Mol. Cancer.
  • AKT is activated by phosphorylation at Thr308 by PIP3 and at Ser473 by the mammalian target of rapamycin (mTOR), as a part of the mTOR complex (mTORC) (Cromwell et al., Mol. Cancer. Ther., 2007; 6:2139-48).
  • mTOR mammalian target of rapamycin
  • mTORC mTOR complex
  • PTEN phosphatase and tensin homolog deleted on chromosome 10
  • PTEN is a well-described negative regulator of the PI3K/AKT signaling pathway, which functions as a tumor suppressor gene by induction of G1 phase cell cycle arrest through decreasing the levels of cyclin D1 (Radu et al., Mol Cell Biol.
  • Rapamycin was initially considered as a promising modality for blocking mTOR phosphorylation in several cancer types; however cancer patients with high AKT activity are reported to minimally respond to mTORC1 inhibitors (O'Reilly et al., Cancer Res. 2006; 66: 1500-1508). Patients with high expression of activated (phosphorylated) AKT were also reported resistant to radiation therapy (Gupta et al., Clin Cancer Res. 2002; 8:855-92). Therefore, what is needed is a new method for the detection of EHCC. In addition what is needed is an accurate and reliable method for determining survival probability for subjects with EHCC.
  • the present disclosure relates to compositions and methods for cancer diagnostics, including but not limited to cancer associated proteins.
  • the present disclosure identifies cancer associated proteins strongly associated with EHCC.
  • the present disclosure further provides novel biomarker indicators, in the form of a ratio-based determination of cancer associated proteins in conjunction with a normalization step, useful for the diagnosis, characterization, prognosis and treatment of EHCC.
  • the present disclosure provides a method for characterizing EHCC tissue in a subject by providing an EHCC tissue sample from a subject; and detecting the level of protein expression of p-AKT, p-mTOR or PTEN in the sample as compared to the level of protein expression in a non-cancerous sample, thereby characterizing the expression profile of cancer associated proteins in the EHCC tissue sample.
  • the subject comprises a human subject. In other embodiments, the subject comprises a non-human mammal.
  • the sample comprises a tumor biopsy. In some embodiments, the sample is a post-surgical tumor tissue sample and the method further comprises the step of identifying EHCC based on detecting changes in protein expression of p-AKT, p-mTOR or PTEN as compared to a normal tissue sample. In some embodiments, characterizing EHCC tissue comprises identifying a stage of EHCC cancer in the tissue. In some embodiments, the stage includes but is not limited to dysplasia, EHCC and metastatic EHCC. In some embodiments, the method further comprises the step of providing a prognosis to the subject. In other embodiments, the prognosis comprises an indicator ratio that determines the relative risk for developing EHCC.
  • the present disclosure provides a kit for characterizing EHCC cancer in a subject, comprising: a reagent capable of specifically detecting the presence of absence of expression of pAKT, p-mTOR or PTEN; and instructions for using the kit for characterizing EHCC cancer in the subject.
  • the reagent comprises an antibody that specifically binds to a pAKT, p-mTOR or PTEN polypeptide.
  • the present disclosure provides a method for characterizing an inconclusive biopsy tissue in a subject, comprising providing an inconclusive biopsy tissue sample from a subject; and detecting the presence of expression of a cancer associated protein in the sample, thereby characterizing the inconclusive biopsy tissue sample.
  • the detection step comprises detecting the presence of a p-AKT polypeptide (e.g., by exposing the p-AKT polypeptide to an antibody specific to the p-AKT polypeptide and detecting the binding of the antibody to the p-AKT polypeptide).
  • the subject comprises a human subject.
  • the presence of p-AKT expression in the inconclusive biopsy tissue is indicative of EHCC cancer in the subject.
  • the method further comprises the step of detecting expression of an additional cancer associated protein, such as p-mTOR or PTEN; and the presence of p-mTOR or PTEN expression (in addition to the presence of p-AKT expression) are indicative of prostate cancer in the subject.
  • the present disclosure further provides a method of detecting p-AKT, p-mTOR or PTEN expression in a bodily fluid, comprising providing a bodily fluid from a subject; and a reagent for detecting p-AKT, p-mTOR or PTEN expression in the biological fluid; and contacting the bodily fluid with the reagent under conditions such that the reagent detects p-AKT, p-mTOR or PTEN expression in the bodily fluid.
  • the bodily fluid is selected from the group consisting of serum, urine, whole blood, lymph fluid, and mucus.
  • the presence of p-AKT, p-mTOR or PTEN in the bodily fluid is indicative of cancer (e.g., EHCC).
  • the presence and/or levels of p-mTOR and p-AKT, or p-MAPK and EGFR, or all four (p-mTOR, p-AKT, p-MAPK and EGFR) in a bodily/biological fluid are indicative of cancer, particularly lung cancer.
  • Tissue Microarrays were constructed from archival formalin fixed, paraffin embedded tissue blocks from each of the above EHCC subjects. For each tumor, a representative tumor area was carefully selected from a hematoxylin and eosin stained section of the donor tissue block, using methods known in the art (see, for example, Hidalago et al., J. Clin. Pathol. 56:144-146, 2003 and Hong et al., Mod. Pathol. 20:562-569, 2007). 20 normal biliary epithelial, 67 biliary dysplasia, and 221 EHCC cases were studied. Each subject was represented with two cores of 1.5 mm diameter from each donor tissue block.
  • TMA Multiplex tissue immunoblotting
  • Intra-array normalization An optional, second normalization process (intra-array normalization) was performed to compensate for variations between sets of membranes under investigation. Intra-array normalization was performed by adjusting the median expression level of normal biliary epithelia.
  • Tissue sections were deparaffinized and hydrated in xylene and serial alcohol solutions, respectively. Endogenous peroxidase was blocked by incubation in 3% H 2 O 2 for 10 minutes. Antigen retrieval was performed in a steam pressure cooker with prewarmed antigen retrieval buffer pH 10 (Dako, Glostrup, Denmark) at 95° C., for 10 minutes. To minimize non-specific staining, sections were incubated with protein block (Dako) for 15 minutes. Primary antibodies were incubated overnight at 4° C. Antigen-antibody reactions were detected with DAKO LSAB+ peroxidase kit and DAB.
  • Anti-p-AKT, anti-p-mTOR, and PTEN antibodies were used at a dilution of 1:200. Immunostained sections were lightly counterstained with hematoxylin, dehydrated in ethanol, and cleared in xylene.
  • FIG. 1A The expression patterns of p-AKT and p-mTOR proteins detected by the multiplex tissue immunoblotting (MTI) assay performed as described in Example 2 were analyzed from 221 patients with EHCC. Representative expression signals of p-AKT, p-mTOR, and total PTEN for 16 cases are shown in FIG. 1A .
  • the signal intensity of FIG. 1A from maximum to minimum is shown as white to grey to black in order. Cases with higher intensity to p-AKT and p-mTOR showed lower intensity to total PTEN.
  • FIG. 1B shows immunohistochemical staining of p-AKT, p-mTOR, and PTEN protein in dysplasia and EHCC.
  • MTI immunohistochemical staining of p-AKT, p-mTOR, and PTEN protein in dysplasia and EHCC.
  • Cases with T1 TNM stage were found to have a significantly higher relative PTEN expression (mean, 16.08; relative expression intensity) than those with other classifications (T2, 8.92; T3, 7.18; T4, 3.98; p ⁇ 0.05, post hoc Duncan test, FIG. 3A ).
  • Patients with invasion of the pancreas were observed to have significantly less PTEN expression (mean, 5.94) than those without pancreas invasion (mean, 9.95; p ⁇ 0.05, post hoc Duncan test).
  • Cases with duodenal invasion had statistically less PTEN expression (mean, 3.98) than those without duodenal invasion (mean, 9.04; p ⁇ 0.05, post hoc Duncan test, FIG. 3B ).
  • FIG. 4 shows a Kaplan-Meier survival analysis of EHCC according to PTEN expression.
  • the 1, 3, and 5 year survival rate for patients with low PTEN expression were 80.0%, 13.3%, and 13.3%, respectively, while 1, 3, and 5 year survival rate for those with high PTEN expression were 80.7%, 52.3%, and 42.2%, respectively.
  • FIG. 5A shows Kaplan-Meier survival analysis of EHCC according to PTEN/p-AKT expression.
  • FIG. 5B shows Kaplan-Meier survival analysis of EHCC according to PTEN/p-mTOR expression.
  • the 1, 3, and 5 year survival rate for patients with low PTEN/p-mTOR group was 76.2%, 22.9%, and 11.4%, respectively, while 1, 3, and 5 year survival rate for those with high PTEN/p-mTOR expression was observed to be 81.3%, 52.9%, and 43.3%, respectively ( FIG. 5B ).
  • T1 tumors Thirty-four cases were T1 tumors, eighty cases were T2 tumors, eighty-four cases were T3 tumors, and twenty-three cases were T4 tumors.
  • the length of the patients' follow-up time ranged from 1 to 128 months, and median survival at last follow up was 34 months.
  • differentiation P ⁇ 0.0001
  • the instant method was utilized to profile proteomic expression profiles of cancer associated proteins, for example by transferring proteins from a paraffin-embedded tissue section to a stack of membranes to which conventional immunoblotting techniques were applied.
  • One of the advantages of the current method is that it allows multiple antigens to be assayed from a single tissue section. This approach permits simultaneously quantifying multiple cancer associated proteins with preservation of the morphologic structure of the tissue.
  • a further benefit of the current method is incorporation of a normalization step that allows for the accurate assessment and comparison of inter- and intra-array samples.
  • the method disclosed herein allows for confirmation of the protein expression profiles observed, by standard immunohistochemistry techniques. Utilizing the current method, quantitative analysis of protein expression profiles, such as PTEN, mTOR and AKT were obtained and provided survival probability information, as well as the capacity to stratify patients.
  • NSCLC non-small cell lung cancer
  • MAPK mitogen-activated protein kinase
  • AKT AKT signaling pathways
  • this example demonstrates that the sum of p-mTOR/p-AKT and p-MAPK/EGFR is a predictive marker of survival in patients with NSCLC.
  • Quantitative image analysis of immunohistochemistry with algebraic-like equations offers novel biomarkers of survival and provides clues in identification of patients for targeted therapy.
  • Lung cancer is the most common cause of cancer deaths in both men and women worldwide. Despite advances in treatment, such as combination chemotherapy and chemoradiation, survival has improved very little over the past few decades (Schiller, Oncology 61 Suppl 1:3-13, 2001).
  • Gefitinib the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor
  • NSCLC non-small cell lung cancer
  • AKT activated by extracellular stimuli in a PI3K-dependent manner, pivotal role in oncogenesis (Franke et al., Cell 88:435-7, 1997). Induction of these pathways is mediated by phosphorylation of the proteins involved.
  • a total of 231 lung cancer cases were selected from the pathology case archive of Toyama University Hospital based on the diagnosis and the quality of the available tissue on the paraffin blocks. These patients did not receive neoadjuvant treatment.
  • the tumors were staged according to the International Union against Cancer's TNM classification and histologically divided and graded according to 2004 WHO guidelines (Fukuoka et al., Clin Cancer Res 10:4314-24, 2004).
  • TMAs were constructed using a TMA arrayer (Pathology Devices, Riverside, Md.) as previously described (Kononen et al., Nat Med 4:844-7, 1998). For each case, areas with the most representative histology were selected from review of hematoxylin-eosin (H&E) stained slides.
  • the cylindrical tissue samples (0.6 mm) were cored from the above described areas in the donor block and extruded into the recipient array. Multiple 5 ⁇ m thick sections were cut with a microtome and H&E staining of TMA slides were examined every 50th sections for the presence of tumor cells.
  • EGFR (M3563) antibody was purchased from DAKO (Carpinteria, Calif.), and p-AKT (T308), p-MAPK and p-mTOR antibodies were purchased from Cell Signaling (Beverly, Mass.).
  • the tissue sections were deparaffinized in xylene and rehydrated through a graded alcohol series to distilled water as described herein.
  • Antigen retrieval for EGFR was performed using Proteinase K (DAKO), for p-mTOR using Pressure Chamber (Pascal, DAKO) with pH 6 Target Retrieval Solution (DAKO), and for other antibodies using it with pH10 Target Retrieval Solution (DAKO). These slides were blocked with hydrogen peroxide/methanol.
  • Hierarchical clustering was performed on the basis of WS. WS above 10% of the highest score was considered high score group to evaluate each antibody. Using the chi-square test, the antibodies were evaluated in association with each other within each category.
  • the number of cases eventually extracted for final analysis is listed in Table 3 along with their clinical data. Survival time and outcome was limited to 204 of 231 cases.
  • the TMA was stained for p-AKT, p-mTOR, EGFR and p-MAPK. Slides were manually reviewed for quality of staining ( FIG. 6 ), and imaged with an Aperio Scanscope CS (Vista, Calif.) with a 20 ⁇ objective. The TMAs were subsequently de-arrayed in Spectrum Plus, and tumor features were annotated by hand for each TMA core for image analysis. Cores with inadequate tumor were excluded. After tuning of the positive pixel and membrane image analysis algorithms, image analysis was performed on the TMA. A value-weighted score was calculated for each tumor.
  • FIG. 8C proves that the described “biomarker algebra” is modeling the behavior of multiple biomarkers in a fashion that is superior.
  • the p-MAPK/EGFR ratio of category 2 is same as category 3
  • the p-mTOR/p-AKT ratio of category 2 is higher than that of category 3.
  • the five-year survival rate of category 2 is 74% and of category 4 is 45%, although stage and lymph node metastasis rate is almost same.
  • Quantitative analysis was usually determined by using a scale for assessment of distribution and/or a scale for assessment of intensity (Vergis et al., Lancet Oncol 9:342-51, 2008; Howard et al., Lung Cancer 46:313-23, 2004; Yano et al., Cancer Res 68:9479-87, 2008).
  • ratios reflect a measure of activity through a pathway, as well as dysregulation of this pathway, and may be useful in identification of patients for targeted therapies.
  • the capacity to add two ratio-based biomarkers into a complex, four-antibody combined biomarker is a reflection of the cross-talk between signaling pathways and may other biologically relevant features of the tumors.
  • Immunohistochemistry was a weak tool for quantitative comparison of multiple antibodies. Quantitative image analysis of immunohistochemistry with algebraic-like equations offers novel biomarkers of survival. As illustrated in this example, the sum of p-mTOR/p-AKT and p-MAPK/EGFR is a predictive marker of survival in patients with NSCLC.
  • gastric (stomach) cancers have been examined in a very large cohort of 946 patients for whom detailed clinico-pathologic data is available. Numerous markers have been interrogated, including mucin genes, p53, e-cadherin, beta-catenin and others. Her2 and Her3 have been examined using “manual” interpretation by a pathologist, resulting in non-continuous data—qualitative data, but with a range of values rather than binary. In a multivariate analysis with hazards ratios, HER2 expression was a negative prognostic factor (HR 1.37) and HER3 was a positive prognostic factor (HR 0.94). Ratio-based metrics have been applied, demonstrating an HR of 0.61. All of the HRs are statistically significant, however the greater deviation from 1.0, the greater the significance.
  • HER2 and HER3 are not up/downstream of each other in a signaling pathway, but instead they form a functional heterodimer.
  • the proposed model is that it is the balance of HER2 to HER3 expression that is predictive, where an excess of HER2 (for instance, through overexpression of HER2 or underexpression of HER3) is a poor prognostic marker.
  • this model is similar to the relationship of the denominator factor(s) being downstream of numerator factor(s), and further supports the concept that the herein-described types of ratiometric biomarkers are functional when there is a connection between the two markers at the biologic level. This also further supports the conclusion that the relationships reported herein are not random observational events.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Medical Informatics (AREA)
  • Biotechnology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Pathology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Evolutionary Biology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Genetics & Genomics (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Theoretical Computer Science (AREA)
  • Microbiology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Data Mining & Analysis (AREA)
  • Databases & Information Systems (AREA)
  • Epidemiology (AREA)
  • Primary Health Care (AREA)
  • Biophysics (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
US13/144,474 2009-01-14 2010-01-13 Ratio based biomarkers and methods of use thereof Abandoned US20110306514A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/144,474 US20110306514A1 (en) 2009-01-14 2010-01-13 Ratio based biomarkers and methods of use thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US14450109P 2009-01-14 2009-01-14
US13/144,474 US20110306514A1 (en) 2009-01-14 2010-01-13 Ratio based biomarkers and methods of use thereof
PCT/US2010/020944 WO2010083252A2 (fr) 2009-01-14 2010-01-13 Biomarqueurs basés sur des taux et méthodes d'utilisation associées

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/020944 A-371-Of-International WO2010083252A2 (fr) 2009-01-14 2010-01-13 Biomarqueurs basés sur des taux et méthodes d'utilisation associées

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/841,176 Continuation-In-Part US20130210648A1 (en) 2009-01-14 2013-03-15 Ratio based biomarkers and methods of use thereof
US15/349,991 Continuation US20170115295A1 (en) 2009-01-14 2016-11-11 Ratio based biomarkers and methods of use thereof

Publications (1)

Publication Number Publication Date
US20110306514A1 true US20110306514A1 (en) 2011-12-15

Family

ID=42028252

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/144,474 Abandoned US20110306514A1 (en) 2009-01-14 2010-01-13 Ratio based biomarkers and methods of use thereof
US15/349,991 Abandoned US20170115295A1 (en) 2009-01-14 2016-11-11 Ratio based biomarkers and methods of use thereof
US16/217,540 Active US11237169B2 (en) 2009-01-14 2018-12-12 Ratio based biomarkers and methods of use thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/349,991 Abandoned US20170115295A1 (en) 2009-01-14 2016-11-11 Ratio based biomarkers and methods of use thereof
US16/217,540 Active US11237169B2 (en) 2009-01-14 2018-12-12 Ratio based biomarkers and methods of use thereof

Country Status (8)

Country Link
US (3) US20110306514A1 (fr)
EP (3) EP2667195B1 (fr)
JP (3) JP5907732B2 (fr)
AU (2) AU2010204741B2 (fr)
CA (1) CA2749601C (fr)
IL (5) IL214046A (fr)
NZ (4) NZ624816A (fr)
WO (1) WO2010083252A2 (fr)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013159016A1 (fr) * 2012-04-20 2013-10-24 University Of Connecticut Pipeline pour la conception rationnelle et l'interprétation de panels de biomarqueurs
WO2013163451A1 (fr) * 2012-04-27 2013-10-31 Bio-Rad Laboratories, Inc. Détermination quantitative et normalisation de protéines sans coloration
US8632971B2 (en) 2011-11-08 2014-01-21 Caldera Health Limited Methods and materials for determining the efficacy of prostate cancer therapies
US20140037642A1 (en) * 2011-02-02 2014-02-06 Amgen Inc. Methods and compositions relating to inhibition of igf-1r
US8658164B2 (en) 2011-11-08 2014-02-25 Caldera Health Limited Methods and materials for the diagnosis of prostate cancers
US8658166B2 (en) 2011-11-08 2014-02-25 Caldera Health Limited Methods and materials for the diagnosis of prostate cancers
US8703127B2 (en) 2011-11-08 2014-04-22 Caldera Health Limited Methods and materials for the diagnosis of prostate cancers
WO2015120416A1 (fr) * 2014-02-07 2015-08-13 The Johns Hopkins University Biomarqueurs pour l'évaluation d'un traitement sur des patients atteints d'un cancer
WO2015035250A3 (fr) * 2013-09-05 2015-11-19 Dendreon Corporation Réponse immunitaire humorale dirigée contre des antigènes tumoraux après traitement à l'aide d'une immunothérapie active spécifique d'antigènes du cancer et son association avec un résultat clinique amélioré
US20170016911A1 (en) * 2014-03-24 2017-01-19 Konica Minolta, Inc. Method for quantifying biological material based on multiple immunostaining
US20170168055A1 (en) * 2015-12-11 2017-06-15 Expression Pathology, Inc. SRM/MRM Assays
WO2017202962A1 (fr) 2016-05-24 2017-11-30 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés et compositions pharmaceutiques pour le traitement du cancer du poumon non à petites cellules (cbnpc) qui coexiste avec la bronchopneumopathie chronique obstructive (bpco)
WO2018046736A1 (fr) 2016-09-12 2018-03-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de prédiction du temps de survie de patients souffrant d'un cancer
WO2018055080A1 (fr) 2016-09-22 2018-03-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes et compositions pharmaceutiques permettant la reprogrammation de l'environnement immunitaire chez un sujet en ayant besoin
US9994912B2 (en) 2014-07-03 2018-06-12 Abbott Molecular Inc. Materials and methods for assessing progression of prostate cancer
WO2018122245A1 (fr) 2016-12-28 2018-07-05 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de prédiction de la durée de survie de patients souffrant d'un cancer colorectal cms3
WO2018122249A1 (fr) 2016-12-28 2018-07-05 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes permettant de prédire le temps de survie de patients souffrant d'un cancer colorectal stable microsatellitaire
WO2018160988A1 (fr) * 2017-03-03 2018-09-07 Expression Pathology, Inc. Profilage immunitaire de tissus tumoraux
WO2018202850A1 (fr) 2017-05-05 2018-11-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Médicaments ciblant les microtubules en tant qu'inhibiteurs de points de contrôle immunitaires et procédés de criblage de nouveaux inhibiteurs de points de contrôle immunitaires pour le traitement de cancers et de maladies infectieuses
WO2020141199A1 (fr) 2019-01-03 2020-07-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes et compositions pharmaceutiques pour améliorer les réponses immunitaires dépendantes des lymphocytes t cd8+ chez des sujets souffrant d'un cancer
EP3696283A1 (fr) * 2013-03-08 2020-08-19 MDxHealth Research B.V. Marqueurs moléculaires dans le cancer de la vessie
WO2020201362A2 (fr) 2019-04-02 2020-10-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes de prédiction et de prévention du cancer chez des patients ayant des lésions prémalignes
WO2020208060A1 (fr) 2019-04-09 2020-10-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Utilisation d'inhibiteurs de sk2 en association avec une thérapie de blocage de point de contrôle immunitaire pour le traitement du cancer
WO2020245155A1 (fr) 2019-06-03 2020-12-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de modulation d'un régime de traitement
US20210069295A1 (en) * 2017-11-10 2021-03-11 Stc. Unm Galectins control mtor in response to endomembrane damage and provide a mechanism and target for the treatment of autophagy-related diseases
WO2021074391A1 (fr) 2019-10-17 2021-04-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de diagnostic d'adénocarcinomes du type intestinal nasal
US11015190B2 (en) * 2013-09-17 2021-05-25 Lucence Life Sciences Pte Ltd Method of treating a patient having renal cancer
US11047860B2 (en) 2015-11-05 2021-06-29 Vanderbilt University Protein quantitation in multicellular tissue specimens
WO2021170777A1 (fr) 2020-02-28 2021-09-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de diagnostic, de pronostic et de gestion du traitement du cancer du sein
US11204355B2 (en) 2017-02-20 2021-12-21 Vanderbilt University Immune checkpoint molecular fitness profiling by mass spectrometry
WO2022002873A1 (fr) 2020-06-30 2022-01-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés pour prédire le risque de récidive et/ou de mort de patients souffrant d'un cancer solide après des traitements adjuvants pré-opératoires
WO2022002874A1 (fr) 2020-06-30 2022-01-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés pour prédire le risque de récidive et/ou de mort de patients souffrant d'un cancer solide après un traitement adjuvant préopératoire et une chirurgie radicale
WO2022020218A1 (fr) * 2020-07-19 2022-01-27 Aravive Inc Médthodes de diagnostic du cancer faisant appel à des récepteurs leurres axl
CN114113611A (zh) * 2021-12-13 2022-03-01 郑州大学 一种用于肝癌诊断的生物标志物及检测试剂盒
US11435360B2 (en) 2016-12-16 2022-09-06 The Brigham And Women's Hospital, Inc. System and sensor array
WO2022200303A1 (fr) 2021-03-23 2022-09-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés pour le diagnostic et le traitement de lymphomes t
WO2023144303A1 (fr) 2022-01-31 2023-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Cd38 en tant que biomarqueur et biocible dans des lymphomes t
WO2023175366A1 (fr) 2022-03-17 2023-09-21 Veracyte Méthodes de prédiction de la réponse à un traitement immunothérapeutique chez un patient atteint d'un cancer
WO2023198874A1 (fr) 2022-04-15 2023-10-19 Institut National de la Santé et de la Recherche Médicale Méthodes pour le diagnostic et le traitement de lymphomes t
WO2023198648A1 (fr) 2022-04-11 2023-10-19 Institut National de la Santé et de la Recherche Médicale Méthodes de diagnostic et de traitement de malignités des lymphocytes t
WO2024003310A1 (fr) 2022-06-30 2024-01-04 Institut National de la Santé et de la Recherche Médicale Méthodes de diagnostic et de traitement de la leucémie lymphoblastique aiguë
WO2024018046A1 (fr) 2022-07-22 2024-01-25 Institut National de la Santé et de la Recherche Médicale Garp utilisée en tant que biomarqueur et biocible dans des malignités de lymphocytes t
WO2024023283A1 (fr) 2022-07-29 2024-02-01 Institut National de la Santé et de la Recherche Médicale Lrrc33 en tant que biomarqueur et biocible dans des lymphomes t cutanés
US11906526B2 (en) 2019-08-05 2024-02-20 Seer, Inc. Systems and methods for sample preparation, data generation, and protein corona analysis
WO2024079192A1 (fr) 2022-10-12 2024-04-18 Institut National de la Santé et de la Recherche Médicale Cd81 utilisé en tant que biomarqueur et cible biologique dans des malignités de lymphocytes t

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1871809A2 (fr) 2005-04-22 2008-01-02 Morphotek, Inc. Anticorps a activite d'effecteur immunitaire et internalises dans des cellules positives vis-a-vis de l'endosialine
BRPI0809665B8 (pt) 2007-04-05 2021-05-25 Eisai Inc anticorpo monoclonal que se liga especificamente a endosialina
NZ624816A (en) 2009-01-14 2015-07-31 Us Health Ratio based biomarkers and methods of use thereof
DK3147373T3 (da) 2010-07-27 2019-08-12 Genomic Health Inc Fremgangsmåde til anvendelse af genekspression til at bestemme prognosen for prostatacancer
WO2012021887A2 (fr) * 2010-08-13 2012-02-16 Arizona Borad Of Regents, A Body Corporate Acting For And On Behalf Of Arizona State University Biomarqueurs pour la détection précoce du cancer du sein
TW201302793A (zh) 2010-09-03 2013-01-16 Glaxo Group Ltd 新穎之抗原結合蛋白
JP6047503B2 (ja) 2010-12-29 2016-12-21 エクスプレッション、パソロジー、インコーポレイテッドExpression Pathology, Inc. Her3タンパク質SRM/MRMアッセイ
JP5862671B2 (ja) * 2011-08-23 2016-02-16 日本電気株式会社 情報処理システム、情報処理方法、情報処理装置およびその制御方法と制御プログラム
EP2776587A4 (fr) * 2011-11-10 2015-07-15 Exosome Diagnostics Inc Dosage du liquide céphalorachidien
WO2014130444A1 (fr) * 2013-02-19 2014-08-28 Genomic Health, Inc. Méthode de prédiction du pronostic du cancer du sein
CA2904973A1 (fr) * 2013-03-15 2014-09-25 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Biomarqueurs bases sur le rapport et leur procedes d'utilisation
ES2754177T3 (es) 2013-06-12 2020-04-16 Massachusetts Gen Hospital Métodos para la detección multiplexada de moléculas diana y sus usos
EP3011059B1 (fr) 2013-06-20 2019-02-06 Immunexpress Pty Ltd Identification de marqueur biologique
US11047010B2 (en) 2014-02-06 2021-06-29 Immunexpress Pty Ltd Biomarker signature method, and apparatus and kits thereof
JP6675716B2 (ja) * 2015-05-08 2020-04-01 学校法人 川崎学園 免疫因子を指標とした肺腺癌の予後演算式作成方法と予後推定方法
SE540173C2 (en) * 2016-04-20 2018-04-24 Hiloprobe Ab Marker genes for colorectal cancer classification, method for judging lymph node metastasis for prognosis of colorectal cancer and kit therefor
US11243208B2 (en) 2016-07-11 2022-02-08 Arizona Board Of Regents On Behalf Of Arizona State University Autoantibody biomarkers for the early detection of ovarian cancer
ES2921701T3 (es) 2016-08-05 2022-08-30 Univ New York State Res Found Queratina 17 como un biomarcador para el cáncer de vejiga
CN106370853B (zh) * 2016-08-26 2018-08-21 苏州大学 人肝癌的预测性生物标志物及其应用
AU2018393024A1 (en) 2017-12-20 2020-07-09 Laboratory Corporation Of America Holdings Compositions and methods to detect head and neck cancer
JP7184373B2 (ja) * 2020-01-29 2022-12-06 学校法人九州文化学園 生体外での腸管腫瘍の検査方法
WO2022154037A1 (fr) * 2021-01-14 2022-07-21 公立大学法人福島県立医科大学 Biomarqueur pronostique pour le cancer
GB202103200D0 (en) * 2021-03-08 2021-04-21 Terasom S R O Lung Cancer diagnosis
JPWO2022264545A1 (fr) * 2021-06-17 2022-12-22

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5866366A (en) 1997-07-01 1999-02-02 Smithkline Beecham Corporation gidB
US6969615B2 (en) 1999-07-26 2005-11-29 20/20 Genesystems, Inc. Methods, devices, arrays and kits for detecting and analyzing biomolecules
EP2277991A1 (fr) 2001-08-21 2011-01-26 Ventana Medical Systems, Inc. Procédé et essai de quantification permettant d'évaluer du système c-kit/scf/pakt
US6951761B2 (en) 2001-08-31 2005-10-04 The United States Of America As Represented By The Department Of Health And Human Services Measurements of multiple molecules using a CryoArray
DE03768629T1 (de) * 2002-11-05 2006-01-26 The Regents Of The University Of California, Oakland Verfahren und materialien zur untersuchung von mit glioblastomprogression assoziierten wegen
GB2424070B (en) * 2002-11-14 2007-06-27 Univ Nottingham Methods for preparing tumour marker proteins
EP1709152A4 (fr) 2003-12-15 2007-11-07 Univ California Signature moleculaire du suppresseur de tumeur pten
CA2596845C (fr) 2005-02-10 2016-11-08 Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Procede de diagnostic et de traitement du cancer au moyen de variants d'epissure de beta-catenine
AR053272A1 (es) * 2005-05-11 2007-04-25 Hoffmann La Roche Determinacion de responsivos a la quimioterapia
WO2006133460A2 (fr) * 2005-06-09 2006-12-14 Yale University Methodes de diagnostic et de traitement du cancer du sein d'apres le rapport her/er
CA2616874A1 (fr) 2005-08-03 2007-02-15 Ventana Medical Systems, Inc. Methodes predictives pour chimiotherapie cancereuse
EP1934378A4 (fr) 2005-09-19 2008-10-29 Veridex Llc Méthodes de diagnostic du cancer du pancréas
CN104749378A (zh) 2006-02-16 2015-07-01 文塔纳医疗***公司 用于癌症预后和病理分期的试剂和方法
ES2394224T3 (es) * 2006-05-05 2013-01-23 Yale University Uso de perfiles de localización subcelular como indicadores de prognóstico o predictivos
JP2010500577A (ja) 2006-08-07 2010-01-07 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム 癌の予後および予測シグナチャーのプロテオミクスパターン
US20100003247A1 (en) * 2006-10-27 2010-01-07 George Mason Intellectual Properties, Inc. Assay for metastatic colorectal cancer
CL2008000614A1 (es) * 2007-03-02 2008-09-05 Genentech Inc F Hoffmann La Ro Metodo para tratar un paciente con un tipo de cancer que expresa el receptor her3 a un nivel bajo mediante la administracion de un inhibidor de la dimerizacion de her.
NZ624816A (en) 2009-01-14 2015-07-31 Us Health Ratio based biomarkers and methods of use thereof
US20130210648A1 (en) 2009-01-14 2013-08-15 The United States of America, as Represented by the Secretary, Department of Health and Human Services Ratio based biomarkers and methods of use thereof
JP4782854B2 (ja) 2009-03-10 2011-09-28 ナショナル ヤン−ミン ユニバーシティ 癌患者の潜在的転移能、予後、又は全生存を予測する方法
CN106399506A (zh) 2009-10-26 2017-02-15 雅培分子公司 用于测定非小细胞肺癌预后的诊断方法
US20120052508A1 (en) 2010-08-27 2012-03-01 Rutgers, The State University Of New Jersey Genetic markers and diagnostic methods for resistance of breast cancer to hormonal therapies

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Campbell et al (British Journal of Cancer, 2008, 98: 931-940) *
Hayashi et al (Clin Cancer Res, 2008, 14(23): 7843-7849) *
Lee et al (Gynecologic Oncology, 2005, 99: 415-421) *
Saxena et al (American Journal of Hematology, 2004, 75: 22-33) *
Tockman et al (Cancer Res., 1992, 52:2711s-2718s) *
Xia et al (Clinical Cancer Research, 1999, 5: 4164-4174) *

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140037642A1 (en) * 2011-02-02 2014-02-06 Amgen Inc. Methods and compositions relating to inhibition of igf-1r
US8703127B2 (en) 2011-11-08 2014-04-22 Caldera Health Limited Methods and materials for the diagnosis of prostate cancers
US8632971B2 (en) 2011-11-08 2014-01-21 Caldera Health Limited Methods and materials for determining the efficacy of prostate cancer therapies
US8658164B2 (en) 2011-11-08 2014-02-25 Caldera Health Limited Methods and materials for the diagnosis of prostate cancers
US8658166B2 (en) 2011-11-08 2014-02-25 Caldera Health Limited Methods and materials for the diagnosis of prostate cancers
WO2013159016A1 (fr) * 2012-04-20 2013-10-24 University Of Connecticut Pipeline pour la conception rationnelle et l'interprétation de panels de biomarqueurs
WO2013163451A1 (fr) * 2012-04-27 2013-10-31 Bio-Rad Laboratories, Inc. Détermination quantitative et normalisation de protéines sans coloration
US10119961B2 (en) 2012-04-27 2018-11-06 Bio-Rad Laboratories, Inc. Stain-free protein quantification and normalization
US9606111B2 (en) 2012-04-27 2017-03-28 Bio-Rad Laboratories, Inc. Stain-free protein quantification and normalization
US10539556B2 (en) 2012-04-27 2020-01-21 Bio-Rad Laboratories, Inc. Stain-free protein quantification and normalization
EP3696283A1 (fr) * 2013-03-08 2020-08-19 MDxHealth Research B.V. Marqueurs moléculaires dans le cancer de la vessie
WO2015035250A3 (fr) * 2013-09-05 2015-11-19 Dendreon Corporation Réponse immunitaire humorale dirigée contre des antigènes tumoraux après traitement à l'aide d'une immunothérapie active spécifique d'antigènes du cancer et son association avec un résultat clinique amélioré
US10261091B2 (en) 2013-09-05 2019-04-16 Dendreon Corporation Humoral immune response against tumor antigens after treatment with a cancer antigen specific active immunotherapy and its association with improved clinical outcome
US11015190B2 (en) * 2013-09-17 2021-05-25 Lucence Life Sciences Pte Ltd Method of treating a patient having renal cancer
WO2015120416A1 (fr) * 2014-02-07 2015-08-13 The Johns Hopkins University Biomarqueurs pour l'évaluation d'un traitement sur des patients atteints d'un cancer
US20170016911A1 (en) * 2014-03-24 2017-01-19 Konica Minolta, Inc. Method for quantifying biological material based on multiple immunostaining
US9994912B2 (en) 2014-07-03 2018-06-12 Abbott Molecular Inc. Materials and methods for assessing progression of prostate cancer
US10604812B2 (en) 2014-07-03 2020-03-31 Abbott Molecular Inc. Materials and methods for assessing progression of prostate cancer
EP3530751A1 (fr) 2014-07-03 2019-08-28 Abbott Molecular Inc. Matériaux et procédés d'évaluation de la progression du cancer de la prostate
US11047860B2 (en) 2015-11-05 2021-06-29 Vanderbilt University Protein quantitation in multicellular tissue specimens
US20170168055A1 (en) * 2015-12-11 2017-06-15 Expression Pathology, Inc. SRM/MRM Assays
WO2017202962A1 (fr) 2016-05-24 2017-11-30 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés et compositions pharmaceutiques pour le traitement du cancer du poumon non à petites cellules (cbnpc) qui coexiste avec la bronchopneumopathie chronique obstructive (bpco)
WO2018046736A1 (fr) 2016-09-12 2018-03-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de prédiction du temps de survie de patients souffrant d'un cancer
WO2018055080A1 (fr) 2016-09-22 2018-03-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes et compositions pharmaceutiques permettant la reprogrammation de l'environnement immunitaire chez un sujet en ayant besoin
US11435360B2 (en) 2016-12-16 2022-09-06 The Brigham And Women's Hospital, Inc. System and sensor array
WO2018122249A1 (fr) 2016-12-28 2018-07-05 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes permettant de prédire le temps de survie de patients souffrant d'un cancer colorectal stable microsatellitaire
WO2018122245A1 (fr) 2016-12-28 2018-07-05 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de prédiction de la durée de survie de patients souffrant d'un cancer colorectal cms3
US11204355B2 (en) 2017-02-20 2021-12-21 Vanderbilt University Immune checkpoint molecular fitness profiling by mass spectrometry
WO2018160988A1 (fr) * 2017-03-03 2018-09-07 Expression Pathology, Inc. Profilage immunitaire de tissus tumoraux
WO2018202850A1 (fr) 2017-05-05 2018-11-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Médicaments ciblant les microtubules en tant qu'inhibiteurs de points de contrôle immunitaires et procédés de criblage de nouveaux inhibiteurs de points de contrôle immunitaires pour le traitement de cancers et de maladies infectieuses
US20210069295A1 (en) * 2017-11-10 2021-03-11 Stc. Unm Galectins control mtor in response to endomembrane damage and provide a mechanism and target for the treatment of autophagy-related diseases
WO2020141199A1 (fr) 2019-01-03 2020-07-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes et compositions pharmaceutiques pour améliorer les réponses immunitaires dépendantes des lymphocytes t cd8+ chez des sujets souffrant d'un cancer
EP4059569A1 (fr) 2019-01-03 2022-09-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) Procédés et compositions pharmaceutiques permettant d'améliorer des réponses immunitaires dépendant de lymphocyte t+cd8 chez des sujets souffrant d'un cancer
WO2020201362A2 (fr) 2019-04-02 2020-10-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes de prédiction et de prévention du cancer chez des patients ayant des lésions prémalignes
WO2020208060A1 (fr) 2019-04-09 2020-10-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Utilisation d'inhibiteurs de sk2 en association avec une thérapie de blocage de point de contrôle immunitaire pour le traitement du cancer
WO2020245155A1 (fr) 2019-06-03 2020-12-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de modulation d'un régime de traitement
US11906526B2 (en) 2019-08-05 2024-02-20 Seer, Inc. Systems and methods for sample preparation, data generation, and protein corona analysis
WO2021074391A1 (fr) 2019-10-17 2021-04-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de diagnostic d'adénocarcinomes du type intestinal nasal
WO2021170777A1 (fr) 2020-02-28 2021-09-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de diagnostic, de pronostic et de gestion du traitement du cancer du sein
WO2022002873A1 (fr) 2020-06-30 2022-01-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés pour prédire le risque de récidive et/ou de mort de patients souffrant d'un cancer solide après des traitements adjuvants pré-opératoires
WO2022002874A1 (fr) 2020-06-30 2022-01-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés pour prédire le risque de récidive et/ou de mort de patients souffrant d'un cancer solide après un traitement adjuvant préopératoire et une chirurgie radicale
WO2022020218A1 (fr) * 2020-07-19 2022-01-27 Aravive Inc Médthodes de diagnostic du cancer faisant appel à des récepteurs leurres axl
WO2022200303A1 (fr) 2021-03-23 2022-09-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés pour le diagnostic et le traitement de lymphomes t
CN114113611A (zh) * 2021-12-13 2022-03-01 郑州大学 一种用于肝癌诊断的生物标志物及检测试剂盒
WO2023144303A1 (fr) 2022-01-31 2023-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Cd38 en tant que biomarqueur et biocible dans des lymphomes t
WO2023175366A1 (fr) 2022-03-17 2023-09-21 Veracyte Méthodes de prédiction de la réponse à un traitement immunothérapeutique chez un patient atteint d'un cancer
WO2023198648A1 (fr) 2022-04-11 2023-10-19 Institut National de la Santé et de la Recherche Médicale Méthodes de diagnostic et de traitement de malignités des lymphocytes t
WO2023198874A1 (fr) 2022-04-15 2023-10-19 Institut National de la Santé et de la Recherche Médicale Méthodes pour le diagnostic et le traitement de lymphomes t
WO2024003310A1 (fr) 2022-06-30 2024-01-04 Institut National de la Santé et de la Recherche Médicale Méthodes de diagnostic et de traitement de la leucémie lymphoblastique aiguë
WO2024018046A1 (fr) 2022-07-22 2024-01-25 Institut National de la Santé et de la Recherche Médicale Garp utilisée en tant que biomarqueur et biocible dans des malignités de lymphocytes t
WO2024023283A1 (fr) 2022-07-29 2024-02-01 Institut National de la Santé et de la Recherche Médicale Lrrc33 en tant que biomarqueur et biocible dans des lymphomes t cutanés
WO2024079192A1 (fr) 2022-10-12 2024-04-18 Institut National de la Santé et de la Recherche Médicale Cd81 utilisé en tant que biomarqueur et cible biologique dans des malignités de lymphocytes t

Also Published As

Publication number Publication date
EP2667194A3 (fr) 2014-06-04
EP2667194A2 (fr) 2013-11-27
EP2667195A2 (fr) 2013-11-27
AU2016202107B2 (en) 2017-11-23
WO2010083252A2 (fr) 2010-07-22
EP2380025B1 (fr) 2013-09-11
IL233097A (en) 2017-01-31
IL214046A0 (en) 2011-08-31
JP2016035475A (ja) 2016-03-17
AU2016202107A1 (en) 2016-04-28
NZ624816A (en) 2015-07-31
IL233096A (en) 2017-01-31
CA2749601C (fr) 2020-07-21
IL233096A0 (en) 2014-07-31
NZ628463A (en) 2015-12-24
EP2667195B1 (fr) 2016-12-14
AU2010204741B2 (en) 2016-01-07
EP2667195A3 (fr) 2014-06-04
IL233095A0 (en) 2014-07-31
IL233098A (en) 2017-01-31
AU2010204741A1 (en) 2011-07-07
IL233097A0 (en) 2014-07-31
JP6502839B2 (ja) 2019-04-17
NZ606687A (en) 2014-08-29
JP5907732B2 (ja) 2016-04-26
WO2010083252A3 (fr) 2010-10-28
US20170115295A1 (en) 2017-04-27
JP2014041162A (ja) 2014-03-06
IL233095A (en) 2017-01-31
US20190113517A1 (en) 2019-04-18
CA2749601A1 (fr) 2010-07-22
IL233098A0 (en) 2014-07-31
JP2012515334A (ja) 2012-07-05
IL214046A (en) 2015-08-31
NZ593514A (en) 2013-03-28
US11237169B2 (en) 2022-02-01
EP2380025A2 (fr) 2011-10-26

Similar Documents

Publication Publication Date Title
US11237169B2 (en) Ratio based biomarkers and methods of use thereof
US20180074076A1 (en) Ratio based biomarkers and methods of use thereof
Mueller et al. Reverse phase protein microarrays advance to use in clinical trials
JP5750152B2 (ja) 肝細胞癌と関連するシグネチャ遺伝子を利用する検査方法、及び該方法に用いられるアレイ又はキット
Blokzijl et al. Profiling protein expression and interactions: proximity ligation as a tool for personalized medicine
KR20110065512A (ko) 암의 진단 방법 및 암 환자의 전반적 생존 및 무질병 생존의 측정 방법
US20080166733A1 (en) Method for determining the prognosis of cancer patients by measuring levels of bag expression
Zhang et al. Elevated growth differentiation factor 15 expression predicts poor prognosis in epithelial ovarian cancer patients
CA2694409A1 (fr) Procede de prediction d'une reponse au tamoxifene
WO2014151079A2 (fr) Biomarqueurs basés sur le rapport et leur procédés d'utilisation
US20140030254A1 (en) Signal pathway alterations and drug target elevations in primary metachronous metastatic colorectal cancer compared to non-metastatic disease
Lenggenhager et al. of Pancreatic Adenocarcinoma
Tseng Advances In Keratin 17 As A Cancer Biomarker: A Systematic Review
JP2017528699A (ja) 生体試料品質を評価するための方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNITED STATES OF AMERICA, AS REPRESENTED BY TH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HEWITT, STEPHEN M.;CHUNG, JOON-YONG;REEL/FRAME:023993/0117

Effective date: 20100225

AS Assignment

Owner name: THE UNITED STATES OF AMERICA, AS REPRESENTED BY TH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HEWITT, STEPHEN M.;CHUNG, JOON-YONG;REEL/FRAME:026593/0077

Effective date: 20100225

STCB Information on status: application discontinuation

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