US20130116150A1 - Lung Cancer Biomarkers and Uses Thereof - Google Patents

Lung Cancer Biomarkers and Uses Thereof Download PDF

Info

Publication number
US20130116150A1
US20130116150A1 US13/808,751 US201113808751A US2013116150A1 US 20130116150 A1 US20130116150 A1 US 20130116150A1 US 201113808751 A US201113808751 A US 201113808751A US 2013116150 A1 US2013116150 A1 US 2013116150A1
Authority
US
United States
Prior art keywords
individual
lung cancer
biomarker
biomarkers
biological sample
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
US13/808,751
Other languages
English (en)
Inventor
Sheri Wilcox
Deborah Ayers
Nebojsa Janjic
Larry Gold
Michael Riel-Mehan
Thale Jarvis
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.)
Madryn Health Partners Lp
Somalogic Inc
Original Assignee
Somalogic Inc
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 Somalogic Inc filed Critical Somalogic Inc
Priority to US13/808,751 priority Critical patent/US20130116150A1/en
Assigned to SOMALOGIC, INC. reassignment SOMALOGIC, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JANJIC, NEBOJSA, RIEL-MEHAN, MICHAEL, AYERS, DEBORAH, GOLD, LARRY, JARVIS, THALE, WILCOX, SHERI K.
Publication of US20130116150A1 publication Critical patent/US20130116150A1/en
Assigned to VISIUM HEALTHCARE PARTNERS, LP reassignment VISIUM HEALTHCARE PARTNERS, LP SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SOMALOGIC, INC.
Assigned to MADRYN HEALTH PARTNERS, LP reassignment MADRYN HEALTH PARTNERS, LP CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: VISIUM HEALTHCARE PARTNERS, LP
Assigned to SOMALOGIC, INC. reassignment SOMALOGIC, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: MADRYN HEALTH PARTNERS, LP, FORMERLY VISIUM HEALTHCARE PARTNERS, LP
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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • G06F19/10
    • 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
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • 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
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/20Allele or variant detection, e.g. single nucleotide polymorphism [SNP] detection
    • 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
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • G16B40/30Unsupervised data analysis
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/16Primer sets for multiplex assays
    • 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
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding

Definitions

  • the present application relates generally to the detection of biomarkers and the diagnosis of cancer in an individual and, more specifically, to one or more biomarkers, methods, devices, reagents, systems, and kits for diagnosing cancer, more particularly lung cancer, in an individual.
  • Lung cancer remains the most common cause of cancer-related mortality. This is true for both men and women. In 2005 in the United States lung cancer accounted for more deaths than breast cancer, prostate cancer, and colon cancer combined. In that year, 107,416 men and 89,271 women were diagnosed with lung cancer, and 90,139 men and 69,078 women died from lung cancer. Among men in the United States, lung cancer is the second most common cancer among white, black, Asian/Pacific Islander, American Indian/Alaska Native, and Hispanic men. Among women in the United States, lung cancer is the second most common cancer among white, black, and American Indian/Alaska Native women, and the third most common cancer among Asian/Pacific Islander and Hispanic women. For those who do not quit smoking, the probability of death from lung cancer is 15% and remains above 5% even for those who quit at age 50-59. The annual healthcare cost of lung cancer in the U.S. alone is $95 billion.
  • NSCLC non-small cell lung cancer
  • NSCLC nuclear cancer
  • squamous cell carcinoma large cell carcinoma
  • adenocarcinoma adenocarcinoma
  • Squamous cell carcinoma accounts for 25-30% of all lung cancers and is generally found in a proximal bronchus.
  • Early stage NSCLC tends to be localized, and if detected early it can often be treated by surgery with a favorable outcome and improved survival.
  • Other treatment options include radiation treatment, drug therapy, and a combination of these methods.
  • NSCLC is staged by the size of the tumor and its presence in other tissues including lymph nodes.
  • cancer cells are found in sputum samples or lavage samples and no tumor is detectable in the lungs.
  • stage 0 only the innermost lining of the lungs exhibit cancer cells and the tumor has not grown through the lining.
  • stage IA the cancer is considered invasive and has grown deep into the lung tissue but the tumor is less than 3 cm across. In this stage, the tumor is not found in the bronchus or lymph nodes.
  • stage IB the tumor is either larger than 3 cm across or has grown into the bronchus or pleura, but has not grown into the lymph nodes.
  • stage IIA the tumor is more than 3 cm across and has grown into the lymph nodes.
  • stage IIB the tumor has either been found in the lymph nodes and is greater than 3 cm across or grown into the bronchus or pleura; or the cancer is not in the lymph nodes but is found in the chest wall, diaphragm, pleura, bronchus, or tissue that surrounds the heart.
  • stage IIIA cancer cells are found in the lymph nodes near the lung and bronchi and in those between the lungs but on the side of the chest where the tumor is located.
  • Stage IIIB cancer cells are located on the opposite side of the chest from the tumor and in the neck. Other organs near the lungs may also have cancer cells and multiple tumors may be found in one lobe of the lungs.
  • stage IV tumors are found in more than one lobe of the same lung or both lungs and cancer cells are found in other parts of the body.
  • Lung nodules are relatively round lesions, or areas of abnormal tissue, located within the lung and may vary in size. Lung nodules may be benign or cancerous, but most are benign. If a nodule is below 4 mm the prevalence is only 1.5%, if 4-8 mm the prevalence is approximately 6%, and if above 20 mm the incidence is approximately 20%. For small and medium-sized nodules, the patient is advised to undergo a repeat scan within three months to a year. For many large nodules, the patient receives a biopsy (which is invasive and may lead to complications) even though most of these are benign.
  • diagnostic methods that can replace or complement CT are needed to reduce the number of surgical procedures conducted and minimize the risk of surgical complications.
  • methods are needed to detect lung cancer at its early stages to improve patient outcomes. Only 16% of lung cancer cases are diagnosed as localized, early stage cancer, where the 5-year survival rate is 46%, compared to 84% of those diagnosed at late stage, where the 5-year survival rate is only 13%. This demonstrates that relying on symptoms for diagnosis is not useful because many of them are common to other lung disease. These symptoms include a persistent cough, bloody sputum, chest pain, and recurring bronchitis or pneumonia.
  • Biomarker selection for a specific disease state involves first the identification of markers that have a measurable and statistically significant difference in a disease population compared to a control population for a specific medical application. Biomarkers can include secreted or shed molecules that parallel disease development or progression and readily diffuse into the blood stream from lung tissue or from distal tissues in response to a lesion.
  • Biomarker or set of biomarkers identified are generally clinically validated or shown to be a reliable indicator for the original intended use for which it was selected.
  • Biomarkers can include small molecules, peptides, proteins, and nucleic acids.
  • biomarkers and diagnose disease A variety of methods have been utilized in an attempt to identify biomarkers and diagnose disease.
  • protein-based markers these include two-dimensional electrophoresis, mass spectrometry, and immunoassay methods.
  • nucleic acid markers these include mRNA expression profiles, microRNA profiles, FISH, serial analysis of gene expression (SAGE), and large scale gene expression arrays.
  • Sandwich immunoassays do not scale to high content, and thus biomarker discovery using stringent sandwich immunoassays is not possible using standard array formats. Lastly, antibody reagents are subject to substantial lot variability and reagent instability. The instant platform for protein biomarker discovery overcomes this problem.
  • sample preparation required to run a sufficiently powered study designed to identify/discover statistically relevant biomarkers in a series of well-defined sample populations is extremely difficult, costly, and time consuming.
  • fractionation a wide range of variability can be introduced into the various samples. For example, a potential marker could be unstable to the process, the concentration of the marker could be changed, inappropriate aggregation or disaggregation could occur, and inadvertent sample contamination could occur and thus obscure the subtle changes anticipated in early disease.
  • biomarker discovery and detection methods using these technologies have serious limitations for the identification of diagnostic biomarkers. These limitations include an inability to detect low-abundance biomarkers, an inability to consistently cover the entire dynamic range of the proteome, irreproducibility in sample processing and fractionation, and overall irreproducibility and lack of robustness of the method. Further, these studies have introduced biases into the data and not adequately addressed the complexity of the sample populations, including appropriate controls, in terms of the distribution and randomization required to identify and validate biomarkers within a target disease population.
  • Biomarker research based on 2D gels or mass spectrometry supports these notions. Very few useful biomarkers have been identified through these approaches. However, it is usually overlooked that 2D gel and mass spectrometry measure proteins that are present in blood at approximately 1 nM concentrations and higher, and that this ensemble of proteins may well be the least likely to change with disease. Other than the instant biomarker discovery platform, proteomic biomarker discovery platforms that are able to accurately measure protein expression levels at much lower concentrations do not exist.
  • biochemical pathways for complex human biology. Many biochemical pathways culminate in or are started by secreted proteins that work locally within the pathology, for example growth factors are secreted to stimulate the replication of other cells in the pathology, and other factors are secreted to ward off the immune system, and so on. While many of these secreted proteins work in a paracrine fashion, some operate distally in the body.
  • One skilled in the art with a basic understanding of biochemical pathways would understand that many pathology-specific proteins ought to exist in blood at concentrations below (even far below) the detection limits of 2D gels and mass spectrometry. What must precede the identification of this relatively abundant number of disease biomarkers is a proteomic platform that can analyze proteins at concentrations below those detectable by 2D gels or mass spectrometry.
  • biomarkers methods, devices, reagents, systems, and kits that enable (a) the differentiation of benign pulmonary nodules from malignant pulmonary nodules; (b) the detection of lung cancer biomarkers; and (c) the diagnosis of lung cancer.
  • SOMAscan This technology, referred to as SOMAscan, is enabled by a new generation of slow off-rate aptamers (SOMAmers) that contain chemically modified nucleotides, which greatly expand the physicochemical diversity of the large randomized nucleic acid libraries from which the aptamers are selected (see U.S. Pat. No. 7,947,447).
  • SOMAmers slow off-rate aptamers
  • Such modifications which are compatible with SELEX, introduce functional groups into aptamers that are often found in protein-protein interaction, antibody-antigen interactions and interactions between small-molecule drugs with their protein targets. Overall, the use of these modifications expands the range of possible aptamer targets, improves their binding properties and facilitates selection of aptamers with slow dissociation rates.
  • proteins in complex matrices such as plasma are measured with a process that transforms a signature of protein concentrations into a corresponding signature of DNA aptamer concentrations, which is then quantified using a DNA microarray platform (Gold et al. Nature Precedings, http://precedings.nature.com/documents/4538/version/1 (2010)).
  • the assay leverages equilibrium binding and kinetic challenge. Both are carried out in solution, not on a surface, to take advantage of more favorable kinetics of binding and dissociation. In essence, the assay takes advantage of the dual nature of aptamers as both folded binding entities with defined shapes and unique sequences recognizable by specific hybridization probes.
  • the assay is capable of simultaneously measuring large numbers of proteins ranging from low to high abundance in serum.
  • samples from 1,326 subjects from four independent studies of non-small cell lung cancer (NSCLC) have been analyzed in long-term tobacco-exposed populations. More than 800 proteins in 15 ⁇ L of serum were measured and a 12-protein panel was developed that distinguishes NSCLC from controls with 91% sensitivity and 84% specificity in a training set and 89% sensitivity and 83% specificity in a blinded, independent verification set.
  • performance was similar for early and late stage NSCLC (Ostroff et al. Nature Precedings, http://precedings.nature.com/documents/4537/version/1 (2010)).
  • the present application demonstrates the utility of the newly discovered microarray platform technology to identify disease-related biomarkers from tissue.
  • the present application includes biomarkers, methods, reagents, devices, systems, and kits for the detection and diagnosis of cancer and more particularly, lung cancer from tissue.
  • the biomarkers of the present application were identified using a multiplex aptamer-based assay which is described in detail in Example 6.
  • this application describes a surprisingly large number of lung cancer biomarkers from tissue that are useful for the detection and diagnosis of lung cancer.
  • identifying these biomarkers over 800 proteins from a number of individual samples were measured, some of which were at concentrations in the low femtomolar range. This is about four orders of magnitude lower than biomarker discovery experiments done with 2D gels and/or mass spectrometry.
  • lung cancer biomarkers While certain of the described lung cancer biomarkers are useful alone for detecting and diagnosing lung cancer, methods are described herein for the grouping of multiple subsets of the lung cancer biomarkers that are useful as a panel of biomarkers. Once an individual biomarker or subset of biomarkers has been identified, the detection or diagnosis of lung cancer in an individual can be accomplished using any assay platform or format that is capable of measuring differences in the levels of the selected biomarker or biomarkers in a biological sample.
  • one or more biomarkers are provided for use either alone or in various combinations to diagnose lung cancer, particularly non-small cell lung cancer (NSCLC) or permit the differential diagnosis of pulmonary nodules as benign or malignant.
  • exemplary embodiments include the biomarkers provided in Table 18, which as noted above, were identified using a multiplex aptamer-based assay, as described generally in Example 1 and more specifically in Example 6.
  • the markers provided in Table 18 are useful in distinguishing benign nodules from cancerous nodules.
  • the markers provided in Table 18 are also useful in distinguishing asymptomatic smokers from smokers having lung cancer.
  • the biomarker is MMP-7.
  • the biomarker is MMP-12.
  • lung cancer biomarkers While certain of the described lung cancer biomarkers are useful alone for detecting and diagnosing lung cancer, methods are also described herein for the grouping of multiple subsets of the lung cancer biomarkers that are each useful as a panel of two or more biomarkers.
  • various embodiments of the instant application provide combinations comprising N biomarkers, wherein N is at least two biomarkers. In other embodiments, N is selected to be any number from 2-36 biomarkers.
  • N is selected to be any number from 2-7, 2-10, 2-15, 2-20, 2-25, 2-30, 2-36. In other embodiments, N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25, 3-30, 3-36. In other embodiments, N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25, 4-30, 4-36. In other embodiments, N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25, 5-30, 5-36. In other embodiments, N is selected to be any number from 6-10, 6-15, 6-20, 6-25, 6-30, 6-36.
  • N is selected to be any number from 7-10, 7-15, 7-20, 7-25, 7-30, 7-36. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25, 8-30, 8-36. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25, 9-30, 9-36. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25, 10-30, 10-36. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
  • a method for diagnosing lung cancer in an individual including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers provided in Table 18, wherein the individual is classified as having lung cancer based on the at least one biomarker value.
  • a method for diagnosing lung cancer in an individual including detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 18, wherein the likelihood of the individual having lung cancer is determined based on the biomarker values.
  • a method for screening smokers for lung cancer including detecting, in a biological sample from an individual who is a smoker, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 18, wherein the individual is classified as having lung cancer, or the likelihood of the individual having lung cancer is determined, based on the at least one biomarker value.
  • a method for diagnosing that an individual does not have lung cancer including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 18, wherein the individual is classified as not having lung cancer based on the at least one biomarker value.
  • a computer-implemented method for indicating a likelihood of lung cancer.
  • the method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, selected from the group of biomarkers set forth in Table 18; performing with the computer a classification of each of the biomarker values; and indicating a likelihood that the individual has lung cancer based upon a plurality of classifications.
  • a computer-implemented method for classifying an individual as either having or not having lung cancer.
  • the method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers provided in Table 18; performing with the computer a classification of each of the biomarker values; and indicating whether the individual has lung cancer based upon a plurality of classifications.
  • a computer program product for indicating a likelihood of lung cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, in the biological sample selected from the group of biomarkers set forth in Table 18; and code that executes a classification method that indicates a likelihood that the individual has lung cancer as a function of the biomarker values.
  • a computer program product for indicating a lung cancer status of an individual.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers in the biological sample selected from the group of biomarkers provided in Table 18; and code that executes a classification method that indicates a lung cancer status of the individual as a function of the biomarker values.
  • a computer-implemented method for indicating a likelihood of lung cancer.
  • the method comprises retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers set forth in Table 18; performing with the computer a classification of the biomarker value; and indicating a likelihood that the individual has lung cancer based upon the classification.
  • a computer-implemented method for classifying an individual as either having or not having lung cancer.
  • the method comprises retrieving from a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers provided in Table 18; performing with the computer a classification of the biomarker value; and indicating whether the individual has lung cancer based upon the classification.
  • a computer program product for indicating a likelihood of lung cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers set forth in Table 18; and code that executes a classification method that indicates a likelihood that the individual has lung cancer as a function of the biomarker value.
  • a computer program product for indicating a lung cancer status of an individual.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers provided in Table 18; and code that executes a classification method that indicates a lung cancer status of the individual as a function of the biomarker value.
  • exemplary embodiments include the biomarkers provided in Table 20, which as noted above, were identified using a multiplex aptamer-based assay, as described generally in Example 1 and more specifically in Example 6.
  • the markers provided in Table 20 are useful in distinguishing benign nodules from cancerous nodules.
  • the markers provided in Table 20 are also useful in distinguishing asymptomatic smokers from smokers having lung cancer.
  • N is selected to be any number from 2-25 biomarkers.
  • the markers provided in Table 20 have been determined to be useful in both tissue and serum samples.
  • N is selected to be any number from 2-7, 2-10, 2-15, 2-20, 2-25. In other embodiments, N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25. In other embodiments, N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25. In other embodiments, N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25. In other embodiments, N is selected to be any number from 6-10, 6-15, 6-20, 6-25. In other embodiments, N is selected to be any number from 7-10, 7-15, 7-20, 7-25. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
  • a method for diagnosing lung cancer in an individual including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers provided in Table 20, wherein the individual is classified as having lung cancer based on the at least one biomarker value.
  • a method for diagnosing lung cancer in an individual including detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 20, wherein the likelihood of the individual having lung cancer is determined based on the biomarker values.
  • a method for screening smokers for lung cancer including detecting, in a biological sample from an individual who is a smoker, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 20, wherein the individual is classified as having lung cancer, or the likelihood of the individual having lung cancer is determined, based on the at least one biomarker value.
  • a method for diagnosing that an individual does not have lung cancer including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 20, wherein the individual is classified as not having lung cancer based on the at least one biomarker value.
  • a computer-implemented method for indicating a likelihood of lung cancer.
  • the method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, selected from the group of biomarkers set forth in Table 20; performing with the computer a classification of each of the biomarker values; and indicating a likelihood that the individual has lung cancer based upon a plurality of classifications.
  • a computer-implemented method for classifying an individual as either having or not having lung cancer.
  • the method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers provided in Table 20; performing with the computer a classification of each of the biomarker values; and indicating whether the individual has lung cancer based upon a plurality of classifications.
  • a computer program product for indicating a likelihood of lung cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, in the biological sample selected from the group of biomarkers set forth in Table 20; and code that executes a classification method that indicates a likelihood that the individual has lung cancer as a function of the biomarker values.
  • a computer program product for indicating a lung cancer status of an individual.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers in the biological sample selected from the group of biomarkers provided in Table 20; and code that executes a classification method that indicates a lung cancer status of the individual as a function of the biomarker values.
  • a computer-implemented method for indicating a likelihood of lung cancer.
  • the method comprises retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers set forth in Table 20; performing with the computer a classification of the biomarker value; and indicating a likelihood that the individual has lung cancer based upon the classification.
  • a computer-implemented method for classifying an individual as either having or not having lung cancer.
  • the method comprises retrieving from a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers provided in Table 20; performing with the computer a classification of the biomarker value; and indicating whether the individual has lung cancer based upon the classification.
  • a computer program product for indicating a likelihood of lung cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers set forth in Table 20; and code that executes a classification method that indicates a likelihood that the individual has lung cancer as a function of the biomarker value.
  • a computer program product for indicating a lung cancer status of an individual.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers provided in Table 20; and code that executes a classification method that indicates a lung cancer status of the individual as a function of the biomarker value.
  • exemplary embodiments include the biomarkers provided in Table 21, which were identified using a multiplex aptamer-based assay, as described generally in Example 1 and more specifically in Examples 2 and 6.
  • the markers provided in Table 21 are useful in distinguishing benign nodules from cancerous nodules.
  • the markers provided in Table 21 are also useful in distinguishing asymptomatic smokers from smokers having lung cancer.
  • N is selected to be any number from 2-86 biomarkers. All of the biomarkers included in Table 21 are useful in providing the information being sought in both tissue and serum samples.
  • N is selected to be any number from 2-7, 2-10, 2-15, 2-20, 2-25, 2-30, 2-35, 2-40, 2-45, 2-50, 2-55, 2-60, 2-65, 2-70, 2-75, 2-80, or 2-86.
  • N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25, 3-30, 3-35, 3-40, 3-45, 3-50, 3-55, 3-60, 3-65, 3-70, 3-75, 3-80, or 3-86.
  • N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25, 4-30, 4-35, 4-40, 4-45, 4-50, 4-55, 4-60, 4-65, 4-70, 4-75, 4-80, or 4-86.
  • N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25, 5-30, 5-35, 5-40, 5-45, 5-50, 5-55, 5-60, 5-65, 5-70, 5-75, 5-80, or 5-86.
  • N is selected to be any number from 6-10, 6-15, 6-20, 6-25, 6-30, 6-35, 6-40, 6-45, 6-50, 6-55, 6-60, 6-65, 6-70, 6-75, 6-80, or 6-86.
  • N is selected to be any number from 7-10, 7-15, 7-20, 7-25, 7-30, 7-35, 7-40, 7-45, 7-50, 7-55, 7-60, 7-65, 7-70, 7-75, 7-80, or 7-86. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25, 8-30, 8-35, 8-40, 8-45, 8-50, 8-55, 8-60, 8-65, 8-70, 8-75, 8-80, or 8-86.
  • N is selected to be any number from 9-15, 9-20, 9-25, 9-30, 9-35, 9-40, 9-45, 9-50, 9-55, 9-60, 9-65, 9-70, 9-75, 9-80, or 9-86. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25, 10-30, 10-35, 10-40, 10-45, 10-50, 10-55, 10-60, 10-65, 10-70, 10-75, 10-80, or 10-86. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
  • a method for diagnosing lung cancer in an individual including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers provided in Table 21, wherein the individual is classified as having lung cancer based on the at least one biomarker value.
  • a method for diagnosing lung cancer in an individual including detecting, in a biological sample from an individual, biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers set forth in Table 21, wherein the likelihood of the individual having lung cancer is determined based on the biomarker values.
  • a method for screening smokers for lung cancer including detecting, in a biological sample from an individual who is a smoker, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 21, wherein the individual is classified as having lung cancer, or the likelihood of the individual having lung cancer is determined, based on the at least one biomarker value.
  • a method for diagnosing that an individual does not have lung cancer including detecting, in a biological sample from an individual, at least one biomarker value corresponding to at least one biomarker selected from the group of biomarkers set forth in Table 21, wherein the individual is classified as not having lung cancer based on the at least one biomarker value.
  • a computer-implemented method for indicating a likelihood of lung cancer.
  • the method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, selected from the group of biomarkers set forth in Table 21; performing with the computer a classification of each of the biomarker values; and indicating a likelihood that the individual has lung cancer based upon a plurality of classifications.
  • a computer-implemented method for classifying an individual as either having or not having lung cancer.
  • the method comprises: retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises biomarker values that each correspond to one of at least N biomarkers selected from the group of biomarkers provided in Table 21; performing with the computer a classification of each of the biomarker values; and indicating whether the individual has lung cancer based upon a plurality of classifications.
  • a computer program product for indicating a likelihood of lung cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers, wherein N is as defined above, in the biological sample selected from the group of biomarkers set forth in Table 21; and code that executes a classification method that indicates a likelihood that the individual has lung cancer as a function of the biomarker values.
  • a computer program product for indicating a lung cancer status of an individual.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises biomarker values that each correspond to one of at least N biomarkers in the biological sample selected from the group of biomarkers provided in Table 21; and code that executes a classification method that indicates a lung cancer status of the individual as a function of the biomarker values.
  • a computer-implemented method for indicating a likelihood of lung cancer.
  • the method comprises retrieving on a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers set forth in Table 21; performing with the computer a classification of the biomarker value; and indicating a likelihood that the individual has lung cancer based upon the classification.
  • a computer-implemented method for classifying an individual as either having or not having lung cancer.
  • the method comprises retrieving from a computer biomarker information for an individual, wherein the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers provided in Table 21; performing with the computer a classification of the biomarker value; and indicating whether the individual has lung cancer based upon the classification.
  • a computer program product for indicating a likelihood of lung cancer.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers set forth in Table 21; and code that executes a classification method that indicates a likelihood that the individual has lung cancer as a function of the biomarker value.
  • a computer program product for indicating a lung cancer status of an individual.
  • the computer program product includes a computer readable medium embodying program code executable by a processor of a computing device or system, the program code comprising: code that retrieves data attributed to a biological sample from an individual, wherein the data comprises a biomarker value corresponding to a biomarker in the biological sample selected from the group of biomarkers provided in Table 21; and code that executes a classification method that indicates a lung cancer status of the individual as a function of the biomarker value.
  • At least one of said N biomarkers selected from Table 21 in each of the above methods is a biomarker selected from the Table 20.
  • said biomarker selected from Table 20 is MMP-12.
  • a method for diagnosing lung cancer including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of biomarkers selected from the group of panels set forth in Tables 22-25 wherein a classification of the biomarker values indicates that the individual has lung cancer.
  • a method for diagnosing an absence of lung cancer including detecting, in a biological sample from an individual, biomarker values that each correspond to a biomarker on a panel of biomarkers selected from the group of panels provided in Tables 22-25, wherein a classification of the biomarker values indicates an absence of lung cancer in the individual.
  • FIG. 1A is a flowchart for an exemplary method for detecting lung cancer in a biological sample.
  • FIG. 1B is a flowchart for an exemplary method for detecting lung cancer in a biological sample using a na ⁇ ve Bayes classification method.
  • FIG. 2 shows a ROC curve for a single biomarker, SCFsR, using a na ⁇ ve Bayes classifier for a test that detects lung cancer in asymptomatic smokers.
  • FIG. 3 shows ROC curves for biomarker panels of from one to ten biomarkers using na ⁇ ve Bayes classifiers for a test that detects lung cancer in asymptomatic smokers.
  • FIG. 4 illustrates the increase in the classification score (specificity+sensitivity) as the number of biomarkers is increased from one to ten using na ⁇ ve Bayes classification for a benign nodule-lung cancer panel.
  • FIG. 5 shows the measured biomarker distributions for SCFsR as a cumulative distribution function (cdf) in log-transformed RFU for the benign nodule control group (solid line) and the lung cancer disease group (dotted line) along with their curve fits to a normal cdf (dashed lines) used to train the na ⁇ ve Bayes classifiers
  • FIG. 6 illustrates an exemplary computer system for use with various computer-implemented methods described herein.
  • FIG. 7 is a flowchart for a method of indicating the likelihood that an individual has lung cancer in accordance with one embodiment.
  • FIG. 8 is a flowchart for a method of indicating the likelihood that an individual has lung cancer in accordance with one embodiment.
  • FIG. 9 illustrates an exemplary aptamer assay that can be used to detect one or more lung cancer biomarkers in a biological sample.
  • FIG. 10 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between NSCLC and benign nodules from an aggregated set of potential biomarkers.
  • FIG. 11 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between NSCLC and asymptomatic smokers from an aggregated set of potential biomarkers.
  • FIG. 12 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between NSCLC and benign nodules from a site-consistent set of potential biomarkers.
  • FIG. 13 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between NSCLC and asymptomatic smokers from a site-consistent set of potential biomarkers.
  • FIG. 14 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between NSCLC and benign nodules from a set of potential biomarkers resulting from a combination of aggregated and site-consistent markers.
  • FIG. 15 shows a histogram of frequencies for which biomarkers were used in building classifiers to distinguish between NSCLC and asymptomatic smokers from a set of potential biomarkers resulting from a combination of aggregated and site-consistent markers.
  • FIG. 16 shows gel images resulting from pull-down experiments that illustrate the specificity of aptamers as capture reagents for the proteins LBP, C9 and IgM.
  • lane 1 is the eluate from the Streptavidin-agarose beads
  • lane 2 is the final eluate
  • lane is a MW marker lane (major bands are at 110, 50, 30, 15, and 3.5 kDa from top to bottom).
  • FIG. 17A shows a pair of histograms summarizing all possible single protein na ⁇ ve Bayes classifier scores (sensitivity+specificity) using the biomarkers set forth in Table 1, Col 5 (solid) and a set of random markers (dotted).
  • FIG. 17B shows a pair of histograms summarizing all possible two-protein protein na ⁇ ve Bayes classifier scores (sensitivity+specificity) using the biomarkers set forth in Table 1, Col 5 (solid) and a set of random markers (dotted).
  • FIG. 17C shows a pair of histograms summarizing all possible three-protein na ⁇ ve Bayes classifier scores (sensitivity+specificity) using the biomarkers set forth in Table 1, Col 5 (solid) and a set of random markers (dotted).
  • FIG. 18A shows a pair of histograms summarizing all possible single protein na ⁇ ve Bayes classifier scores (sensitivity+specificity) using the biomarkers set forth in Table 1, Col 6 (solid) and a set of random markers (dotted).
  • FIG. 18B shows a pair of histograms summarizing all possible two-protein protein na ⁇ ve Bayes classifier scores (sensitivity+specificity) using the biomarkers set forth in Table 1, Col 6 (solid) and a set of random markers (dotted).
  • FIG. 18C shows a pair of histograms summarizing all possible three-protein na ⁇ ve Bayes classifier scores (sensitivity+specificity) using the biomarkers set forth in Table 1, Col 6 (solid) and a set of random markers (dotted).
  • FIG. 19A shows the sensitivity+specificity score for na ⁇ ve Bayes classifiers using from 2-10 markers selected from the full panel ( ⁇ ) and the scores obtained by dropping the best 5 ( ⁇ ), 10 ( ⁇ ) and 15 (x) markers during classifier generation for the benign nodule control group.
  • FIG. 19B shows the sensitivity+specificity score for na ⁇ ve Bayes classifiers using from 2-10 markers selected from the full panel ( ⁇ ) and the scores obtained by dropping the best 5 ( ⁇ ), 10 ( ⁇ ) and 15 (x) markers during classifier generation for the smoker control group.
  • FIG. 20A shows a set of ROC curves modeled from the data in Tables 38 and 39 for panels of from one to five markers.
  • FIG. 20B shows a set of ROC curves computed from the training data for panels of from one to five markers as in FIG. 19A .
  • FIG. 21 shows relative changes in protein expression for 813 proteins from eight NSCLC resection samples between adjacent and distant tissue ( FIG. 21A ), tumor and adjacent tissue ( FIG. 21B ) and tumor and distant tissue ( FIG. 21C ) expressed as log 2 median ratios.
  • FIG. 22 show a heat map of protein levels in tumor tissue samples.
  • the samples are arranged in columns and are separated into distant, adjacent, and tumor samples. Within each tissue type, the samples are separated into adenocarcinoma (AC) and squamous cell carcinoma (SCC). The numbers above each column correspond to patient codes.
  • the proteins are displayed in rows and were ordered using hierarchial clustering.
  • FIG. 23 depicts proteins with increased levels in tumor tissue compared with adjacent or distal tissue.
  • FIG. 24 depicts proteins with decreased levels in tumor tissue compared with adjacent or distal tissue from the eight NSCLC samples used in this study.
  • FIG. 25 shows SOMAmer histochemistry on frozen tissue sections for selected biomarkers detected in this study.
  • A Thrombospondin-2 (red) staining the fibrocollagenous matrix surrounding a tumor nest.
  • B Corresponding normal lung specimen stained with Thrombospondin-2 SOMAmer (red).
  • C Macrophage Mannose Receptor SOMAmer (red) staining scattered macrophages in a lung adenocarcinoma.
  • D Macrophage Mannose Receptor SOMAmer (red) staining numerous alveolar macrophages in a section of normal lung parenchyma.
  • FIG. 26 shows changes in protein expression in NSCLC tissue compared to serum.
  • the top two panels show the log 2 ratio (LR) derived from serum samples versus log ratios derived from adjacent tissue and distant tissue, respectively.
  • the bottom four panels feature zoomed portions of plots above, indicated by the color of the plot (green for decreased and red for increased expression compared to non-tumor tissue).
  • Analytes shown in FIGS. 23 and 24 have been labeled and analytes mentioned in the publication on the serum samples are shown in filled red symbols red.
  • FIG. 27 depicts thrombospondin-2 histochemical identification in tissue samples.
  • Thrombospondin-2 is identified in a serial frozen section of a single lung carcinoma specimen by (A) a home-made rabbit polyclonal thrombospondin-2 polyclonal antibody, (B) the pre-immune serum from rabbits used to make the home-made polyclonal antibody, (C) a commercial (Novus) rabbit polyclonal thrombospondin-2 antibody, and (D) the thrombospondin-2 SOMAmer.
  • the thrombospondin-2 SOMAmer was then used to stain frozen sections of normal and malignant lung tissue, with standard Avidin-Biotin-Peroxidase color development, to demonstrate different morphologic distributions: (E) Strong staining of the fibrotic stroma surrounding tumor nests, with minimal cytosolic staining of carcinoma cells, (F) Strong staining of the fibrotic stroma surrounding tumor nests in a mucinous adenocarcinoma, with no significant staining of the carcinoma cells, (G) normal lung tissue, showing strong cytosolic staining of bronchial epithelium and scattered alveolar macrophages, and (H) strong cytosolic staining of an adenocarcinoma, with no significant staining of the non-fibrotic, predominantly inflammatory stroma.
  • the term “about” represents an insignificant modification or variation of the numerical value such that the basic function of the item to which the numerical value relates is unchanged.
  • the terms “comprises,” “comprising,” “includes,” “including,” “contains,” “containing,” and any variations thereof, are intended to cover a non-exclusive inclusion, such that a process, method, product-by-process, or composition of matter that comprises, includes, or contains an element or list of elements does not include only those elements but may include other elements not expressly listed or inherent to such process, method, product-by-process, or composition of matter.
  • the present application includes biomarkers, methods, devices, reagents, systems, and kits for the detection and diagnosis of lung cancer.
  • one or more biomarkers are provided for use either alone or in various combinations to diagnose lung cancer, permit the differential diagnosis of pulmonary nodules as benign or malignant, monitor lung cancer recurrence, or address other clinical indications.
  • said biomarker(s) can be used in determining information about lung cancer in an individual such as, prognosis, cancer classification, prediction of disease risk or selection of treatment.
  • exemplary embodiments include the biomarkers provided in Tables 18, and 21, which were identified using a multiplex aptamer-based assay that is described generally in Example 1 and more specifically in Examples 2 and 6. Each of the biomarkers is useful in assaying any type of sample as defined below.
  • Table 1 Col. 2 sets forth the findings obtained from analyzing hundreds of individual blood samples from NSCLC cancer cases, and hundreds of equivalent individual blood samples from smokers and from individuals diagnosed with benign lung nodules.
  • the smoker and benign nodule groups were designed to match the populations with which a lung cancer diagnostic test can have the most benefit. (These cases and controls were obtained from multiple clinical sites to mimic the range of real world conditions under which such a test can be applied).
  • the potential biomarkers were measured in individual samples rather than pooling the disease and control blood; this allowed a better understanding of the individual and group variations in the phenotypes associated with the presence and absence of disease (in this case lung cancer). Since over 800 protein measurements were made on each sample, and several hundred samples from each of the disease and the control populations were individually measured, Table 1, Col. 2 resulted from an analysis of an uncommonly large set of data. The measurements were analyzed using the methods described in the section, “Classification of Biomarkers and Calculation of Disease Scores” herein.
  • Table 1, Col. 2 lists the biomarkers found to be useful in distinguishing samples obtained from individuals with NSCLC from “control” samples obtained from smokers and individuals with benign lung nodules.
  • Using a multiplex aptamer assay as described herein thirty-eight biomarkers were discovered that distinguished the samples obtained from individuals who had lung cancer from the samples obtained from individuals in the smoker control group (see Table 1, Col. 6).
  • using a multiplex aptamer assay forty biomarkers were discovered that distinguished samples obtained from individuals with NSCLC from samples obtained from people who had benign lung nodules (see Table 1, Col. 5).
  • the two lists of 38 and 40 biomarkers are comprised of 61 unique biomarkers, because there is considerable overlap between the list of biomarkers for distinguishing NSCLC from benign nodules and the list for distinguishing NSCLC from smokers who do not have lung cancer.
  • Table 18 sets forth the findings obtained from analyzing eight individual tissue samples of smokers diagnosed with NSCLC as described in Example 6. All of the patients were smokers ranging from 47 to 75 years old and covering NSCLC stages 1A through 3B. Three samples were obtained from each individual: tumor tissue, adjacent healthy tissue (within 1 cm of the tumor) and distant uninvolved lung tissue. The samples were chosen to match the populations with which a lung cancer diagnostic test can have the most benefit. The potential biomarkers were measured in individual samples rather than pooling the disease and control tissue; this allowed a better understanding of the individual and group variations in the phenotypes associated with the presence and absence of disease (in this case lung cancer). The measurements were analyzed using the Mann-Whitney test.
  • Table 18 lists the biomarkers found to be useful in distinguishing samples obtained from individuals with NSCLC from “control” samples obtained from adjacent and distal uninvolved lung tissue obtained from the same individuals.
  • thirty-six biomarkers were discovered that distinguished the tumor tissue samples from samples obtained from adjacent and distal lung tissue in individuals who had been diagnosed with NSCLC.
  • Table 1 col. 2, it can be seen that eleven of the biomarkers overlap those identified in serum samples as described in Example 2.
  • An additional marker which was not measured in the original serum profiling, MMP-12 has since been found to be a useful biomarker in both serum and in tissue.
  • Table 21 provides a list of the total number of biomarkers (eighty-six) identified in both the serum and tumor tissue samples combined.
  • Table 20 provides a list of the biomarkers identified which were unique to the tumor tissue samples (twenty-five).
  • N biomarkers are at least two biomarkers.
  • N is selected from 2-86 biomarkers (Table 21); 2-36 biomarkers (Table 18) or 2-25 biomarkers (Table 20).
  • N is selected from 2-86 (Table 21) and at least one of said N biomarkers is MMP-12.
  • N is selected from 2-25 (Table 20) and at least one of said N biomarkers is MMP-12.
  • Representative panels of 2-5 biomarkers including MMP-12 as one of the markers are set forth in Tables 22-25.
  • the biomarkers are selected from those listed in Table 18 and N is selected to be any number from 2-7, 2-10, 2-15, 2-20, 2-25, 2-30, 2-36. In other embodiments, N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25, 3-30, 3-36. In other embodiments, N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25, 4-30, 4-36. In other embodiments, N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25, 5-30, 5-36. In other embodiments, N is selected to be any number from 6-10, 6-15, 6-20, 6-25, 6-30, 6-36.
  • N is selected to be any number from 7-10, 7-15, 7-20, 7-25, 7-30, 7-36. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25, 8-30, 8-36. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25, 9-30, 9-36. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25, 10-30, 10-36. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
  • the biomarkers are selected from those listed in Table 20 and N is selected to be any number from 2-7, 2-10, 2-15, 2-20, 2-25. In other embodiments, N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25. In other embodiments, N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25. In other embodiments, N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25. In other embodiments, N is selected to be any number from 6-10, 6-15, 6-20, 6-25. In other embodiments, N is selected to be any number from 7-10, 7-15, 7-20, 7-25.
  • N is selected to be any number from 8-10, 8-15, 8-20, 8-25. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25. In other embodiments, N is selected to be any number from 9-15, 9-20, 9-25. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
  • the biomarkers are selected from those listed in Table 21 and N is selected to be any number from 2-7, 2-10, 2-15, 2-20, 2-25, 2-30, 2-35, 2-40, 2-45, 2-50, 2-55, 2-60, 2-65, 2-70, 2-75, 2-80, or 2-86.
  • N is selected to be any number from 3-7, 3-10, 3-15, 3-20, 3-25, 3-30, 3-35, 3-40, 3-45, 3-50, 3-55, 3-60, 3-65, 3-70, 3-75, 3-80, or 3-86.
  • N is selected to be any number from 4-7, 4-10, 4-15, 4-20, 4-25, 4-30, 4-35, 4-40, 4-45, 4-50, 4-55, 4-60, 4-65, 4-70, 4-75, 4-80, or 4-86.
  • N is selected to be any number from 5-7, 5-10, 5-15, 5-20, 5-25, 5-30, 5-35, 5-40, 5-45, 5-50, 5-55, 5-60, 5-65, 5-70, 5-75, 5-80, or 5-86.
  • N is selected to be any number from 6-10, 6-15, 6-20, 6-25, 6-30, 6-35, 6-40, 6-45, 6-50, 6-55, 6-60, 6-65, 6-70, 6-75, 6-80, or 6-86.
  • N is selected to be any number from 7-10, 7-15, 7-20, 7-25, 7-30, 7-35, 7-40, 7-45, 7-50, 7-55, 7-60, 7-65, 7-70, 7-75, 7-80, or 7-86. In other embodiments, N is selected to be any number from 8-10, 8-15, 8-20, 8-25, 8-30, 8-35, 8-40, 8-45, 8-50, 8-55, 8-60, 8-65, 8-70, 8-75, 8-80, or 8-86.
  • N is selected to be any number from 9-15, 9-20, 9-25, 9-30, 9-35, 9-40, 9-45, 9-50, 9-55, 9-60, 9-65, 9-70, 9-75, 9-80, or 9-86. In other embodiments, N is selected to be any number from 10-15, 10-20, 10-25, 10-30, 10-35, 10-40, 10-45, 10-50, 10-55, 10-60, 10-65, 10-70, 10-75, 10-80, or 10-86. It will be appreciated that N can be selected to encompass similar, but higher order, ranges.
  • the number of biomarkers useful for a biomarker subset or panel is based on the sensitivity and specificity value for the particular combination of biomarker values.
  • sensitivity and “specificity” are used herein with respect to the ability to correctly classify an individual, based on one or more biomarker values detected in their biological sample, as having lung cancer or not having lung cancer.
  • Sensitivity indicates the performance of the biomarker(s) with respect to correctly classifying individuals that have lung cancer.
  • Specificity indicates the performance of the biomarker(s) with respect to correctly classifying individuals who do not have lung cancer.
  • 85% specificity and 90% sensitivity for a panel of markers used to test a set of control samples and lung cancer samples indicates that 85% of the control samples were correctly classified as control samples by the panel, and 90% of the lung cancer samples were correctly classified as lung cancer samples by the panel.
  • the desired or preferred minimum value can be determined as described in Example 3.
  • lung cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to at least one of the biomarkers MMP-7, MMP-12, or IGFBP-2 and at least N additional biomarkers selected from the list of biomarkers in Table 21, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • lung cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarkers MMP-7, MMP-12, or IGFBP-2 and one of at least N additional biomarkers selected from the list of biomarkers in Table 21, wherein N equals 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13.
  • lung cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker MMP-7 and one of at least N additional biomarkers selected from the list of biomarkers in Table 21, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • lung cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker MMP-12 and one of at least N additional biomarkers selected from the list of biomarkers in Table 21, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • lung cancer is detected or diagnosed in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker IGFBP-2 and one of at least N additional biomarkers selected from the list of biomarkers in Table 21, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • the lung cancer biomarkers identified herein represent a relatively large number of choices for subsets or panels of biomarkers that can be used to effectively detect or diagnose lung cancer. Selection of the desired number of such biomarkers depends on the specific combination of biomarkers chosen. It is important to remember that panels of biomarkers for detecting or diagnosing lung cancer may also include biomarkers not found in Tables 18, 20 or 21, and that the inclusion of additional biomarkers not found in Tables 18, 20 or 21 may reduce the number of biomarkers in the particular subset or panel that is selected from Tables 18, 20 or 21. The number of biomarkers from Tables 18, 20 or 21 used in a subset or panel may also be reduced if additional biomedical information is used in conjunction with the biomarker values to establish acceptable sensitivity and specificity values for a given assay.
  • Another factor that can affect the number of biomarkers to be used in a subset or panel of biomarkers is the procedures used to obtain biological samples from individuals who are being diagnosed for lung cancer. In a carefully controlled sample procurement environment, the number of biomarkers necessary to meet desired sensitivity and specificity values will be lower than in a situation where there can be more variation in sample collection, handling and storage. In developing the list of biomarkers set forth in Tables 18, 20 or 21, multiple sample collection sites were utilized to collect data for classifier training. This provides for more robust biomarkers that are less sensitive to variations in sample collection, handling and storage, but can also require that the number of biomarkers in a subset or panel be larger than if the training data were all obtained under very similar conditions.
  • a biological sample is obtained from an individual or individuals of interest.
  • the biological sample is then assayed to detect the presence of one or more (N) biomarkers of interest and to determine a biomarker value for each of said N biomarkers (referred to in FIG. 1B as marker RFU).
  • N one or more biomarkers of interest
  • RFU a biomarker value for each of said N biomarkers
  • lung may be interchangeably referred to as “pulmonary”.
  • smoke refers to an individual who has a history of tobacco smoke inhalation.
  • Bio sample “sample”, and “test sample” are used interchangeably herein to refer to any material, biological fluid, tissue, or cell obtained or otherwise derived from an individual.
  • a blood sample can be fractionated into serum or into fractions containing particular types of blood cells, such as red blood cells or white blood cells (leukocytes).
  • a sample can be a combination of samples from an individual, such as a combination of a tissue and fluid sample.
  • biological sample also includes materials containing homogenized solid material, such as from a stool sample, a tissue sample, or a tissue biopsy, for example.
  • biological sample also includes materials derived from a tissue culture or a cell culture.
  • any suitable methods for obtaining a biological sample can be employed; exemplary methods include, e.g., phlebotomy, swab (e.g., buccal swab), and a fine needle aspirate biopsy procedure.
  • tissue susceptible to fine needle aspiration include lymph node, lung, lung washes, BAL (bronchoalveolar lavage), thyroid, breast, and liver.
  • Samples can also be collected, e.g., by micro dissection (e.g., laser capture micro dissection (LCM) or laser micro dissection (LMD)), bladder wash, smear (e.g., a PAP smear), or ductal lavage.
  • a “biological sample” obtained or derived from an individual includes any such sample that has been processed in any suitable manner after being obtained from the individual.
  • tissue sample refers to a certain subset of the biological samples described above. According to this definition, tissues are collections of macromolecules in a heterogeneous environment. As used herein, tissue refers to a single cell type, a collection of cell types, an aggregate of cells, or an aggregate of macromolecules. Tissues are generally a physical array of macromolecules that can be either fluid or rigid, both in terms of structure and composition. Extracellular matrix is an example of a more rigid tissue, both structurally and compositionally, while a membrane bilayer is more fluid in structure and composition.
  • Tissue includes, but is not limited to, an aggregate of cells usually of a particular kind together with their intercellular substance that form one of the structural materials commonly used to denote the general cellular fabric of a given organ, e.g., kidney tissue, brain tissue, lung tissue.
  • the four general classes of tissues are epithelial tissue, connective tissue, nerve tissue, and muscle tissue.
  • tissues which fall within this definition include, but are not limited to, heterogeneous aggregates of macromolecules such as fibrin clots which are acellular; homogeneous or heterogeneous aggregates of cells; higher ordered structures containing cells which have a specific function, such as organs, tumors, lymph nodes, arteries, etc.; and individual cells.
  • Tissues or cells can be in their natural environment, isolated, or in tissue culture. The tissue can be intact or modified. The modification can include numerous changes such as transformation, transfection, activation, and substructure isolation, e.g., cell membranes, cell nuclei, cell organelles, etc.
  • Sources of the tissue, cell or subcellular structures can be obtained from prokaryotes as well as eukaryotes. This includes human, animal, plant, bacterial, fungal and viral structures.
  • a biological sample can be derived by taking biological samples from a number of individuals and pooling them or pooling an aliquot of each individual's biological sample.
  • the pooled sample can be treated as a sample from a single individual and if the presence of cancer is established in the pooled sample, then each individual biological sample can be re-tested to determine which individual/s have lung cancer.
  • the phrase “data attributed to a biological sample from an individual” is intended to mean that the data in some form derived from, or were generated using, the biological sample of the individual.
  • the data may have been reformatted, revised, or mathematically altered to some degree after having been generated, such as by conversion from units in one measurement system to units in another measurement system; but, the data are understood to have been derived from, or were generated using, the biological sample.
  • Target “target molecule”, and “analyte” are used interchangeably herein to refer to any molecule of interest that may be present in a biological sample.
  • a “molecule of interest” includes any minor variation of a particular molecule, such as, in the case of a protein, for example, minor variations in amino acid sequence, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component, which does not substantially alter the identity of the molecule.
  • a “target molecule”, “target”, or “analyte” is a set of copies of one type or species of molecule or multi-molecular structure.
  • Target molecules refer to more than one such set of molecules.
  • exemplary target molecules include proteins, polypeptides, nucleic acids, carbohydrates, lipids, polysaccharides, glycoproteins, hormones, receptors, antigens, antibodies, affybodies, antibody mimics, viruses, pathogens, toxic substances, substrates, metabolites, transition state analogs, cofactors, inhibitors, drugs, dyes, nutrients, growth factors, cells, tissues, and any fragment or portion of any of the foregoing.
  • polypeptide As used herein, “polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to polymers of amino acids of any length.
  • the polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • Polypeptides can be single chains or associated chains. Also included within the definition are preproteins and intact mature proteins; peptides or polypeptides derived from a mature protein; fragments of a protein; splice variants; recombinant forms of a protein; protein variants with amino acid modifications, deletions, or substitutions; digests; and post-translational modifications, such as glycosylation, acetylation, phosphorylation, and the like.
  • marker and “biomarker” are used interchangeably to refer to a target molecule that indicates or is a sign of a normal or abnormal process in an individual or of a disease or other condition in an individual. More specifically, a “marker” or “biomarker” is an anatomic, physiologic, biochemical, or molecular parameter associated with the presence of a specific physiological state or process, whether normal or abnormal, and, if abnormal, whether chronic or acute. Biomarkers are detectable and measurable by a variety of methods including laboratory assays and medical imaging.
  • a biomarker is a protein
  • biomarker value As used herein, “biomarker value”, “value”, “biomarker level”, and “level” are used interchangeably to refer to a measurement that is made using any analytical method for detecting the biomarker in a biological sample and that indicates the presence, absence, absolute amount or concentration, relative amount or concentration, titer, a level, an expression level, a ratio of measured levels, or the like, of, for, or corresponding to the biomarker in the biological sample.
  • the exact nature of the “value” or “level” depends on the specific design and components of the particular analytical method employed to detect the biomarker.
  • biomarker indicates or is a sign of an abnormal process or a disease or other condition in an individual
  • that biomarker is generally described as being either over-expressed or under-expressed as compared to an expression level or value of the biomarker that indicates or is a sign of a normal process or an absence of a disease or other condition in an individual.
  • Up-regulation “up-regulated”, “over-expression”, “over-expressed”, and any variations thereof are used interchangeably to refer to a value or level of a biomarker in a biological sample that is greater than a value or level (or range of values or levels) of the biomarker that is typically detected in similar biological samples from healthy or normal individuals.
  • the terms may also refer to a value or level of a biomarker in a biological sample that is greater than a value or level (or range of values or levels) of the biomarker that may be detected at a different stage of a particular disease.
  • Down-regulation “down-regulated”, “under-expression”, “under-expressed”, and any variations thereof are used interchangeably to refer to a value or level of a biomarker in a biological sample that is less than a value or level (or range of values or levels) of the biomarker that is typically detected in similar biological samples from healthy or normal individuals.
  • the terms may also refer to a value or level of a biomarker in a biological sample that is less than a value or level (or range of values or levels) of the biomarker that may be detected at a different stage of a particular disease.
  • a biomarker that is either over-expressed or under-expressed can also be referred to as being “differentially expressed” or as having a “differential level” or “differential value” as compared to a “normal” expression level or value of the biomarker that indicates or is a sign of a normal process or an absence of a disease or other condition in an individual.
  • “differential expression” of a biomarker can also be referred to as a variation from a “normal” expression level of the biomarker.
  • differential gene expression and “differential expression” are used interchangeably to refer to a gene (or its corresponding protein expression product) whose expression is activated to a higher or lower level in a subject suffering from a specific disease, relative to its expression in a normal or control subject.
  • the terms also include genes (or the corresponding protein expression products) whose expression is activated to a higher or lower level at different stages of the same disease. It is also understood that a differentially expressed gene may be either activated or inhibited at the nucleic acid level or protein level, or may be subject to alternative splicing to result in a different polypeptide product.
  • Differential gene expression may include a comparison of expression between two or more genes or their gene products; or a comparison of the ratios of the expression between two or more genes or their gene products; or even a comparison of two differently processed products of the same gene, which differ between normal subjects and subjects suffering from a disease; or between various stages of the same disease.
  • Differential expression includes both quantitative, as well as qualitative, differences in the temporal or cellular expression pattern in a gene or its expression products among, for example, normal and diseased cells, or among cells which have undergone different disease events or disease stages.
  • “individual” refers to a test subject or patient.
  • the individual can be a mammal or a non-mammal.
  • the individual is a mammal.
  • a mammalian individual can be a human or non-human.
  • the individual is a human.
  • a healthy or normal individual is an individual in which the disease or condition of interest (including, for example, lung diseases, lung-associated diseases, or other lung conditions) is not detectable by conventional diagnostic methods.
  • Diagnose”, “diagnosing”, “diagnosis”, and variations thereof refer to the detection, determination, or recognition of a health status or condition of an individual on the basis of one or more signs, symptoms, data, or other information pertaining to that individual.
  • the health status of an individual can be diagnosed as healthy/normal (i.e., a diagnosis of the absence of a disease or condition) or diagnosed as ill/abnormal (i.e., a diagnosis of the presence, or an assessment of the characteristics, of a disease or condition).
  • diagnosis encompass, with respect to a particular disease or condition, the initial detection of the disease; the characterization or classification of the disease; the detection of the progression, remission, or recurrence of the disease; and the detection of disease response after the administration of a treatment or therapy to the individual.
  • diagnosis of lung cancer includes distinguishing individuals, including smokers and nonsmokers, who have cancer from individuals who do not. It further includes distinguishing benign pulmonary nodules from cancerous pulmonary nodules.
  • Prognose refers to the prediction of a future course of a disease or condition in an individual who has the disease or condition (e.g., predicting patient survival), and such terms encompass the evaluation of disease response after the administration of a treatment or therapy to the individual.
  • “Evaluate”, “evaluating”, “evaluation”, and variations thereof encompass both “diagnose” and “prognose” and also encompass determinations or predictions about the future course of a disease or condition in an individual who does not have the disease as well as determinations or predictions regarding the likelihood that a disease or condition will recur in an individual who apparently has been cured of the disease.
  • the term “evaluate” also encompasses assessing an individual's response to a therapy, such as, for example, predicting whether an individual is likely to respond favorably to a therapeutic agent or is unlikely to respond to a therapeutic agent (or will experience toxic or other undesirable side effects, for example), selecting a therapeutic agent for administration to an individual, or monitoring or determining an individual's response to a therapy that has been administered to the individual.
  • “evaluating” lung cancer can include, for example, any of the following: prognosing the future course of lung cancer in an individual; predicting the recurrence of lung cancer in an individual who apparently has been cured of lung cancer; or determining or predicting an individual's response to a lung cancer treatment or selecting a lung cancer treatment to administer to an individual based upon a determination of the biomarker values derived from the individual's biological sample.
  • any of the following examples may be referred to as either “diagnosing” or “evaluating” lung cancer: initially detecting the presence or absence of lung cancer; determining a specific stage, type or sub-type, or other classification or characteristic of lung cancer; determining whether a pulmonary nodule is a benign lesion or a malignant lung tumor; or detecting/monitoring lung cancer progression (e.g., monitoring lung tumor growth or metastatic spread), remission, or recurrence.
  • additional biomedical information refers to one or more evaluations of an individual, other than using any of the biomarkers described herein, that are associated with lung cancer risk.
  • “Additional biomedical information” includes any of the following: physical descriptors of an individual, physical descriptors of a pulmonary nodule observed by CT imaging, the height and/or weight of an individual, the gender of an individual, the ethnicity of an individual, smoking history, occupational history, exposure to known carcinogens (e.g., exposure to any of asbestos, radon gas, chemicals, smoke from fires, and air pollution, which can include emissions from stationary or mobile sources such as industrial/factory or auto/marine/aircraft emissions), exposure to second-hand smoke, family history of lung cancer (or other cancer), the presence of pulmonary nodules, size of nodules, location of nodules, morphology of nodules (e.g., as observed through CT imaging, ground glass opacity (GGO), solid, non-solid), edge characteristics of
  • Smoking history is usually quantified in terms of “pack years”, which refers to the number of years a person has smoked multiplied by the average number of packs smoked per day. For example, a person who has smoked, on average, one pack of cigarettes per day for 35 years is referred to as having 35 pack years of smoking history.
  • Additional biomedical information can be obtained from an individual using routine techniques known in the art, such as from the individual themselves by use of a routine patient questionnaire or health history questionnaire, etc., or from a medical practitioner, etc. Alternately, additional biomedical information can be obtained from routine imaging techniques, including CT imaging (e.g., low-dose CT imaging) and X-ray.
  • CT imaging e.g., low-dose CT imaging
  • X-ray X-ray
  • Testing of biomarker levels in combination with an evaluation of any additional biomedical information may, for example, improve sensitivity, specificity, and/or AUC for detecting lung cancer (or other lung cancer-related uses) as compared to biomarker testing alone or evaluating any particular item of additional biomedical information alone (e.g., CT imaging alone).
  • AUC area under the curve
  • ROC receiver operating characteristic
  • the feature data across the entire population e.g., the cases and controls
  • the true positive and false positive rates for the data are calculated.
  • the true positive rate is determined by counting the number of cases above the value for that feature and then dividing by the total number of cases.
  • the false positive rate is determined by counting the number of controls above the value for that feature and then dividing by the total number of controls.
  • ROC curves can be generated for a single feature as well as for other single outputs, for example, a combination of two or more features can be mathematically combined (e.g., added, subtracted, multiplied, etc.) to provide a single sum value, and this single sum value can be plotted in a ROC curve. Additionally, any combination of multiple features, in which the combination derives a single output value, can be plotted in a ROC curve. These combinations of features may comprise a test.
  • the ROC curve is the plot of the true positive rate (sensitivity) of a test against the false positive rate (1-specificity) of the test.
  • detecting or “determining” with respect to a biomarker value includes the use of both the instrument required to observe and record a signal corresponding to a biomarker value and the material/s required to generate that signal.
  • the biomarker value is detected using any suitable method, including fluorescence, chemiluminescence, surface plasmon resonance, surface acoustic waves, mass spectrometry, infrared spectroscopy, Raman spectroscopy, atomic force microscopy, scanning tunneling microscopy, electrochemical detection methods, nuclear magnetic resonance, quantum dots, and the like.
  • Solid support refers herein to any substrate having a surface to which molecules may be attached, directly or indirectly, through either covalent or non-covalent bonds.
  • a “solid support” can have a variety of physical formats, which can include, for example, a membrane; a chip (e.g., a protein chip); a slide (e.g., a glass slide or coverslip); a column; a hollow, solid, semi-solid, pore- or cavity-containing particle, such as, for example, a bead; a gel; a fiber, including a fiber optic material; a matrix; and a sample receptacle.
  • Exemplary sample receptacles include sample wells, tubes, capillaries, vials, and any other vessel, groove or indentation capable of holding a sample.
  • a sample receptacle can be contained on a multi-sample platform, such as a microtiter plate, slide, microfluidics device, and the like.
  • a support can be composed of a natural or synthetic material, an organic or inorganic material. The composition of the solid support on which capture reagents are attached generally depends on the method of attachment (e.g., covalent attachment).
  • Other exemplary receptacles include microdroplets and microfluidic controlled or bulk oil/aqueous emulsions within which assays and related manipulations can occur.
  • Suitable solid supports include, for example, plastics, resins, polysaccharides, silica or silica-based materials, functionalized glass, modified silicon, carbon, metals, inorganic glasses, membranes, nylon, natural fibers (such as, for example, silk, wool and cotton), polymers, and the like.
  • the material composing the solid support can include reactive groups such as, for example, carboxy, amino, or hydroxyl groups, which are used for attachment of the capture reagents.
  • Polymeric solid supports can include, e.g., polystyrene, polyethylene glycol tetraphthalate, polyvinyl acetate, polyvinyl chloride, polyvinyl pyrrolidone, polyacrylonitrile, polymethyl methacrylate, polytetrafluoroethylene, butyl rubber, styrenebutadiene rubber, natural rubber, polyethylene, polypropylene, (poly)tetrafluoroethylene, (poly)vinylidenefluoride, polycarbonate, and polymethylpentene.
  • Suitable solid support particles that can be used include, e.g., encoded particles, such as Luminex®-type encoded particles, magnetic particles, and glass particles.
  • methods are provided for diagnosing lung cancer in an individual by detecting one or more biomarker values corresponding to one or more biomarkers that are present in the lung tissue of an individual by any number of analytical methods, including any of the analytical methods described herein.
  • biomarkers are, for example, differentially expressed in individuals with lung cancer as compared to individuals without lung cancer, particularly NSCLC.
  • Detection of the differential expression of a biomarker in an individual can be used, for example, to permit the early diagnosis of lung cancer, to distinguish between a benign and malignant pulmonary nodule (such as, for example, a nodule observed on a computed tomography (CT) scan), to monitor lung cancer recurrence, or for other clinical indications, including determination of prognosis and methods of treatment.
  • a benign and malignant pulmonary nodule such as, for example, a nodule observed on a computed tomography (CT) scan
  • CT computed tomography
  • any of the biomarkers described herein may be used in a variety of clinical indications for lung cancer, including any of the following: detection of lung cancer (such as in a high-risk individual or population); characterizing lung cancer (e.g., determining lung cancer type, sub-type, or stage), such as by distinguishing between non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) and/or between adenocarcinoma and squamous cell carcinoma (or otherwise facilitating histopathology); determining whether a lung nodule is a benign nodule or a malignant lung tumor; determining lung cancer prognosis; monitoring lung cancer progression or remission; monitoring for lung cancer recurrence; monitoring metastasis; treatment selection; monitoring response to a therapeutic agent or other treatment; stratification of individuals for computed tomography (CT) screening (e.g., identifying those individuals at greater risk of lung cancer and thereby most likely to benefit from spiral-CT screening, thus increasing the positive predictive value of CT); combining biomark
  • facilitating the diagnosis of a pulmonary nodule as malignant or benign facilitating clinical decision making once a pulmonary nodule is observed on CT (e.g., ordering repeat CT scans if the nodule is deemed to be low risk, such as if a biomarker-based test is negative, with or without categorization of nodule size, or considering biopsy if the nodule is deemed medium to high risk, such as if a biomarker-based test is positive, with or without categorization of nodule size); and facilitating decisions regarding clinical follow-up (e.g., whether to implement repeat CT scans, fine needle biopsy, or thoracotomy after observing a non-calcified nodule on CT).
  • clinical follow-up e.g., whether to implement repeat CT scans, fine needle biopsy, or thoracotomy after observing a non-calcified nodule on CT.
  • Biomarker testing may improve positive predictive value (PPV) over CT screening alone.
  • the biomarkers described herein can also be used in conjunction with any other imaging modalities used for lung cancer, such as chest X-ray.
  • the described biomarkers may also be useful in permitting certain of these uses before indications of lung cancer are detected by imaging modalities or other clinical correlates, or before symptoms appear.
  • differential expression of one or more of the described biomarkers in an individual who is not known to have lung cancer may indicate that the individual has lung cancer, thereby enabling detection of lung cancer at an early stage of the disease when treatment is most effective, perhaps before the lung cancer is detected by other means or before symptoms appear.
  • Over-expression of one or more of the biomarkers during the course of lung cancer may be indicative of lung cancer progression, e.g., a lung tumor is growing and/or metastasizing (and thus indicate a poor prognosis), whereas a decrease in the degree to which one or more of the biomarkers is differentially expressed (i.e., in subsequent biomarker tests, the expression level in the individual is moving toward or approaching a “normal” expression level) may be indicative of lung cancer remission, e.g., a lung tumor is shrinking (and thus indicate a good or better prognosis).
  • an increase in the degree to which one or more of the biomarkers is differentially expressed may indicate that the lung cancer is progressing and therefore indicate that the treatment is ineffective
  • a decrease in differential expression of one or more of the biomarkers during the course of lung cancer treatment may be indicative of lung cancer remission and therefore indicate that the treatment is working successfully.
  • an increase or decrease in the differential expression of one or more of the biomarkers after an individual has apparently been cured of lung cancer may be indicative of lung cancer recurrence.
  • the individual can be re-started on therapy (or the therapeutic regimen modified such as to increase dosage amount and/or frequency, if the individual has maintained therapy) at an earlier stage than if the recurrence of lung cancer was not detected until later.
  • a differential expression level of one or more of the biomarkers in an individual may be predictive of the individual's response to a particular therapeutic agent.
  • changes in the biomarker expression levels may indicate the need for repeat imaging (e.g., repeat CT scanning), such as to determine lung cancer activity or to determine the need for changes in treatment.
  • Detection of any of the biomarkers described herein may be particularly useful following, or in conjunction with, lung cancer treatment, such as to evaluate the success of the treatment or to monitor lung cancer remission, recurrence, and/or progression (including metastasis) following treatment.
  • Lung cancer treatment may include, for example, administration of a therapeutic agent to the individual, performance of surgery (e.g., surgical resection of at least a portion of a lung tumor), administration of radiation therapy, or any other type of lung cancer treatment used in the art, and any combination of these treatments.
  • any of the biomarkers may be detected at least once after treatment or may be detected multiple times after treatment (such as at periodic intervals), or may be detected both before and after treatment.
  • Differential expression levels of any of the biomarkers in an individual over time may be indicative of lung cancer progression, remission, or recurrence, examples of which include any of the following: an increase or decrease in the expression level of the biomarkers after treatment compared with the expression level of the biomarker before treatment; an increase or decrease in the expression level of the biomarker at a later time point after treatment compared with the expression level of the biomarker at an earlier time point after treatment; and a differential expression level of the biomarker at a single time point after treatment compared with normal levels of the biomarker.
  • the biomarker levels for any of the biomarkers described herein can be determined in pre-surgery and post-surgery (e.g., 2-4 weeks after surgery) serum samples.
  • An increase in the biomarker expression level(s) in the post-surgery sample compared with the pre-surgery sample can indicate progression of lung cancer (e.g., unsuccessful surgery), whereas a decrease in the biomarker expression level(s) in the post-surgery sample compared with the pre-surgery sample can indicate regression of lung cancer (e.g., the surgery successfully removed the lung tumor).
  • Similar analyses of the biomarker levels can be carried out before and after other forms of treatment, such as before and after radiation therapy or administration of a therapeutic agent or cancer vaccine.
  • biomarker levels can also be done in conjunction with determination of SNPs or other genetic lesions or variability that are indicative of increased risk of susceptibility of disease. (See, e.g., Amos et al., Nature Genetics 40, 616-622 (2009)).
  • biomarker levels can also be done in conjunction with CT screening.
  • the biomarkers may facilitate the medical and economic justification for implementing CT screening, such as for screening large asymptomatic populations at risk for lung cancer (e.g., smokers).
  • a “pre-CT” test of biomarker levels could be used to stratify high-risk individuals for CT screening, such as for identifying those who are at highest risk for lung cancer based on their biomarker levels and who should be prioritized for CT screening.
  • biomarker levels e.g., as determined by an aptamer assay of serum or plasma samples
  • additional biomedical information e.g., tumor parameters determined by CT testing
  • PPV positive predictive value
  • a “post-CT” aptamer panel for determining biomarker levels can be used to determine the likelihood that a pulmonary nodule observed by CT (or other imaging modality) is malignant or benign.
  • biomarker testing may eliminate or reduce a significant number of false positive tests over CT alone. Further, biomarker testing may facilitate treatment of patients. By way of example, if a lung nodule is less than 5 mm in size, results of biomarker testing may advance patients from “watch and wait” to biopsy at an earlier time; if a lung nodule is 5-9 mm, biomarker testing may eliminate the use of a biopsy or thoracotomy on false positive scans; and if a lung nodule is larger than 10 mm, biomarker testing may eliminate surgery for a sub-population of these patients with benign nodules.
  • Eliminating the need for biopsy in some patients based on biomarker testing would be beneficial because there is significant morbidity associated with nodule biopsy and difficulty in obtaining nodule tissue depending on the location of nodule. Similarly, eliminating the need for surgery in some patients, such as those whose nodules are actually benign, would avoid unnecessary risks and costs associated with surgery.
  • biomarker levels in conjunction with CT screening e.g., assessing biomarker levels in conjunction with size or other characteristics of a lung nodule observed on a CT scan
  • information regarding the biomarkers can also be evaluated in conjunction with other types of data, particularly data that indicates an individual's risk for lung cancer (e.g., patient clinical history, symptoms, family history of cancer, risk factors such as whether or not the individual is a smoker, and/or status of other biomarkers, etc.).
  • data that indicates an individual's risk for lung cancer e.g., patient clinical history, symptoms, family history of cancer, risk factors such as whether or not the individual is a smoker, and/or status of other biomarkers, etc.
  • These various data can be assessed by automated methods, such as a computer program/software, which can be embodied in a computer or other apparatus/device.
  • an imaging agent can be coupled to any of the described biomarkers, which can be used to aid in lung cancer diagnosis, to monitor disease progression/remission or metastasis, to monitor for disease recurrence, or to monitor response to therapy, among other uses.
  • a biomarker value for the biomarkers described herein can be detected using any of a variety of known analytical methods.
  • a biomarker value is detected using a capture reagent.
  • a “capture agent” or “capture reagent” refers to a molecule that is capable of binding specifically to a biomarker.
  • the capture reagent can be exposed to the biomarker in solution or can be exposed to the biomarker while the capture reagent is immobilized on a solid support.
  • the capture reagent contains a feature that is reactive with a secondary feature on a solid support.
  • the capture reagent can be exposed to the biomarker in solution, and then the feature on the capture reagent can be used in conjunction with the secondary feature on the solid support to immobilize the biomarker on the solid support.
  • the capture reagent is selected based on the type of analysis to be conducted.
  • Capture reagents include but are not limited to aptamers, antibodies, adnectins, ankyrins, other antibody mimetics and other protein scaffolds, autoantibodies, chimeras, small molecules, an F(ab′) 2 fragment, a single chain antibody fragment, an Fv fragment, a single chain Fv fragment, a nucleic acid, a lectin, a ligand-binding receptor, affybodies, nanobodies, imprinted polymers, avimers, peptidomimetics, a hormone receptor, a cytokine receptor, and synthetic receptors, and modifications and fragments of these.
  • a biomarker value is detected using a biomarker/capture reagent complex.
  • the biomarker value is derived from the biomarker/capture reagent complex and is detected indirectly, such as, for example, as a result of a reaction that is subsequent to the biomarker/capture reagent interaction, but is dependent on the formation of the biomarker/capture reagent complex.
  • the biomarker value is detected directly from the biomarker in a biological sample.
  • the biomarkers are detected using a multiplexed format that allows for the simultaneous detection of two or more biomarkers in a biological sample.
  • capture reagents are immobilized, directly or indirectly, covalently or non-covalently, in discrete locations on a solid support.
  • a multiplexed format uses discrete solid supports where each solid support has a unique capture reagent associated with that solid support, such as, for example quantum dots.
  • an individual device is used for the detection of each one of multiple biomarkers to be detected in a biological sample. Individual devices can be configured to permit each biomarker in the biological sample to be processed simultaneously. For example, a microtiter plate can be used such that each well in the plate is used to uniquely analyze one of multiple biomarkers to be detected in a biological sample.
  • a fluorescent tag can be used to label a component of the biomarker/capture complex to enable the detection of the biomarker value.
  • the fluorescent label can be conjugated to a capture reagent specific to any of the biomarkers described herein using known techniques, and the fluorescent label can then be used to detect the corresponding biomarker value.
  • Suitable fluorescent labels include rare earth chelates, fluorescein and its derivatives, rhodamine and its derivatives, dansyl, allophycocyanin, PBXL-3, Qdot 605, Lissamine, phycoerythrin, Texas Red, and other such compounds.
  • the fluorescent label is a fluorescent dye molecule.
  • the fluorescent dye molecule includes at least one substituted indolium ring system in which the substituent on the 3-carbon of the indolium ring contains a chemically reactive group or a conjugated substance.
  • the dye molecule includes an AlexFluor molecule, such as, for example, AlexaFluor 488, AlexaFluor 532, AlexaFluor 647, AlexaFluor 680, or AlexaFluor 700.
  • the dye molecule includes a first type and a second type of dye molecule, such as, e.g., two different AlexaFluor molecules.
  • the dye molecule includes a first type and a second type of dye molecule, and the two dye molecules have different emission spectra.
  • Fluorescence can be measured with a variety of instrumentation compatible with a wide range of assay formats.
  • spectrofluorimeters have been designed to analyze microtiter plates, microscope slides, printed arrays, cuvettes, etc. See Principles of Fluorescence Spectroscopy, by J. R. Lakowicz, Springer Science+Business Media, Inc., 2004; Bioluminescence & Chemiluminescence: Progress & Current Applications; Philip E. Stanley and Larry J. Kricka editors, World Scientific Publishing Company, January 2002.
  • a chemiluminescence tag can optionally be used to label a component of the biomarker/capture complex to enable the detection of a biomarker value.
  • Suitable chemiluminescent materials include any of oxalyl chloride, Rodamin 6G, Ru(bipy) 3 2+ , TMAE (tetrakis(dimethylamino)ethylene), pyrogallol (1,2,3-trihydroxibenzene), Lucigenin, peroxyoxalates, aryl oxalates, acridinium esters, dioxetanes, and others.
  • the detection method includes an enzyme/substrate combination that generates a detectable signal that corresponds to the biomarker value.
  • the enzyme catalyzes a chemical alteration of the chromogenic substrate which can be measured using various techniques, including spectrophotometry, fluorescence, and chemiluminescence.
  • Suitable enzymes include, for example, luciferases, luciferin, malate dehydrogenase, urease, horseradish peroxidase (HRPO), alkaline phosphatase, beta-galactosidase, glucoamylase, lysozyme, glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase, uricase, xanthine oxidase, lactoperoxidase, microperoxidase, and the like.
  • HRPO horseradish peroxidase
  • alkaline phosphatase beta-galactosidase
  • glucoamylase lysozyme
  • glucose oxidase galactose oxidase
  • glucose-6-phosphate dehydrogenase uricase
  • xanthine oxidase lactoperoxidase
  • microperoxidase and the like.
  • the detection method can be a combination of fluorescence, chemiluminescence, radionuclide or enzyme/substrate combinations that generate a measurable signal.
  • Multimodal signaling could have unique and advantageous characteristics in biomarker assay formats.
  • biomarker values for the biomarkers described herein can be detected using known analytical methods including, singleplex aptamer assays, multiplexed aptamer assays, singleplex or multiplexed immunoassays, mRNA expression profiling, miRNA expression profiling, mass spectrometric analysis, histological/cytological methods, etc. as detailed below.
  • Assays directed to the detection and quantification of physiologically significant molecules in biological samples and other samples are important tools in scientific research and in the health care field.
  • One class of such assays involves the use of a microarray that includes one or more aptamers immobilized on a solid support.
  • the aptamers are each capable of binding to a target molecule in a highly specific manner and with very high affinity. See, e.g., U.S. Pat. No. 5,475,096 entitled “Nucleic Acid Ligands,” see also, e.g., U.S. Pat. No. 6,242,246, U.S. Pat. No. 6,458,543, and U.S. Pat. No. 6,503,715, each of which is entitled “Nucleic Acid Ligand Diagnostic Biochip”.
  • the aptamers bind to their respective target molecules present in the sample and thereby enable a determination of a biomarker value corresponding to a biomarker.
  • an “aptamer” refers to a nucleic acid that has a specific binding affinity for a target molecule. It is recognized that affinity interactions are a matter of degree; however, in this context, the “specific binding affinity” of an aptamer for its target means that the aptamer binds to its target generally with a much higher degree of affinity than it binds to other components in a test sample.
  • An “aptamer” is a set of copies of one type or species of nucleic acid molecule that has a particular nucleotide sequence.
  • An aptamer can include any suitable number of nucleotides, including any number of chemically modified nucleotides. “Aptamers” refers to more than one such set of molecules.
  • aptamers can have either the same or different numbers of nucleotides.
  • Aptamers can be DNA or RNA or chemically modified nucleic acids and can be single stranded, double stranded, or contain double stranded regions, and can include higher ordered structures.
  • An aptamer can also be a photoaptamer, where a photoreactive or chemically reactive functional group is included in the aptamer to allow it to be covalently linked to its corresponding target. Any of the aptamer methods disclosed herein can include the use of two or more aptamers that specifically bind the same target molecule.
  • an aptamer may include a tag. If an aptamer includes a tag, all copies of the aptamer need not have the same tag. Moreover, if different aptamers each include a tag, these different aptamers can have either the same tag or a different tag.
  • An aptamer can be identified using any known method, including the SELEX process. Once identified, an aptamer can be prepared or synthesized in accordance with any known method, including chemical synthetic methods and enzymatic synthetic methods.
  • SELEX and “SELEX process” are used interchangeably herein to refer generally to a combination of (1) the selection of aptamers that interact with a target molecule in a desirable manner, for example binding with high affinity to a protein, with (2) the amplification of those selected nucleic acids.
  • the SELEX process can be used to identify aptamers with high affinity to a specific target or biomarker.
  • SELEX generally includes preparing a candidate mixture of nucleic acids, binding of the candidate mixture to the desired target molecule to form an affinity complex, separating the affinity complexes from the unbound candidate nucleic acids, separating and isolating the nucleic acid from the affinity complex, purifying the nucleic acid, and identifying a specific aptamer sequence.
  • the process may include multiple rounds to further refine the affinity of the selected aptamer.
  • the process can include amplification steps at one or more points in the process. See, e.g., U.S. Pat. No.
  • the SELEX process can be used to generate an aptamer that covalently binds its target as well as an aptamer that non-covalently binds its target. See, e.g., U.S. Pat. No. 5,705,337 entitled “Systematic Evolution of Nucleic Acid Ligands by Exponential Enrichment: Chemi-SELEX.”
  • the SELEX process can be used to identify high-affinity aptamers containing modified nucleotides that confer improved characteristics on the aptamer, such as, for example, improved in vivo stability or improved delivery characteristics. Examples of such modifications include chemical substitutions at the ribose and/or phosphate and/or base positions. SELEX process-identified aptamers containing modified nucleotides are described in U.S. Pat. No. 5,660,985, entitled “High Affinity Nucleic Acid Ligands Containing Modified Nucleotides,” which describes oligonucleotides containing nucleotide derivatives chemically modified at the 5′- and 2′-positions of pyrimidines. U.S. Pat. No.
  • SELEX can also be used to identify aptamers that have desirable off-rate characteristics. See U.S. Patent Application Publication 20090004667, entitled “Method for Generating Aptamers with Improved Off-Rates,” which describes improved SELEX methods for generating aptamers that can bind to target molecules. Methods for producing aptamers and photoaptamers having slower rates of dissociation from their respective target molecules are described. The methods involve contacting the candidate mixture with the target molecule, allowing the formation of nucleic acid-target complexes to occur, and performing a slow off-rate enrichment process wherein nucleic acid-target complexes with fast dissociation rates will dissociate and not reform, while complexes with slow dissociation rates will remain intact. Additionally, the methods include the use of modified nucleotides in the production of candidate nucleic acid mixtures to generate aptamers with improved off-rate performance.
  • a variation of this assay employs aptamers that include photoreactive functional groups that enable the aptamers to covalently bind or “photocrosslink” their target molecules. See, e.g., U.S. Pat. No. 6,544,776 entitled “Nucleic Acid Ligand Diagnostic Biochip.” These photoreactive aptamers are also referred to as photoaptamers. See, e.g., U.S. Pat. No. 5,763,177, U.S. Pat. No. 6,001,577, and U.S. Pat. No.
  • the aptamers are immobilized on the solid support prior to being contacted with the sample. Under certain circumstances, however, immobilization of the aptamers prior to contact with the sample may not provide an optimal assay. For example, pre-immobilization of the aptamers may result in inefficient mixing of the aptamers with the target molecules on the surface of the solid support, perhaps leading to lengthy reaction times and, therefore, extended incubation periods to permit efficient binding of the aptamers to their target molecules. Further, when photoaptamers are employed in the assay and depending upon the material utilized as a solid support, the solid support may tend to scatter or absorb the light used to effect the formation of covalent bonds between the photoaptamers and their target molecules.
  • immobilization of the aptamers on the solid support generally involves an aptamer-preparation step (i.e., the immobilization) prior to exposure of the aptamers to the sample, and this preparation step may affect the activity or functionality of the aptamers.
  • aptamer assays that permit an aptamer to capture its target in solution and then employ separation steps that are designed to remove specific components of the aptamer-target mixture prior to detection have also been described (see U.S. Patent Application Publication 20090042206, entitled “Multiplexed Analyses of Test Samples”).
  • the described aptamer assay methods enable the detection and quantification of a non-nucleic acid target (e.g., a protein target) in a test sample by detecting and quantifying a nucleic acid (i.e., an aptamer).
  • the described methods create a nucleic acid surrogate (i.e, the aptamer) for detecting and quantifying a non-nucleic acid target, thus allowing the wide variety of nucleic acid technologies, including amplification, to be applied to a broader range of desired targets, including protein targets.
  • a nucleic acid surrogate i.e, the aptamer
  • Aptamers can be constructed to facilitate the separation of the assay components from an aptamer biomarker complex (or photoaptamer biomarker covalent complex) and permit isolation of the aptamer for detection and/or quantification.
  • these constructs can include a cleavable or releasable element within the aptamer sequence.
  • additional functionality can be introduced into the aptamer, for example, a labeled or detectable component, a spacer component, or a specific binding tag or immobilization element.
  • the aptamer can include a tag connected to the aptamer via a cleavable moiety, a label, a spacer component separating the label, and the cleavable moiety.
  • a cleavable element is a photocleavable linker.
  • the photocleavable linker can be attached to a biotin moiety and a spacer section, can include an NHS group for derivatization of amines, and can be used to introduce a biotin group to an aptamer, thereby allowing for the release of the aptamer later in an assay method.
  • Homogenous assays done with all assay components in solution, do not require separation of sample and reagents prior to the detection of signal. These methods are rapid and easy to use. These methods generate signal based on a molecular capture or binding reagent that reacts with its specific target.
  • the molecular capture reagents would be an aptamer or an antibody or the like and the specific target would be a lung cancer biomarker of Table 20.
  • a method for signal generation takes advantage of anisotropy signal change due to the interaction of a fluorophore-labeled capture reagent with its specific biomarker target.
  • the labeled capture reacts with its target, the increased molecular weight causes the rotational motion of the fluorophore attached to the complex to become much slower changing the anisotropy value.
  • binding events may be used to quantitatively measure the biomarkers in solutions.
  • Other methods include fluorescence polarization assays, molecular beacon methods, time resolved fluorescence quenching, chemiluminescence, fluorescence resonance energy transfer, and the like.
  • An exemplary solution-based aptamer assay that can be used to detect a biomarker value corresponding to a biomarker in a biological sample includes the following: (a) preparing a mixture by contacting the biological sample with an aptamer that includes a first tag and has a specific affinity for the biomarker, wherein an aptamer affinity complex is formed when the biomarker is present in the sample; (b) exposing the mixture to a first solid support including a first capture element, and allowing the first tag to associate with the first capture element; (c) removing any components of the mixture not associated with the first solid support; (d) attaching a second tag to the biomarker component of the aptamer affinity complex; (e) releasing the aptamer affinity complex from the first solid support; (f) exposing the released aptamer affinity complex to a second solid support that includes a second capture element and allowing the second tag to associate with the second capture element; (g) removing any non-complexed aptamer from the mixture
  • Immunoassay methods are based on the reaction of an antibody to its corresponding target or analyte and can detect the analyte in a sample depending on the specific assay format.
  • monoclonal antibodies are often used because of their specific epitope recognition.
  • Polyclonal antibodies have also been successfully used in various immunoassays because of their increased affinity for the target as compared to monoclonal antibodies.
  • Immunoassays have been designed for use with a wide range of biological sample matrices. Immunoassay formats have been designed to provide qualitative, semi-quantitative, and quantitative results.
  • Quantitative results are generated through the use of a standard curve created with known concentrations of the specific analyte to be detected.
  • the response or signal from an unknown sample is plotted onto the standard curve, and a quantity or value corresponding to the target in the unknown sample is established.
  • ELISA or EIA can be quantitative for the detection of an analyte. This method relies on attachment of a label to either the analyte or the antibody and the label component includes, either directly or indirectly, an enzyme. ELISA tests may be formatted for direct, indirect, competitive, or sandwich detection of the analyte. Other methods rely on labels such as, for example, radioisotopes (I 125 ) or fluorescence.
  • Additional techniques include, for example, agglutination, nephelometry, turbidimetry, Western blot, immunoprecipitation, immunocytochemistry, immunohistochemistry, flow cytometry, Luminex assay, and others (see ImmunoAssay: A Practical Guide, edited by Brian Law, published by Taylor & Francis, Ltd., 2005 edition).
  • Exemplary assay formats include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay, fluorescent, chemiluminescence, and fluorescence resonance energy transfer (FRET) or time resolved-FRET (TR-FRET) immunoassays.
  • ELISA enzyme-linked immunosorbent assay
  • FRET fluorescence resonance energy transfer
  • TR-FRET time resolved-FRET
  • biomarkers include biomarker immunoprecipitation followed by quantitative methods that allow size and peptide level discrimination, such as gel electrophoresis, capillary electrophoresis, planar electrochromatography, and the like.
  • Methods of detecting and/or quantifying a detectable label or signal generating material depend on the nature of the label.
  • the products of reactions catalyzed by appropriate enzymes can be, without limitation, fluorescent, luminescent, or radioactive or they may absorb visible or ultraviolet light.
  • detectors suitable for detecting such detectable labels include, without limitation, x-ray film, radioactivity counters, scintillation counters, spectrophotometers, colorimeters, fluorometers, luminometers, and densitometers.
  • Any of the methods for detection can be performed in any format that allows for any suitable preparation, processing, and analysis of the reactions. This can be, for example, in multi-well assay plates (e.g., 96 wells or 384 wells) or using any suitable array or microarray. Stock solutions for various agents can be made manually or robotically, and all subsequent pipetting, diluting, mixing, distribution, washing, incubating, sample readout, data collection and analysis can be done robotically using commercially available analysis software, robotics, and detection instrumentation capable of detecting a detectable label.
  • Measuring mRNA in a biological sample may be used as a surrogate for detection of the level of the corresponding protein in the biological sample.
  • any of the biomarkers or biomarker panels described herein can also be detected by detecting the appropriate RNA.
  • mRNA expression levels are measured by reverse transcription quantitative polymerase chain reaction (RT-PCR followed with qPCR).
  • RT-PCR is used to create a cDNA from the mRNA.
  • the cDNA may be used in a qPCR assay to produce fluorescence as the DNA amplification process progresses. By comparison to a standard curve, qPCR can produce an absolute measurement such as number of copies of mRNA per cell.
  • Northern blots, microarrays, Invader assays, and RT-PCR combined with capillary electrophoresis have all been used to measure expression levels of mRNA in a sample (see Gene Expression Profiling: Methods and Protocols, Richard A. Shimkets, editor, Humana Press, 2004).
  • miRNA molecules are small RNAs that are non-coding but may regulate gene expression. Any of the methods suited to the measurement of mRNA expression levels can also be used for the corresponding miRNA. Recently many laboratories have investigated the use of miRNAs as biomarkers for disease. Many diseases involve wide-spread transcriptional regulation, and it is not surprising that miRNAs might find a role as biomarkers. The connection between miRNA concentrations and disease is often even less clear than the connections between protein levels and disease, yet the value of miRNA biomarkers might be substantial.
  • RNA biomarkers have similar requirements, although many potential protein biomarkers are secreted intentionally at the site of pathology and function, during disease, in a paracrine fashion. Many potential protein biomarkers are designed to function outside the cells within which those proteins are synthesized.
  • any of the described biomarkers may also be used in molecular imaging tests.
  • an imaging agent can be coupled to any of the described biomarkers, which can be used to aid in lung cancer diagnosis, to monitor disease progression/remission or metastasis, to monitor for disease recurrence, or to monitor response to therapy, among other uses.
  • In vivo imaging technologies provide non-invasive methods for determining the state of a particular disease in the body of an individual. For example, entire portions of the body, or even the entire body, may be viewed as a three dimensional image, thereby providing valuable information concerning morphology and structures in the body. Such technologies may be combined with the detection of the biomarkers described herein to provide information concerning the cancer status, in particular the lung cancer status, of an individual.
  • in vivo molecular imaging technologies are expanding due to various advances in technology. These advances include the development of new contrast agents or labels, such as radiolabels and/or fluorescent labels, which can provide strong signals within the body; and the development of powerful new imaging technology, which can detect and analyze these signals from outside the body, with sufficient sensitivity and accuracy to provide useful information.
  • the contrast agent can be visualized in an appropriate imaging system, thereby providing an image of the portion or portions of the body in which the contrast agent is located.
  • the contrast agent may be bound to or associated with a capture reagent, such as an aptamer or an antibody, for example, and/or with a peptide or protein, or an oligonucleotide (for example, for the detection of gene expression), or a complex containing any of these with one or more macromolecules and/or other particulate forms.
  • a capture reagent such as an aptamer or an antibody, for example, and/or with a peptide or protein, or an oligonucleotide (for example, for the detection of gene expression), or a complex containing any of these with one or more macromolecules and/or other particulate forms.
  • the contrast agent may also feature a radioactive atom that is useful in imaging.
  • Suitable radioactive atoms include technetium-99m or iodine-123 for scintigraphic studies.
  • Other readily detectable moieties include, for example, spin labels for magnetic resonance imaging (MRI) such as, for example, iodine-123 again, iodine-131, indium-111, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
  • MRI magnetic resonance imaging
  • Standard imaging techniques include but are not limited to magnetic resonance imaging, computed tomography scanning, positron emission tomography (PET), single photon emission computed tomography (SPECT), and the like.
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • the type of detection instrument available is a major factor in selecting a given contrast agent, such as a given radionuclide and the particular biomarker that it is used to target (protein, mRNA, and the like).
  • the radionuclide chosen typically has a type of decay that is detectable by a given type of instrument.
  • its half-life should be long enough to enable detection at the time of maximum uptake by the target tissue but short enough that deleterious radiation of the host is minimized.
  • Exemplary imaging techniques include but are not limited to PET and SPECT, which are imaging techniques in which a radionuclide is synthetically or locally administered to an individual. The subsequent uptake of the radiotracer is measured over time and used to obtain information about the targeted tissue and the biomarker. Because of the high-energy (gamma-ray) emissions of the specific isotopes employed and the sensitivity and sophistication of the instruments used to detect them, the two-dimensional distribution of radioactivity may be inferred from outside of the body.
  • PET and SPECT are imaging techniques in which a radionuclide is synthetically or locally administered to an individual. The subsequent uptake of the radiotracer is measured over time and used to obtain information about the targeted tissue and the biomarker. Because of the high-energy (gamma-ray) emissions of the specific isotopes employed and the sensitivity and sophistication of the instruments used to detect them, the two-dimensional distribution of radioactivity may be inferred from outside of the body.
  • positron-emitting nuclides in PET include, for example, carbon-11, nitrogen-13, oxygen-15, and fluorine-18.
  • Isotopes that decay by electron capture and/or gamma-emission are used in SPECT and include, for example iodine-123 and technetium-99m.
  • An exemplary method for labeling amino acids with technetium-99m is the reduction of pertechnetate ion in the presence of a chelating precursor to form the labile technetium-99m-precursor complex, which, in turn, reacts with the metal binding group of a bifunctionally modified chemotactic peptide to form a technetium-99m-chemotactic peptide conjugate.
  • Antibodies are frequently used for such in vivo imaging diagnostic methods.
  • the preparation and use of antibodies for in vivo diagnosis is well known in the art.
  • Labeled antibodies which specifically bind any of the biomarkers in Table 20 can be injected into an individual suspected of having a certain type of cancer (e.g., lung cancer), detectable according to the particular biomarker used, for the purpose of diagnosing or evaluating the disease status of the individual.
  • the label used will be selected in accordance with the imaging modality to be used, as previously described. Localization of the label permits determination of the spread of the cancer.
  • the amount of label within an organ or tissue also allows determination of the presence or absence of cancer in that organ or tissue.
  • aptamers may be used for such in vivo imaging diagnostic methods.
  • an aptamer that was used to identify a particular biomarker described in Table 20 (and therefore binds specifically to that particular biomarker) may be appropriately labeled and injected into an individual suspected of having lung cancer, detectable according to the particular biomarker, for the purpose of diagnosing or evaluating the lung cancer status of the individual.
  • the label used will be selected in accordance with the imaging modality to be used, as previously described. Localization of the label permits determination of the spread of the cancer.
  • the amount of label within an organ or tissue also allows determination of the presence or absence of cancer in that organ or tissue.
  • Aptamer-directed imaging agents could have unique and advantageous characteristics relating to tissue penetration, tissue distribution, kinetics, elimination, potency, and selectivity as compared to other imaging agents.
  • Such techniques may also optionally be performed with labeled oligonucleotides, for example, for detection of gene expression through imaging with antisense oligonucleotides. These methods are used for in situ hybridization, for example, with fluorescent molecules or radionuclides as the label. Other methods for detection of gene expression include, for example, detection of the activity of a reporter gene.
  • optical imaging Another general type of imaging technology is optical imaging, in which fluorescent signals within the subject are detected by an optical device that is external to the subject. These signals may be due to actual fluorescence and/or to bioluminescence. Improvements in the sensitivity of optical detection devices have increased the usefulness of optical imaging for in vivo diagnostic assays.
  • in vivo molecular biomarker imaging is increasing, including for clinical trials, for example, to more rapidly measure clinical efficacy in trials for new cancer therapies and/or to avoid prolonged treatment with a placebo for those diseases, such as multiple sclerosis, in which such prolonged treatment may be considered to be ethically questionable.
  • tissue samples may be used in histological or cytological methods. Sample selection depends on the primary tumor location and sites of metastases. For example, endo- and trans-bronchial biopsies, fine needle aspirates, cutting needles, and core biopsies can be used for histology. Bronchial washing and brushing, pleural aspiration, and sputum, can be used for cytology. While cytological analysis is still used in the diagnosis of lung cancer, histological methods are known to provide better sensitivity for the detection of cancer. Any of the biomarkers identified herein that were shown to be up-regulated (see Table 19) in the individuals with lung cancer can be used to stain a histological specimen as an indication of disease.
  • one or more capture reagent(s) specific to the corresponding biomarker(s) are used in a cytological evaluation of a lung cell sample and may include one or more of the following: collecting a cell sample, fixing the cell sample, dehydrating, clearing, immobilizing the cell sample on a microscope slide, permeabilizing the cell sample, treating for analyte retrieval, staining, destaining, washing, blocking, and reacting with one or more capture reagent/s in a buffered solution.
  • the cell sample is produced from a cell block.
  • one or more capture reagent/s specific to the corresponding biomarkers are used in a histological evaluation of a lung tissue sample and may include one or more of the following: collecting a tissue specimen, fixing the tissue sample, dehydrating, clearing, immobilizing the tissue sample on a microscope slide, permeabilizing the tissue sample, treating for analyte retrieval, staining, destaining, washing, blocking, rehydrating, and reacting with capture reagent/s in a buffered solution.
  • fixing and dehydrating are replaced with freezing.
  • the one or more aptamer/s specific to the corresponding biomarker/s are reacted with the histological or cytological sample and can serve as the nucleic acid target in a nucleic acid amplification method.
  • Suitable nucleic acid amplification methods include, for example, PCR, q-beta replicase, rolling circle amplification, strand displacement, helicase dependent amplification, loop mediated isothermal amplification, ligase chain reaction, and restriction and circularization aided rolling circle amplification.
  • the one or more capture reagent(s) specific to the corresponding biomarkers for use in the histological or cytological evaluation are mixed in a buffered solution that can include any of the following: blocking materials, competitors, detergents, stabilizers, carrier nucleic acid, polyanionic materials, etc.
  • a “cytology protocol” generally includes sample collection, sample fixation, sample immobilization, and staining.
  • Cell preparation can include several processing steps after sample collection, including the use of one or more slow off-rate aptamers for the staining of the prepared cells.
  • Sample collection can include directly placing the sample in an untreated transport container, placing the sample in a transport container containing some type of media, or placing the sample directly onto a slide (immobilization) without any treatment or fixation.
  • Sample immobilization can be improved by applying a portion of the collected specimen to a glass slide that is treated with polylysine, gelatin, or a silane. Slides can be prepared by smearing a thin and even layer of cells across the slide. Care is generally taken to minimize mechanical distortion and drying artifacts. Liquid specimens can be processed in a cell block method. Or, alternatively, liquid specimens can be mixed 1:1 with the fixative solution for about 10 minutes at room temperature.
  • Cell blocks can be prepared from residual effusions, sputum, urine sediments, gastrointestinal fluids, cell scraping, or fine needle aspirates. Cells are concentrated or packed by centrifugation or membrane filtration. A number of methods for cell block preparation have been developed. Representative procedures include the fixed sediment, bacterial agar, or membrane filtration methods. In the fixed sediment method, the cell sediment is mixed with a fixative like Bouins, picric acid, or buffered formalin and then the mixture is centrifuged to pellet the fixed cells. The supernatant is removed, drying the cell pellet as completely as possible. The pellet is collected and wrapped in lens paper and then placed in a tissue cassette. The tissue cassette is placed in a jar with additional fixative and processed as a tissue sample.
  • a fixative like Bouins, picric acid, or buffered formalin
  • Agar method is very similar but the pellet is removed and dried on paper towel and then cut in half. The cut side is placed in a drop of melted agar on a glass slide and then the pellet is covered with agar making sure that no bubbles form in the agar. The agar is allowed to harden and then any excess agar is trimmed away. This is placed in a tissue cassette and the tissue process completed.
  • the pellet may be directly suspended in 2% liquid agar at 65° C. and the sample centrifuged. The agar cell pellet is allowed to solidify for an hour at 4° C. The solid agar may be removed from the centrifuge tube and sliced in half. The agar is wrapped in filter paper and then the tissue cassette. Processing from this point forward is as described above. Centrifugation can be replaced in any these procedures with membrane filtration. Any of these processes may be used to generate a “cell block sample”.
  • Cell blocks can be prepared using specialized resin including Lowicryl resins, LR White, LR Gold, Unicryl, and MonoStep. These resins have low viscosity and can be polymerized at low temperatures and with ultra violet (UV) light.
  • the embedding process relies on progressively cooling the sample during dehydration, transferring the sample to the resin, and polymerizing a block at the final low temperature at the appropriate UV wavelength.
  • Cell block sections can be stained with hematoxylin-eosin for cytomorphological examination while additional sections are used for examination for specific markers.
  • the sample may be fixed prior to additional processing to prevent sample degradation.
  • This process is called “fixation” and describes a wide range of materials and procedures that may be used interchangeably.
  • the sample fixation protocol and reagents are best selected empirically based on the targets to be detected and the specific cell/tissue type to be analyzed.
  • Sample fixation relies on reagents such as ethanol, polyethylene glycol, methanol, formalin, or isopropanol.
  • the samples should be fixed as soon after collection and affixation to the slide as possible.
  • the fixative selected can introduce structural changes into various molecular targets making their subsequent detection more difficult.
  • fixation and immobilization processes and their sequence can modify the appearance of the cell and these changes must be anticipated and recognized by the cytotechnologist.
  • Fixatives can cause shrinkage of certain cell types and cause the cytoplasm to appear granular or reticular.
  • Many fixatives function by crosslinking cellular components. This can damage or modify specific epitopes, generate new epitopes, cause molecular associations, and reduce membrane permeability.
  • Formalin fixation is one of the most common cytological/histological approaches. Formalin forms methyl bridges between neighboring proteins or within proteins. Precipitation or coagulation is also used for fixation and ethanol is frequently used in this type of fixation.
  • a combination of cros slinking and precipitation can also be used for fixation.
  • a strong fixation process is best at preserving morphological information while a weaker fixation process is best for the preservation of molecular targets.
  • a representative fixative is 50% absolute ethanol, 2 mM polyethylene glycol (PEG), 1.85% formaldehyde. Variations on this formulation include ethanol (50% to 95%), methanol (20%-50%), and formalin (formaldehyde) only.
  • Another common fixative is 2% PEG 1500, 50% ethanol, and 3% methanol. Slides are place in the fixative for about 10 to 15 minutes at room temperature and then removed and allowed to dry. Once slides are fixed they can be rinsed with a buffered solution like PBS.
  • a wide range of dyes can be used to differentially highlight and contrast or “stain” cellular, sub-cellular, and tissue features or morphological structures.
  • Hematoylin is used to stain nuclei a blue or black color.
  • Orange G-6 and Eosin Azure both stain the cell's cytoplasm.
  • Orange G stains keratin and glycogen containing cells yellow.
  • Eosin Y is used to stain nucleoli, cilia, red blood cells, and superficial epithelial squamous cells.
  • Romanowsky stains are used for air dried slides and are useful in enhancing pleomorphism and distinguishing extracellular from intracytoplasmic material.
  • the staining process can include a treatment to increase the permeability of the cells to the stain.
  • Treatment of the cells with a detergent can be used to increase permeability.
  • fixed samples can be further treated with solvents, saponins, or non-ionic detergents. Enzymatic digestion can also improve the accessibility of specific targets in a tissue sample.
  • the sample is dehydrated using a succession of alcohol rinses with increasing alcohol concentration.
  • the final wash is done with xylene or a xylene substitute, such as a citrus terpene, that has a refractive index close to that of the coverslip to be applied to the slide. This final step is referred to as clearing.
  • a mounting medium is applied. The mounting medium is selected to have a refractive index close to the glass and is capable of bonding the coverslip to the slide. It will also inhibit the additional drying, shrinking, or fading of the cell sample.
  • the final evaluation of the lung cytological specimen is made by some type of microscopy to permit a visual inspection of the morphology and a determination of the marker's presence or absence.
  • exemplary microscopic methods include brightfield, phase contrast, fluorescence, and differential interference contrast.
  • the coverslip may be removed and the slide destained. Destaining involves using the original solvent systems used in staining the slide originally without the added dye and in a reverse order to the original staining procedure. Destaining may also be completed by soaking the slide in an acid alcohol until the cells are colorless. Once colorless the slides are rinsed well in a water bath and the second staining procedure applied.
  • specific molecular differentiation may be possible in conjunction with the cellular morphological analysis through the use of specific molecular reagents such as antibodies or nucleic acid probes or aptamers. This improves the accuracy of diagnostic cytology.
  • Micro-dissection can be used to isolate a subset of cells for additional evaluation, in particular, for genetic evaluation of abnormal chromosomes, gene expression, or mutations.
  • Preparation of a tissue sample for histological evaluation involves fixation, dehydration, infiltration, embedding, and sectioning.
  • the fixation reagents used in histology are very similar or identical to those used in cytology and have the same issues of preserving morphological features at the expense of molecular ones such as individual proteins.
  • Time can be saved if the tissue sample is not fixed and dehydrated but instead is frozen and then sectioned while frozen. This is a more gentle processing procedure and can preserve more individual markers.
  • freezing is not acceptable for long term storage of a tissue sample as subcellular information is lost due to the introduction of ice crystals. Ice in the frozen tissue sample also prevents the sectioning process from producing a very thin slice and thus some microscopic resolution and imaging of subcellular structures can be lost.
  • osmium tetroxide is used to fix and stain phospholipids (membranes).
  • Dehydration of tissues is accomplished with successive washes of increasing alcohol concentration. Clearing employs a material that is miscible with alcohol and the embedding material and involves a stepwise process starting at 50:50 alcohol:clearing reagent and then 100% clearing agent (xylene or xylene substitute). Infiltration involves incubating the tissue with a liquid form of the embedding agent (warm wax, nitrocellulose solution) first at 50:50 embedding agent: clearing agent and the 100% embedding agent. Embedding is completed by placing the tissue in a mold or cassette and filling with melted embedding agent such as wax, agar, or gelatin. The embedding agent is allowed to harden. The hardened tissue sample may then be sliced into thin section for staining and subsequent examination.
  • the tissue section Prior to staining, the tissue section is dewaxed and rehydrated. Xylene is used to dewax the section, one or more changes of xylene may be used, and the tissue is rehydrated by successive washes in alcohol of decreasing concentration. Prior to dewax, the tissue section may be heat immobilized to a glass slide at about 80° C. for about 20 minutes.
  • Laser capture micro-dissection allows the isolation of a subset of cells for further analysis from a tissue section.
  • the tissue section or slice can be stained with a variety of stains.
  • a large menu of commercially available stains can be used to enhance or identify specific features.
  • the first such technique uses high temperature heating of a fixed sample. This method is also referred to as heat-induced epitope retrieval or HIER.
  • HIER heat-induced epitope retrieval
  • a variety of heating techniques have been used, including steam heating, microwaving, autoclaving, water baths, and pressure cooking or a combination of these methods of heating.
  • Analyte retrieval solutions include, for example, water, citrate, and normal saline buffers.
  • the key to analyte retrieval is the time at high temperature but lower temperatures for longer times have also been successfully used.
  • Another key to analyte retrieval is the pH of the heating solution.
  • the section is first dewaxed and hydrated.
  • the slide is then placed in 10 mM sodium citrate buffer pH 6.0 in a dish or jar.
  • a representative procedure uses an 1100 W microwave and microwaves the slide at 100% power for 2 minutes followed by microwaving the slides using 20% power for 18 minutes after checking to be sure the slide remains covered in liquid.
  • the slide is then allowed to cool in the uncovered container and then rinsed with distilled water.
  • HIER may be used in combination with an enzymatic digestion to improve the reactivity of the target to immunochemical reagents.
  • One such enzymatic digestion protocol uses proteinase K.
  • a 20 ⁇ g/ml concentration of proteinase K is prepared in 50 mM Tris Base, 1 mM EDTA, 0.5% Triton X-100, pH 8.0 buffer.
  • the process first involves dewaxing sections in 2 changes of xylene, 5 minutes each. Then the sample is hydrated in 2 changes of 100% ethanol for 3 minutes each, 95% and 80% ethanol for 1 minute each, and then rinsed in distilled water. Sections are covered with Proteinase K working solution and incubated 10-20 minutes at 37° C. in humidified chamber (optimal incubation time may vary depending on tissue type and degree of fixation).
  • the sections are cooled at room temperature for 10 minutes and then rinsed in PBS Tween 20 for 2 ⁇ 2 min. If desired, sections can be blocked to eliminate potential interference from endogenous compounds and enzymes.
  • the section is then incubated with primary antibody at appropriate dilution in primary antibody dilution buffer for 1 hour at room temperature or overnight at 4° C.
  • the section is then rinsed with PBS Tween 20 for 2 ⁇ 2 min. Additional blocking can be performed, if required for the specific application, followed by additional rinsing with PBS Tween 20 for 3 ⁇ 2 min and then finally the immunostaining protocol completed.
  • a simple treatment with 1% SDS at room temperature has also been demonstrated to improve immunohistochemical staining.
  • Analyte retrieval methods have been applied to slide mounted sections as well as free floating sections.
  • Another treatment option is to place the slide in a jar containing citric acid and 0.1 Nonident P40 at pH 6.0 and heating to 95° C. The slide is then washed with a buffer solution like PBS.
  • tissue proteins For immunological staining of tissues it may be useful to block non-specific association of the antibody with tissue proteins by soaking the section in a protein solution like serum or non-fat dry milk.
  • Blocking reactions may include the need to reduce the level of endogenous biotin; eliminate endogenous charge effects; inactivate endogenous nucleases; and/or inactivate endogenous enzymes like peroxidase and alkaline phosphatase.
  • Endogenous nucleases may be inactivated by degradation with proteinase K, by heat treatment, use of a chelating agent such as EDTA or EGTA, the introduction of carrier DNA or RNA, treatment with a chaotrope such as urea, thiourea, guanidine hydrochloride, guanidine thiocyanate, lithium perchlorate, etc, or diethyl pyrocarbonate.
  • Alkaline phosphatase may be inactivated by treated with 0.1N HCl for 5 minutes at room temperature or treatment with 1 mM levamisole. Peroxidase activity may be eliminated by treatment with 0.03% hydrogen peroxide.
  • Endogenous biotin may be blocked by soaking the slide or section in an avidin (streptavidin, neutravidin may be substituted) solution for at least 15 minutes at room temperature. The slide or section is then washed for at least 10 minutes in buffer. This may be repeated at least three times. Then the slide or section is soaked in a biotin solution for 10 minutes. This may be repeated at least three times with a fresh biotin solution each time. The buffer wash procedure is repeated.
  • Blocking protocols should be minimized to prevent damaging either the cell or tissue structure or the target or targets of interest but one or more of these protocols could be combined to “block” a slide or section prior to reaction with one or more slow off-rate aptamers. See Basic Medical Histology: the Biology of Cells, Tissues and Organs, authored by Richard G. Kessel, Oxford University Press, 1998.
  • mass spectrometers can be used to detect biomarker values.
  • Several types of mass spectrometers are available or can be produced with various configurations.
  • a mass spectrometer has the following major components: a sample inlet, an ion source, a mass analyzer, a detector, a vacuum system, and instrument-control system, and a data system. Difference in the sample inlet, ion source, and mass analyzer generally define the type of instrument and its capabilities.
  • an inlet can be a capillary-column liquid chromatography source or can be a direct probe or stage such as used in matrix-assisted laser desorption.
  • Common ion sources are, for example, electrospray, including nanospray and microspray or matrix-assisted laser desorption.
  • Common mass analyzers include a quadrupole mass filter, ion trap mass analyzer and time-of-flight mass analyzer. Additional mass spectrometry methods are well known in the art (see Burlingame et al. Anal. Chem. 70:647 R-716R (1998); Kinter and Sherman. Protein sequencing and identification using tandem mass spectrometry. New York: Wiley-Interscience (2000).
  • Protein biomarkers and biomarker values can be detected and measured by any of the following: electrospray ionization mass spectrometry (ESI-MS), ESI-MS/MS, ESI-MS/(MS)n, matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS), surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF-MS), desorption/ionization on silicon (DIOS), secondary ion mass spectrometry (SIMS), quadrupole time-of-flight (Q-TOF), tandem time-of-flight (TOF/TOF) technology, called ultraflex III TOF/TOF, atmospheric pressure chemical ionization mass spectrometry (APCI-MS), APCI-MS/MS, APCI-(MS) N , atmospheric pressure photoionization mass spectrometry (APPI-MS), APPI-MS/MS
  • Labeling methods include but are not limited to isobaric tag for relative and absolute quantitation (iTRAQ) and stable isotope labeling with amino acids in cell culture (SILAC).
  • Capture reagents used to selectively enrich samples for candidate biomarker proteins prior to mass spectroscopic analysis include but are not limited to aptamers, antibodies, nucleic acid probes, chimeras, small molecules, an F(ab′) 2 fragment, a single chain antibody fragment, an Fv fragment, a single chain Fv fragment, a nucleic acid, a lectin, a ligand-binding receptor, affybodies, nanobodies, ankyrins, domain antibodies, alternative antibody scaffolds (e.g.
  • the foregoing assays enable the detection of biomarker values that are useful in methods for diagnosing lung cancer, where the methods comprise detecting, in a biological sample from an individual, at least N biomarker values that each correspond to a biomarker selected from the group consisting of the biomarkers provided in Tables 18, 20 or 21, wherein a classification, as described in detail below, using the biomarker values indicates whether the individual has lung cancer. While certain of the described lung cancer biomarkers are useful alone for detecting and diagnosing lung cancer, methods are also described herein for the grouping of multiple subsets of the lung cancer biomarkers that are each useful as a panel of three or more biomarkers. Thus, various embodiments of the instant application provide combinations comprising N biomarkers, wherein N is at least three biomarkers.
  • N is selected to be any number from 2-86 biomarkers. It will be appreciated that N can be selected to be any number from any of the above described ranges, as well as similar, but higher order, ranges.
  • biomarker values can be detected and classified individually or they can be detected and classified collectively, as for example in a multiplex assay format.
  • methods for detecting an absence of lung cancer, the methods comprising detecting, in a biological sample from an individual, at least N biomarker values that each correspond to a biomarker selected from the group consisting of the biomarkers provided in Tables 18, 20 or 21, wherein a classification, as described in detail below, of the biomarker values indicates an absence of lung cancer in the individual. While certain of the described lung cancer biomarkers are useful alone for detecting and diagnosing the absence of lung cancer, methods are also described herein for the grouping of multiple subsets of the lung cancer biomarkers that are each useful as a panel of three or more biomarkers. Thus, various embodiments of the instant application provide combinations comprising N biomarkers, wherein N is at least three biomarkers.
  • N is selected to be any number from 2-86 biomarkers. It will be appreciated that N can be selected to be any number from any of the above described ranges, as well as similar, but higher order, ranges.
  • biomarker values can be detected and classified individually or they can be detected and classified collectively, as for example in a multiplex assay format.
  • a biomarker “signature” for a given diagnostic test contains a set of markers, each marker having different levels in the populations of interest. Different levels, in this context, may refer to different means of the marker levels for the individuals in two or more groups, or different variances in the two or more groups, or a combination of both.
  • these markers can be used to assign an unknown sample from an individual into one of two groups, either diseased or not diseased.
  • classification The assignment of a sample into one of two or more groups is known as classification, and the procedure used to accomplish this assignment is known as a classifier or a classification method. Classification methods may also be referred to as scoring methods. There are many classification methods that can be used to construct a diagnostic classifier from a set of biomarker values.
  • classification methods are most easily performed using supervised learning techniques where a data set is collected using samples obtained from individuals within two (or more, for multiple classification states) distinct groups one wishes to distinguish. Since the class (group or population) to which each sample belongs is known in advance for each sample, the classification method can be trained to give the desired classification response. It is also possible to use unsupervised learning techniques to produce a diagnostic classifier.
  • diagnostic classifiers include decision trees; bagging+boosting+forests; rule inference based learning; Parzen Windows; linear models; logistic; neural network methods; unsupervised clustering; K-means; hierarchical ascending/descending; semi-supervised learning; prototype methods; nearest neighbor; kernel density estimation; support vector machines; hidden Markov models; Boltzmann Learning; and classifiers may be combined either simply or in ways which minimize particular objective functions.
  • Pattern Classification R. O. Duda, et al., editors, John Wiley & Sons, 2nd edition, 2001
  • training data includes samples from the distinct groups (classes) to which unknown samples will later be assigned.
  • samples collected from individuals in a control population and individuals in a particular disease population can constitute training data to develop a classifier that can classify unknown samples (or, more particularly, the individuals from whom the samples were obtained) as either having the disease or being free from the disease.
  • the development of the classifier from the training data is known as training the classifier.
  • Specific details on classifier training depend on the nature of the supervised learning technique. For purposes of illustration, an example of training a na ⁇ ve Bayesian classifier will be described below (see, e.g., Pattern Classification, R. O.
  • Over-fitting occurs when a statistical model describes random error or noise instead of the underlying relationship. Over-fitting can be avoided in a variety of way, including, for example, by limiting the number of markers used in developing the classifier, by assuming that the marker responses are independent of one another, by limiting the complexity of the underlying statistical model employed, and by ensuring that the underlying statistical model conforms to the data.
  • An illustrative example of the development of a diagnostic test using a set of biomarkers includes the application of a na ⁇ ve Bayes classifier, a simple probabilistic classifier based on Bayes theorem with strict independent treatment of the biomarkers.
  • Each biomarker is described by a class-dependent probability density function (pdf) for the measured RFU values or log RFU (relative fluorescence units) values in each class.
  • the joint pdfs for the set of markers in one class is assumed to be the product of the individual class-dependent pdfs for each biomarker.
  • Training a na ⁇ ve Bayes classifier in this context amounts to assigning parameters (“parameterization”) to characterize the class dependent pdfs. Any underlying model for the class-dependent pdfs may be used, but the model should generally conform to the data observed in the training set.
  • the class-dependent probability of measuring a value x i for biomarker i in the disease class is written as p(x i
  • d) and the overall na ⁇ ve Bayes probability of observing n markers with values ⁇ tilde under (x) ⁇ (x 1 , x 2 , . . . x n ) is written as
  • the individual x i s are the measured biomarker levels in RFU or log RFU.
  • the classification assignment for an unknown is facilitated by calculating the probability of being diseased p(d
  • the ratio of these probabilities is computed from the class-dependent pdfs by application of Bayes theorem, i.e.,
  • log likelihood ratio This form is known as the log likelihood ratio and simply states that the log likelihood of being free of the particular disease versus having the disease and is primarily composed of the sum of individual log likelihood ratios of the n individual biomarkers.
  • an unknown sample or, more particularly, the individual from whom the sample was obtained is classified as being free of the disease if the above ratio is greater than zero and having the disease if the ratio is less than zero.
  • d) are assumed to be normal or log-normal distributions in the measured RFU values x i , i.e.
  • the Bayes classifier is fully determined and may be used to classify unknown samples with measured values ⁇ tilde under (x) ⁇ .
  • the performance of the na ⁇ ve Bayes classifier is dependent upon the number and quality of the biomarkers used to construct and train the classifier.
  • a single biomarker will perform in accordance with its KS-distance (Kolmogorov-Smirnov), as defined in Example 3, below. If a classifier performance metric is defined as the sum of the sensitivity (fraction of true positives, f TP ) and specificity (one minus the fraction of false positives, 1 ⁇ f FP ), a perfect classifier will have a score of two and a random classifier, on average, will have a score of one. Using the definition of the KS-distance, that value x* which maximizes the difference in the cdf functions can be found by solving
  • d ) ⁇ ⁇ x 1 - ⁇ x * ⁇ ⁇ p ⁇ ( x
  • d ) ⁇ ⁇ x 1 - f FP - f FN ,
  • Example 4 The algorithm approach used here is described in detail in Example 4. Briefly, all single analyte classifiers are generated from a table of potential biomarkers and added to a list. Next, all possible additions of a second analyte to each of the stored single analyte classifiers is then performed, saving a predetermined number of the best scoring pairs, say, for example, a thousand, on a new list. All possible three marker classifiers are explored using this new list of the best two-marker classifiers, again saving the best thousand of these. This process continues until the score either plateaus or begins to deteriorate as additional markers are added. Those high scoring classifiers that remain after convergence can be evaluated for the desired performance for an intended use.
  • classifiers with a high sensitivity and modest specificity may be more desirable than modest sensitivity and high specificity.
  • classifiers with a high specificity and a modest sensitivity may be more desirable.
  • the desired level of performance is generally selected based upon a trade-off that must be made between the number of false positives and false negatives that can each be tolerated for the particular diagnostic application. Such trade-offs generally depend on the medical consequences of an error, either false positive or false negative.
  • Various other techniques are known in the art and may be employed to generate many potential classifiers from a list of biomarkers using a na ⁇ ve Bayes classifier.
  • a genetic algorithm can be used to combine different markers using the fitness score as defined above. Genetic algorithms are particularly well suited to exploring a large diverse population of potential classifiers.
  • so-called ant colony optimization can be used to generate sets of classifiers.
  • Other strategies that are known in the art can also be employed, including, for example, other evolutionary strategies as well as simulated annealing and other stochastic search methods. Metaheuristic methods, such as, for example, harmony search may also be employed.
  • Exemplary embodiments use any number of the lung cancer biomarkers listed in Tables 18, 20 or 21 in various combinations to produce diagnostic tests for detecting lung cancer (see Examples 2 and 6 for a detailed description of how these biomarkers were identified).
  • a method for diagnosing lung cancer uses a na ⁇ ve Bayes classification method in conjunction with any number of the lung cancer biomarkers listed in Tables 18, 20 or 21.
  • the score for a classifier constructed of two biomarkers is not a simple sum of the KS-distances; KS-distances are not additive when combining biomarkers and it takes many more weak markers to achieve the same level of performance as a strong marker.
  • Adding additional biomarkers such as, for example, PTN, BTK, CD30, Kallikrein 7, LRIG3, LDH-H1, and PARC, produces a series of lung cancer tests summarized in Table 14 and displayed as a series of ROC curves in FIG. 3 .
  • the score of the classifiers as a function of the number of analytes used in classifier construction is displayed in FIG. 4 .
  • the sensitivity and specificity of this exemplary ten-marker classifier is >87% and the AUC is 0.91.
  • panels of biomarkers are comprised of different numbers of analytes depending on a specific diagnostic performance criterion that is selected. For example, certain combinations of biomarkers will produce tests that are more sensitive (or more specific) than other combinations.
  • a panel is defined to include a particular set of biomarkers from Tables 18, 20 or 21 and a classifier is constructed from a set of training data
  • the definition of the diagnostic test is complete.
  • the procedure used to classify an unknown sample is outlined in FIG. 1A .
  • the procedure used to classify an unknown sample is outlined in FIG. 1B .
  • the biological sample is appropriately diluted and then run in one or more assays to produce the relevant quantitative biomarker levels used for classification.
  • the measured biomarker levels are used as input for the classification method that outputs a classification and an optional score for the sample that reflects the confidence of the class assignment.
  • Table 21 identifies eighty-six biomarkers that are useful for diagnosing lung cancer in both tissue and blood samples.
  • Table 20 identifies twenty-five biomarkers that were identified in tissue samples, but which are useful in serum and plasma samples as well. This is a surprisingly larger number than expected when compared to what is typically found during biomarker discovery efforts and may be attributable to the scale of the described study, which encompassed over 800 proteins measured in hundreds of individual samples, in some cases at concentrations in the low femtomolar range.
  • the large number of discovered biomarkers reflects the diverse biochemical pathways implicated in both tumor biology and the body's response to the tumor's presence; each pathway and process involves many proteins. The results show that no single protein of a small group of proteins is uniquely informative about such complex processes; rather, that multiple proteins are involved in relevant processes, such as apoptosis or extracellular matrix repair, for example.
  • Example 4 The results from Example 4 suggest certain possible conclusions: First, the identification of a large number of biomarkers enables their aggregation into a vast number of classifiers that offer similarly high performance. Second, classifiers can be constructed such that particular biomarkers may be substituted for other biomarkers in a manner that reflects the redundancies that undoubtedly pervade the complexities of the underlying disease processes. That is to say, the information about the disease contributed by any individual biomarker identified in Table 1 overlaps with the information contributed by other biomarkers, such that it may be that no particular biomarker or small group of biomarkers in Table 1 must be included in any classifier.
  • Exemplary embodiments use na ⁇ ve Bayes classifiers constructed from the data in Tables 38 and 39 to classify an unknown sample.
  • the procedure is outlined in FIGS. 1A and B.
  • the biological sample is optionally diluted and run in a multiplexed aptamer assay.
  • the data from the assay are normalized and calibrated as outlined in Example 3, and the resulting biomarker levels are used as input to a Bayes classification scheme.
  • the log-likelihood ratio is computed for each measured biomarker individually and then summed to produce a final classification score, which is also referred to as a diagnostic score.
  • the resulting assignment as well as the overall classification score can be reported.
  • the individual log-likelihood risk factors computed for each biomarker level can be reported as well.
  • the details of the classification score calculation are presented in Example 3.
  • aptamer-based proteomic technology for use in discovery of disease-related biomarkers from tissues
  • homogenized tissues samples from surgical resections obtained from eight non-small cell lung cancer (NSCLC) patients were analyzed, as described in Example 6. All NSCLC patients were smokers, ranging in age from 47 to 75 years old and covering NSCLC stages 1A through 3B (Table 17). Three samples were obtained from each resection: tumor tissue sample, adjacent non-tumor tissue.
  • the protein concentration measurements allow large-scale comparisons of protein signatures among samples (see FIG. 21 ).
  • ROU relative fluorescence units
  • NSCLC tissue biomarkers protein expression levels between tumor, adjacent and distant tissue samples were compared using the Mann-Whitney test as described in Ostroff et al. Nature Precedings, http://precedings.nature.com/documents/4537/version/1 (2010)). Thirty-six proteins with the greatest fold change and with statistically significant differences between tumor and non-tumor tissue were identified with a false discovery rate cutoff of q ⁇ 0.05 for significance FIGS. 23 and 24 , and Table 18). Twenty of these proteins were up-regulated and 16 were down-regulated in tumor tissue. Although the number of samples used for this study was relatively small, a powerful individual-based study design in which each tumor sample had its own healthy tissue controls was employed.
  • the biomarkers identified according to the method of Example 6 can be classified broadly into four biological processes associated with important hallmarks of tumor biology (Hanahan & Weinberg (2011) Cell 144:646-74) as shown in Table 19: 1) angiogenesis, 2) growth and metabolism, 3) inflammation and apoptosis, and 4) invasion and metastasis. Admittedly, these are convenient albeit inexact classifications that approximate a highly complex and dynamic system in which these molecules often play multiple and nuanced roles. Therefore, the specific state of a given system ultimately affects the expression and function of any particular molecule. Biological understanding is far from complete in these systems. With the SOMAscan platform, the quantitative expression of large numbers of proteins in various tissues and disease processes is made possible. These data provide new coordinates to help map the dynamics of these systems, which in turn will provide a more complete understanding of the biology of lung cancer as well as other diseases. The results from the current study provide a new perspective on NSCLC tumor biology, with both familiar and new elements.
  • Angiogenesis drives growth of new blood vessels to support tumor growth and metabolism.
  • the regulation of angiogenesis is a complex biological phenomenon controlled by both positive and negative signals (Hanahan & Weinberg, (2011) Cell 144:646-74).
  • NSCLC tissue biomarkers identified in this study were well known positive and negative angiogenesis regulators ( FIGS. 23 and 24 and Table 19), all of which have been observed previously in NSCLC tumor tissue (Fontanini et al. (1999) British Journal of Cancer 79(2):363-369; Imoto et al. (1998) J. Thorac. Carciovasc. Surg 115:1007-1011; Ohta et al. (2006) Ann. Thorac. Surg.
  • VEGF endostatin and thrombospondin-1
  • TSP-1 endostatin and thrombospondin-1
  • VEGF is a powerful growth factor that promotes new blood vessel growth and was strongly up-regulated in NSCLC tumor tissue, consistent with previous observations (Imoto et al. (1998) J. Thorac. Carciovasc. Surg 115:1007-1011), and including our study of serum samples from NSCLC patients (Ostroff et al. Nature Precedings, http://precedings.nature.com/documents/4537/version/1 (2010)).
  • Endostatin is a proteolytic fragment of Collagen XVIII and a strong inhibitor of endothelial cell proliferation and angiogenesis (Iizasa et al. (August 2004) Clinical Cancer Research 10:5361-5366).
  • TSP-1 and the related thrombospondin-2 (TSP-2) were substantially up-regulated in NSCLC tumor tissue.
  • TSP-1 and TSP-2 are extracellular matrix proteins with complex, context-dependent effects modulated through a variety of interactions with cell-surface receptors, growth factors, cytokines, matrix metalloproteinases, and other molecules.
  • TSP-1 and TSP-2 inhibit angiogenesis by inhibiting endothelial cell proliferation through the CD47 receptor and inducing endothelial cell apoptosis through the CD36 receptor.
  • proangiogenic influences for TSP-1 and TSP-2 (Bornstein (2009) J. Cell Commun. Signal. 3(3-4):189-200).
  • TSP-1 and TSP-2 relative and absolute expression levels in NSCLC tissue vary (Chijiwa et al. (2009) Oncology Reports 22:279-283; Chen et al. (2009) J Int Med Res 37:551-556; Oshika 1998, Fontanini et al.
  • IGFBP-2 insulin-like growth factor binding proteins
  • chemokines such as BCA-1, CXCL16, IL-8, and NAP-2, are altered (Table 18), consistent with the hypothesis that invasion of tumors with cells from the innate and adaptive arms of the immune system provide bioactive molecules that affect proliferative and angiogenic signals (Hanahan & Weinberg (2011) Cell 144:646-74).
  • the largest group of potential biomarkers contains proteins that function in cell-cell and cell-matrix interactions and are involved in invasion and metastasis. Many have been previously reported to be associated with NSCLC. Most notable are two of the matrix metalloproteases, MMP-7 and MMP-12, which contribute to proteolytic degradation of extracellular matrix components and processing of substrates such as growth factors (see e.g. Su et al. (2004) Chinese Journal of Clinical Oncology 1(2):126-130; Wegmann et al. (1993) Eur. J. Cancer 29A(11):1578-1584). Such processes are well known to play a role in creating tumor microenvironments.
  • MMP-7 and MMP-12 were up-regulated in NSCLC tissue (Table 18), which is consistent with similar study that used antibody-based measurements (Shah et al. (2010) The Journal of Thoracic and Cardiovascular Surgery 139(4):984-990).
  • the over-expression of MMP-7 and MMP-12 has been associated with poor prognosis in NSCLC (Shah et al. (2010) The Journal of Thoracic and Cardiovascular Surgery 139(4):984-990).
  • MMP-12 levels have been correlated with local recurrence and metastatic disease (Hofmann et al. (2005) Clin. Cancer Res. 11:1086-92, Hoffman et al. (2006) Oncol. Rep. 16:587-95).).
  • Two of the eight subjects studied had normal levels of MMP-12, whereas the other six had 15-50 ⁇ elevation of MMP-12 in tumor tissue compared to non-tumor tissue.
  • FIG. 25 demonstrates the ability of two of the identified SOMAmers to stain fresh frozen tissues obtained from the same tumor resections used for the discovery of these biomarkers.
  • TSP2 Thrombospondin-2
  • MRC1 macrophage mannose receptor
  • one third, or twelve of the thirty-six tissue biomarkers reported herein (BCA-1 (BCL), cadherin-1 (cadherin-E), catalase, endostatin, IGFBP-2, MRC1 (macrophage mannose receptor), MAPK-13 (MK13), MMP-7, MMP-12, NAGK, VEGF and YES have been previously identified in serum.
  • BCA-1 BCL
  • cadherin-1 cadherin-1
  • cadherin-E catalase
  • endostatin IGFBP-2
  • MRC1 macrophage mannose receptor
  • MAPK-13 MK13
  • MMP-7 MMP-7
  • MMP-12 MMP-12
  • NAGK vascular endothelial growth factor
  • VEGF vascular endothelial growth factor
  • any combination of the biomarkers of Table 20 can be detected using a suitable kit, such as for use in performing the methods disclosed herein.
  • a suitable kit such as for use in performing the methods disclosed herein.
  • any kit can contain one or more detectable labels as described herein, such as a fluorescent moiety, etc.
  • a kit in one embodiment, includes (a) one or more capture reagents (such as, for example, at least one aptamer or antibody) for detecting one or more biomarkers in a biological sample, wherein the biomarkers include any of the biomarkers set forth in Tables 18, 20 or 21 and optionally (b) one or more software or computer program products for classifying the individual from whom the biological sample was obtained as either having or not having lung cancer or for determining the likelihood that the individual has lung cancer, as further described herein.
  • one or more instructions for manually performing the above steps by a human can be provided.
  • kit The combination of a solid support with a corresponding capture reagent and a signal generating material is referred to herein as a “detection device” or “kit”.
  • the kit can also include instructions for using the devices and reagents, handling the sample, and analyzing the data. Further the kit may be used with a computer system or software to analyze and report the result of the analysis of the biological sample.
  • kits can also contain one or more reagents (e.g., solubilization buffers, detergents, washes, or buffers) for processing a biological sample.
  • reagents e.g., solubilization buffers, detergents, washes, or buffers
  • Any of the kits described herein can also include, e.g., buffers, blocking agents, mass spectrometry matrix materials, antibody capture agents, positive control samples, negative control samples, software and information such as protocols, guidance and reference data.
  • kits for the analysis of lung cancer status include PCR primers for one or more biomarkers selected from Tables 18, 20, or 21.
  • the kit may further include instructions for use and correlation of the biomarkers with lung cancer.
  • the kit may also include a DNA array containing the complement of one or more of the biomarkers selected from Table 20, reagents, and/or enzymes for amplifying or isolating sample DNA.
  • the kits may include reagents for real-time PCR, for example, TaqMan probes and/or primers, and enzymes.
  • a kit can comprise (a) reagents comprising at least capture reagent for quantifying one or more biomarkers in a test sample, wherein said biomarkers comprise the biomarkers set forth in Tables 18, 20, or 21, or any other biomarkers or biomarkers panels described herein, and optionally (b) one or more algorithms or computer programs for performing the steps of comparing the amount of each biomarker quantified in the test sample to one or more predetermined cutoffs and assigning a score for each biomarker quantified based on said comparison, combining the assigned scores for each biomarker quantified to obtain a total score, comparing the total score with a predetermined score, and using said comparison to determine whether an individual has lung cancer.
  • one or more instructions for manually performing the above steps by a human can be provided.
  • a method for diagnosing an individual can comprise the following: 1) collect or otherwise obtain a biological sample; 2) perform an analytical method to detect and measure the biomarker or biomarkers in the panel in the biological sample; 3) perform any data normalization or standardization required for the method used to collect biomarker values; 4) calculate the marker score; 5) combine the marker scores to obtain a total diagnostic score; and 6) report the individual's diagnostic score.
  • the diagnostic score may be a single number determined from the sum of all the marker calculations that is compared to a preset threshold value that is an indication of the presence or absence of disease.
  • the diagnostic score may be a series of bars that each represent a biomarker value and the pattern of the responses may be compared to a pre-set pattern for determination of the presence or absence of disease.
  • FIG. 6 An example of a computer system 100 is shown in FIG. 6 .
  • system 100 is shown comprised of hardware elements that are electrically coupled via bus 108 , including a processor 101 , input device 102 , output device 103 , storage device 104 , computer-readable storage media reader 105 a , communications system 106 processing acceleration (e.g., DSP or special-purpose processors) 107 and memory 109 .
  • Computer-readable storage media reader 105 a is further coupled to computer-readable storage media 105 b , the combination comprehensively representing remote, local, fixed and/or removable storage devices plus storage media, memory, etc.
  • System 100 for temporarily and/or more permanently containing computer-readable information, which can include storage device 104 , memory 109 and/or any other such accessible system 100 resource.
  • System 100 also comprises software elements (shown as being currently located within working memory 191 ) including an operating system 192 and other code 193 , such as programs, data and the like.
  • system 100 has extensive flexibility and configurability.
  • a single architecture might be utilized to implement one or more servers that can be further configured in accordance with currently desirable protocols, protocol variations, extensions, etc.
  • embodiments may well be utilized in accordance with more specific application requirements.
  • one or more system elements might be implemented as sub-elements within a system 100 component (e.g., within communications system 106 ).
  • Customized hardware might also be utilized and/or particular elements might be implemented in hardware, software or both.
  • connection to other computing devices such as network input/output devices (not shown) may be employed, it is to be understood that wired, wireless, modem, and/or other connection or connections to other computing devices might also be utilized.
  • the system can comprise a database containing features of biomarkers characteristic of lung cancer.
  • the biomarker data (or biomarker information) can be utilized as an input to the computer for use as part of a computer implemented method.
  • the biomarker data can include the data as described herein.
  • system further comprises one or more devices for providing input data to the one or more processors.
  • the system further comprises a memory for storing a data set of ranked data elements.
  • the device for providing input data comprises a detector for detecting the characteristic of the data element, e.g., such as a mass spectrometer or gene chip reader.
  • the system additionally may comprise a database management system.
  • User requests or queries can be formatted in an appropriate language understood by the database management system that processes the query to extract the relevant information from the database of training sets.
  • the system may be connectable to a network to which a network server and one or more clients are connected.
  • the network may be a local area network (LAN) or a wide area network (WAN), as is known in the art.
  • the server includes the hardware necessary for running computer program products (e.g., software) to access database data for processing user requests.
  • the system may include an operating system (e.g., UNIX or Linux) for executing instructions from a database management system.
  • the operating system can operate on a global communications network, such as the internet, and utilize a global communications network server to connect to such a network.
  • the system may include one or more devices that comprise a graphical display interface comprising interface elements such as buttons, pull down menus, scroll bars, fields for entering text, and the like as are routinely found in graphical user interfaces known in the art.
  • Requests entered on a user interface can be transmitted to an application program in the system for formatting to search for relevant information in one or more of the system databases.
  • Requests or queries entered by a user may be constructed in any suitable database language.
  • the graphical user interface may be generated by a graphical user interface code as part of the operating system and can be used to input data and/or to display inputted data.
  • the result of processed data can be displayed in the interface, printed on a printer in communication with the system, saved in a memory device, and/or transmitted over the network or can be provided in the form of the computer readable medium.
  • the system can be in communication with an input device for providing data regarding data elements to the system (e.g., expression values).
  • the input device can include a gene expression profiling system including, e.g., a mass spectrometer, gene chip or array reader, and the like.
  • the methods and apparatus for analyzing lung cancer biomarker information may be implemented in any suitable manner, for example, using a computer program operating on a computer system.
  • a conventional computer system comprising a processor and a random access memory, such as a remotely-accessible application server, network server, personal computer or workstation may be used.
  • Additional computer system components may include memory devices or information storage systems, such as a mass storage system and a user interface, for example a conventional monitor, keyboard and tracking device.
  • the computer system may be a stand-alone system or part of a network of computers including a server and one or more databases.
  • the lung cancer biomarker analysis system can provide functions and operations to complete data analysis, such as data gathering, processing, analysis, reporting and/or diagnosis.
  • the computer system can execute the computer program that may receive, store, search, analyze, and report information relating to the lung cancer biomarkers.
  • the computer program may comprise multiple modules performing various functions or operations, such as a processing module for processing raw data and generating supplemental data and an analysis module for analyzing raw data and supplemental data to generate a lung cancer status and/or diagnosis.
  • Diagnosing lung cancer status may comprise generating or collecting any other information, including additional biomedical information, regarding the condition of the individual relative to the disease, identifying whether further tests may be desirable, or otherwise evaluating the health status of the individual.
  • biomarker information can be retrieved for an individual.
  • the biomarker information can be retrieved from a computer database, for example, after testing of the individual's biological sample is performed.
  • a computer can be utilized to classify each of the biomarker values.
  • a determination can be made as to the likelihood that an individual has lung cancer based upon a plurality of classifications.
  • the indication can be output to a display or other indicating device so that it is viewable by a person. Thus, for example, it can be displayed on a display screen of a computer or other output device.
  • a computer can be utilized to retrieve biomarker information for an individual.
  • the biomarker information comprises a biomarker value corresponding to a biomarker selected from the group of biomarkers provided in Tables 18, 20 or 21.
  • a classification of the biomarker value can be performed with the computer.
  • an indication can be made as to the likelihood that the individual has lung cancer based upon the classification.
  • the indication can be output to a display or other indicating device so that it is viewable by a person. Thus, for example, it can be displayed on a display screen of a computer or other output device.
  • a computer program product may include a computer readable medium having computer readable program code embodied in the medium for causing an application program to execute on a computer with a database.
  • a “computer program product” refers to an organized set of instructions in the form of natural or programming language statements that are contained on a physical media of any nature (e.g., written, electronic, magnetic, optical or otherwise) and that may be used with a computer or other automated data processing system. Such programming language statements, when executed by a computer or data processing system, cause the computer or data processing system to act in accordance with the particular content of the statements.
  • Computer program products include without limitation: programs in source and object code and/or test or data libraries embedded in a computer readable medium.
  • the computer program product that enables a computer system or data processing equipment device to act in pre-selected ways may be provided in a number of forms, including, but not limited to, original source code, assembly code, object code, machine language, encrypted or compressed versions of the foregoing and any and all equivalents.
  • a computer program product for indicating a likelihood of lung cancer.
  • a computer program product for indicating a likelihood of lung cancer.
  • the embodiments may be embodied as code stored in a computer-readable memory of virtually any kind including, without limitation, RAM, ROM, magnetic media, optical media, or magneto-optical media. Even more generally, the embodiments could be implemented in software, or in hardware, or any combination thereof including, but not limited to, software running on a general purpose processor, microcode, PLAs, or ASICs.
  • embodiments could be accomplished as computer signals embodied in a carrier wave, as well as signals (e.g., electrical and optical) propagated through a transmission medium.
  • signals e.g., electrical and optical
  • the various types of information discussed above could be formatted in a structure, such as a data structure, and transmitted as an electrical signal through a transmission medium or stored on a computer readable medium.
  • This example describes the multiplex aptamer assay used to analyze the samples and controls for the identification of the biomarkers set forth in Table 1, Col. 2 (see FIG. 9 ).
  • the multiplexed analysis utilized 820 aptamers, each unique to a specific target.
  • aptamers without a photo-cleavable biotin linker custom stock aptamer solutions for 10%, 1% and 0.03% serum were prepared at 8 ⁇ concentration in 1 ⁇ SB17, 0.05% Tween-20 with appropriate photo-cleavable, biotinylated primers, where the resultant primer concentration was 3 times the relevant aptamer concentration.
  • the primers hybridized to all or part of the corresponding aptamer.
  • Each of the 3, 8 ⁇ aptamer solutions were diluted separately 1:4 into 1 ⁇ SB17, 0.05% Tween-20 (1500 ⁇ L of 8 ⁇ stock into 4500 ⁇ L of 1 ⁇ SB17, 0.05% Tween-20) to achieve a 2 ⁇ concentration.
  • Each diluted aptamer master mix was then split, 1500 ⁇ L each, into 4, 2 mL screw cap tubes and brought to 95° C. for 5 minutes, followed by a 37° C. incubation for 15 minutes.
  • a 20% sample solution was prepared by transferring 16 ⁇ L of sample using a 50 ⁇ L 8-channel spanning pipettor into 96-well Hybaid plates, each well containing 64 ⁇ L of the appropriate sample diluent at 4° C. (0.8 ⁇ SB17, 0.05% Tween-20, 2 ⁇ M Z-block — 2, 0.6 mM MgCl 2 for serum). This plate was stored on ice until the next sample dilution steps were initiated.
  • the 20% sample plate was briefly centrifuged and placed on the Beckman FX where it was mixed by pipetting up and down with the 96-well pipettor.
  • a 2% sample was then prepared by diluting 10 ⁇ L of the 20% sample into 90 ⁇ L of 1 ⁇ SB17, 0.05% Tween-20.
  • dilution of 6 ⁇ L of the resultant 2% sample into 194 ⁇ L of 1 ⁇ SB17, 0.05% Tween-20 made a 0.06% sample plate. Dilutions were done on the Beckman Biomek Fx P . After each transfer, the solutions were mixed by pipetting up and down.
  • the 3 sample dilution plates were then transferred to their respective aptamer solutions by adding 55 ⁇ L of the sample to 55 ⁇ L of the appropriate 2 ⁇ aptamer mix.
  • the sample and aptamer solutions were mixed on the robot by pipetting up and down.
  • the sample/aptamer plates were foil sealed and placed into a 37° C. incubator for 3.5 hours before proceeding to the Catch 1 step.
  • the beads were washed in the filter plates with 200 ⁇ L 1 ⁇ SB17, 0.05% Tween-20 and then resuspended in 200 ⁇ L 1 ⁇ SB17, 0.05% Tween-20.
  • the bottoms of the filter plates were blotted and the plates stored for use in the assay.
  • the cytomat was loaded with all tips, plates, all reagents in troughs (except NHS-biotin reagent which was prepared fresh right before addition to the plates), 3 prepared catch 1 filter plates and 1 prepared MyOne plate.
  • the sample/aptamer plates were removed from the incubator, centrifuged for about 1 minute, foil removed, and placed on the deck of the Beckman Biomek Fx P .
  • the Beckman Biomek Fx P program was initiated. All subsequent steps in Catch 1 were performed by the Beckman Biomek Fx P robot unless otherwise noted. Within the program, the vacuum was applied to the Catch 1 filter plates to remove the bead supernatant.
  • One hundred microlitres of each of the 10%, 1% and 0.03% equilibration binding reactions were added to their respective Catch 1 filtration plates, and each plate was mixed using an on-deck orbital shaker at 800 rpm for 10 minutes.
  • Unbound solution was removed via vacuum filtration.
  • the catch 1 beads were washed with 190 ⁇ L of 100 ⁇ M biotin in 1 ⁇ SB17, 0.05% Tween-20 followed by 190 ⁇ L of 1 ⁇ SB17, 0.05% Tween-20 by dispensing the solution and immediately drawing a vacuum to filter the solution through the plate.
  • the robot removed this wash via vacuum filtration and blotted the bottom of the filter plate to remove droplets using the on-deck blot station.
  • the NHS-PEO4-biotin reagent was dissolved at 100 mM concentration in anhydrous DMSO and had been stored frozen at ⁇ 20° C.
  • the diluted NHS-PEO4-biotin reagent was manually added to an on-deck trough and the robot program was manually re-initiated to dispense 100 ⁇ L of the NHS-PEO4-biotin into each well of each Catch 1 filter plate. This solution was allowed to incubate with Catch 1 beads shaking at 800 rpm for 5 minutes on the orbital shakers.
  • the tagging reaction was quenched by the addition of 150 ⁇ L of 20 mM glycine in 1 ⁇ SB17, 0.05% Tween-20 to the Catch 1 plates while still containing the NHS tag. The plates were then incubated for 1 minute on orbital shakers at 800 rpm. The NHS-tag/glycine solution was removed via vacuum filtration. Next, 190 ⁇ L 20 mM glycine (1 ⁇ SB17, 0.05% Tween-20) was added to each plate and incubated for 1 minute on orbital shakers at 800 rpm before removal by vacuum filtration.
  • the wells of the Catch 1 plates were subsequently washed three times by adding 190 ⁇ L 1 ⁇ SB17, 0.05% Tween-20, placing the plates on orbital shakers for 1 minute at 800 rpm followed by vacuum filtration. After the last wash the plates were placed on top of a 1 mL deep-well plate and removed from the deck. The Catch 1 plates were centrifuged at 1000 rpm for 1 minute to remove as much extraneous volume from the agarose beads before elution as possible.
  • the plates were placed back onto the Beckman Biomek Fx P and 85 ⁇ L of 10 mM DxSO 4 in 1 ⁇ SB17, 0.05% Tween-20 was added to each well of the filter plates.
  • the filter plates were removed from the deck, placed onto a Variomag Thermoshaker (Thermo Fisher Scientific, Inc., Waltham, Mass.) under the BlackRay (Ted Pella, Inc., Redding, Calif.) light sources, and irradiated for 10 minutes while shaking at 800 rpm.
  • Variomag Thermoshaker Thermo Fisher Scientific, Inc., Waltham, Mass.
  • BlackRay Ted Pella, Inc., Redding, Calif.
  • the photocleaved solutions were sequentially eluted from each Catch 1 plate into a common deep well plate by first placing the 10% Catch 1 filter plate on top of a 1 mL deep-well plate and centrifuging at 1000 rpm for 1 minute. The 1% and 0.03% catch 1 plates were then sequentially centrifuged into the same deep well plate.
  • the 1 mL deep well block containing the combined eluates of catch 1 was placed on the deck of the Beckman Biomek Fx P for catch 2.
  • the robot transferred all of the photo-cleaved eluate from the 1 mL deep-well plate onto the Hybaid plate containing the previously prepared catch 2 MyOne magnetic beads (after removal of the MyOne buffer via magnetic separation).
  • the solution was incubated while shaking at 1350 rpm for 5 minutes at 25° C. on a Variomag Thermoshaker (Thermo Fisher Scientific, Inc., Waltham, Mass.).
  • the robot transferred the plate to the on deck magnetic separator station.
  • the plate was incubated on the magnet for 90 seconds before removal and discarding of the supernatant.
  • the catch 2 plate was moved to the on-deck thermal shaker and 75 ⁇ L of 1 ⁇ SB17, 0.05% Tween-20 was transferred to each well.
  • the plate was mixed for 1 minute at 1350 rpm and 37° C. to resuspend and warm the beads.
  • 75 ⁇ L of 60% glycerol at 37° C. was transferred and the plate continued to mix for another minute at 1350 rpm and 37° C.
  • the robot transferred the plate to the 37° C. magnetic separator where it was incubated on the magnet for 2 minutes and then the robot removed and discarded the supernatant. These washes were repeated two more times.
  • the catch 2 beads were washed a final time using 150 ⁇ L 1 ⁇ SB19, 0.05% Tween-20 with incubation for 1 minute while shaking at 1350 rpm, prior to magnetic separation.
  • the aptamers were eluted from catch 2 beads by adding 105 ⁇ L of 100 mM CAPSO with 1M NaCl, 0.05% Tween-20 to each well. The beads were incubated with this solution with shaking at 1300 rpm for 5 minutes.
  • the catch 2 plate was then placed onto the magnetic separator for 90 seconds prior to transferring 90 ⁇ L of the eluate to a new 96-well plate containing 10 ⁇ L of 500 mM HCl, 500 mM HEPES, 0.05% Tween-20 in each well. After transfer, the solution was mixed robotically by pipetting 90 ⁇ L up and down five times.
  • the Beckman Biomek Fx P transferred 20 ⁇ L of the neutralized catch 2 eluate to a fresh Hybaid plate, and 5 ⁇ L of 10 ⁇ Agilent Block, containing a 10 ⁇ spike of hybridization controls, was added to each well.
  • 25 ⁇ L of 2 ⁇ Agilent Hybridization buffer was manually pipetted to the each well of the plate containing the neutralized samples and blocking buffer and the solution was mixed by manually pipetting 25 ⁇ L up and down 15 times slowly to avoid extensive bubble formation.
  • the plate was spun at 1000 rpm for 1 minute.
  • a gasket slide was placed into an Agilent hybridization chamber and 40 ⁇ L of each of the samples containing hybridization and blocking solution was manually pipetted into each gasket.
  • An 8-channel variable spanning pipettor was used in a manner intended to minimize bubble formation.
  • Custom Agilent microarray slides (Agilent Technologies, Inc., Santa Clara, Calif.), with their Number Barcode facing up, were then slowly lowered onto the gasket slides (see Agilent manual for detailed description).
  • the top of the hybridization chambers were placed onto the slide/backing sandwich and clamping brackets slid over the whole assembly. These assemblies were tightly clamped by turning the screws securely.
  • the assembled hybridization chambers were incubated in an Agilent hybridization oven for 19 hours at 60° C. rotating at 20 rpm.
  • a staining dish for Agilent Wash 2 was prepared by placing a stir bar into an empty glass staining dish.
  • a fourth glass staining dish was set aside for the final acetonitrile wash.
  • Each of six hybridization chambers was disassembled. One-by-one, the slide/backing sandwich was removed from its hybridization chamber and submerged into the staining dish containing Wash 1. The slide/backing sandwich was pried apart using a pair of tweezers, while still submerging the microarray slide. The slide was quickly transferred into the slide rack in the Wash 1 staining dish on the magnetic stir plate.
  • the slide rack was gently raised and lowered 5 times.
  • the magnetic stirrer was turned on at a low setting and the slides incubated for 5 minutes.
  • wash Buffer 2 pre-warmed to 37° C. in an incubator was added to the second prepared staining dish.
  • the slide rack was quickly transferred to Wash Buffer 2 and any excess buffer on the bottom of the rack was removed by scraping it on the top of the stain dish.
  • the slide rack was gently raised and lowered 5 times.
  • the magnetic stirrer was turned on at a low setting and the slides incubated for 5 minutes.
  • the slide rack was slowly pulled out of Wash 2, taking approximately 15 seconds to remove the slides from the solution.
  • acetonitrile ACN
  • the slide rack was transferred to the acetonitrile stain dish.
  • the slide rack was gently raised and lowered 5 times.
  • the magnetic stirrer was turned on at a low setting and the slides incubated for 5 minutes.
  • the slide rack was slowly pulled out of the ACN stain dish and placed on an absorbent towel. The bottom edges of the slides were quickly dried and the slide was placed into a clean slide box.
  • microarray slides were placed into Agilent scanner slide holders and loaded into the Agilent Microarray scanner according to the manufacturer's instructions.
  • the slides were imaged in the Cy3-channel at 5 ⁇ m resolution at the 100% PMT setting and the XRD option enabled at 0.05.
  • the resulting tiff images were processed using Agilent feature extraction software version 10.5.
  • the identification of potential lung cancer biomarkers was performed for three different diagnostic applications, diagnosis of suspicious nodules from a CT scan, screening of asymptomatic smokers for lung cancer, and diagnosing an individual with lung cancer. Serum samples were collected from four different sites in support of these three applications and include 48 NSCLC cases, 218 high risk controls composed of heavy smokers and patients with benign nodules.
  • the multiplexed aptamer affinity assay as described in Example 1 was used to measure and report the RFU value for 820 analytes in each of these 264 samples. The KS-test was then applied to each analyte.
  • the KS-distance (Kolmogorov-Smirnov statistic) between values from two sets of samples is a non parametric measurement of the extent to which the empirical distribution of the values from one set (Set A) differs from the distribution of values from the other set (Set B). For any value of a threshold T some proportion of the values from Set A will be less than T, and some proportion of the values from Set B will be less than T.
  • the KS-distance measures the maximum (unsigned) difference between the proportion of the values from the two sets for any choice of T.
  • This example describes the selection of biomarkers from Table 21 to form panels that can be used as classifiers in any of the methods described herein. Panels of biomarkers containing MMP-12 and Subsets of the biomarkers in Table 21 were selected to construct classifiers with good performance. This method was also used to determine which potential markers were included as biomarkers in Example 2.
  • the measure of classifier performance used here is the area under the ROC curve (AUC); a performance of 0.5 is the baseline expectation for a random (coin toss) classifier, a classifier worse than random would score between 0.0 and 0.5, a classifier with better than random performance would score between 0.5 and 1.0. A perfect classifier with no errors would have a sensitivity of 1.0, a specificity of 1.0 and an AUC of 1.0.
  • any weighting scheme which results in a single performance measure can be used.
  • Different applications will have different benefits for true positive and true negative findings, and also different costs associated with false positive findings from false negative findings. For example, screening asymptomatic smokers and the differential diagnosis of benign nodules found on CT will not in general have the same optimal trade-off between specificity and sensitivity.
  • the different demands of the two tests will in general require setting different weighting to positive and negative misclassifications, reflected in the performance measure. Changing the performance measure will in general change the exact subset of markers selected from Table 21 for a given set of data.
  • the classifier was completely parameterized by the distributions of biomarkers in the disease and benign training samples, and the list of biomarkers was chosen from Table 21; that is to say, the subset of markers chosen for inclusion determined a classifier in a one-to-one manner given a set of training data.
  • the greedy method employed here was used to search for the optimal subset of markers from Table 21. For small numbers of markers or classifiers with relatively few markers, every possible subset of markers was enumerated and evaluated in terms of the performance of the classifier constructed with that particular set of markers (see Example 4, Part 2). (This approach is well known in the field of statistics as “best subset selection”; see, e.g., The Elements of Statistical Learning—Data Mining, Inference, and Prediction, T. Hastie, et al., editors, Springer Science+Business Media, LLC, 2nd edition, 2009).
  • the number of combinations of multiple markers can be very large, and it was not feasible to evaluate every possible set of five markers, for example, from the list of 86 markers (Table 21) (i.e., 34,826,302 combinations). Because of the impracticality of searching through every subset of markers, the single optimal subset may not be found; however, by using this approach, many excellent subsets were found, and, in many cases, any of these subsets may represent an optimal one.
  • a “greedy” forward stepwise approach may be followed (see, e.g., Dabney A R, Storey J D (2007) Optimality Driven Nearest Centroid Classification from Genomic Data. PLoS ONE 2(10): e1002. doi:10.1371/journal.pone.0001002).
  • a classifier is started with the best single marker (based on KS-distance for the individual markers) and is grown at each step by trying, in turn, each member of a marker list that is not currently a member of the set of markers in the classifier. The one marker which scores best in combination with the existing classifier is added to the classifier. This is repeated until no further improvement in performance is achieved.
  • this approach may miss valuable combinations of markers for which some of the individual markers are not all chosen before the process stops.
  • the greedy procedure used here was an elaboration of the preceding forward stepwise approach, in that, to broaden the search, rather than keeping just a single candidate classifier (marker subset) at each step, a list of candidate classifiers was kept.
  • the list was seeded with every single marker subset (using every marker in the table on its own).
  • the list was expanded in steps by deriving new classifiers (marker subsets) from the ones currently on the list and adding them to the list.
  • Each marker subset currently on the list was extended by adding any marker from Table 1 not already part of that classifier, and which would not, on its addition to the subset, duplicate an existing subset (these are termed “permissible markers”). Every existing marker subset was extended by every permissible marker from the list.
  • biomarkers selected in Table 1 gave rise to classifiers which perform better than classifiers built with “non-markers” (i.e., proteins having signals that did not meet the criteria for inclusion in Table 1 (as described in Example 2)).
  • a performance of 1.0 is the baseline expectation for a random (coin toss) classifier.
  • the histogram of classifier performance was compared with the histogram of performance from a similar exhaustive enumeration of classifiers built from a “non-marker” table of 40 non-marker signals; the 40 signals were randomly chosen from 400 aptamers that did not demonstrate differential signaling between control and disease populations (KS-distance ⁇ 1.4).
  • FIG. 17 shows histograms of the performance of all possible one, two, and three-marker classifiers built from the biomarker parameters in Table 13 for biomarkers that can discriminate between benign nodules and NSCLC and compares these classifiers with all possible one, two, and three-marker classifiers built using the 40 “non-marker” aptamer RFU signals.
  • FIG. 17A shows the histograms of single marker classifier performance
  • FIG. 17B shows the histogram of two marker classifier performance
  • FIG. 17C shows the histogram of three marker classifier performance.
  • the solid lines represent the histograms of the classifier performance of all one, two, and three-marker classifiers using the biomarker data for benign nodules and NSCLC in Table 13.
  • the dotted lines are the histograms of the classifier performance of all one, two, and three-marker classifiers using the data for benign nodules and NSCLC but using the set of random non-marker signals.
  • FIG. 18 shows histograms of the performance of all possible one, two, and three-marker classifiers built from the biomarker parameters in Table 12 for biomarkers that can discriminate between asymptomatic smokers and NSCLC and compares these with all possible one, two, and three-marker classifiers built using 40 “non-marker” aptamer RFU signals.
  • FIG. 18A shows the histograms of single marker classifier performance
  • FIG. 18B shows the histogram of two marker classifier performance
  • FIG. 18C shows the histogram of three marker classifier performance.
  • the solid lines represent the histograms of the classifier performance of all one, two, and three-marker classifiers using the biomarker parameters for asymptomatic smokers and NSCLC in Table 12.
  • the dotted lines are the histograms of the classifier performance of all one, two, and three-marker classifiers using the data for asymptomatic smokers and NSCLC but using the set of random non-marker signals.
  • the classifiers built from the markers listed in Table 1 form a distinct histogram, well separated from the classifiers built with signals from the “non-markers” for all one-marker, two-marker, and three-marker comparisons.
  • the performance and AUC score of the classifiers built from the biomarkers in Table 1 also increase faster with the number of markers than do the classifiers built from the non-markers, the separation increases between the marker and non-marker classifiers as the number of markers per classifier increases. All classifiers built using the biomarkers listed in Tables 38 and 39 perform distinctly better than classifiers built using the “non-markers”.
  • FIG. 19 compares the performance of classifiers built with the full list of biomarkers in Tables 12 and 13 with the performance of classifiers built with a set of biomarkers from Tables 38 and 39 excluding top ranked markers.
  • FIG. 19 demonstrates that classifiers constructed without the best markers perform well, implying that the performance of the classifiers was not due to some small core group of markers and that the changes in the underlying processes associated with disease are reflected in the activities of many proteins. Many subsets of the biomarkers in Table 1 performed close to optimally, even after removing the top 15 of the 40 markers from Table 1.
  • FIG. 19A shows the effect on classifiers for discriminating benign nodules from NSCLC built with 2 to 10 markers. Even after dropping the 15 top-ranked markers (ranked by KS-distance) from Table 13, the benign nodule vs. NSCLC performance increased with the number of markers selected from the table to reach over 1.65 (Sensitivity+Specificity).
  • FIG. 19B shows the effect on classifiers for discriminating asymptomatic smokers from NSCLC built with 2 to 10 markers. Even after dropping the 15 top-ranked markers (ranked by KS-distance) from Table 12, the asymptomatic smokers vs. NSCLC performance increased with the number of markers selected from the table to reach over 1.7 (Sensitivity+Specificity), and closely approached the performance of the best classifier selected from the full list of biomarkers in Table 12.
  • FIG. 20 shows how the ROC performance of typical classifiers constructed from the list of parameters in Tables 12 and 13 according to Example 3.
  • FIG. 20A shows the model performance from assuming the independence of markers as in Example 3
  • FIG. 20B shows the actual ROC curves using the assay data set used to generate the parameters in Tables 12 and 13. It can be seen that the performance for a given number of selected markers was qualitatively in agreement, and that quantitative agreement degraded as the number of markers increases. (This is consistent with the notion that the information contributed by any particular biomarker concerning the disease processes is redundant with the information contributed by other biomarkers provided in Tables 12 and 13).
  • FIG. 20 shows how the ROC performance of typical classifiers constructed from the list of parameters in Tables 12 and 13 according to Example 3.
  • FIG. 20A shows the model performance from assuming the independence of markers as in Example 3
  • FIG. 20B shows the actual ROC curves using the assay data set used to generate the parameters in Tables 12 and 13.
  • Tables 12 and 13 in combination with the methods described in Example 3 enable the construction and evaluation of a great many classifiers useful for the discrimination of NSCLC from benign nodules and the discrimination of asymptomatic smokers who have NSCLC from those who do not have NSCLC.
  • the final readout on the multiplex assay is based on the amount of aptamer recovered after the successive capture steps in the assay.
  • the multiplex assay is based on the premise that the amount of aptamer recovered at the end of the assay is proportional to the amount of protein in the original complex mixture (e.g., plasma).
  • the original complex mixture e.g., plasma
  • This assay can be used to visually demonstrate that a desired protein is in fact pulled out from plasma after equilibration with an aptamer as well as to demonstrate that aptamers bound to their intended protein targets can survive as a complex through the kinetic challenge steps in the assay.
  • recovery of protein at the end of this pull-down assay requires that the protein remain non-covalently bound to the aptamer for nearly two hours after equilibration.
  • non-specifically bound proteins dissociate during these steps and do not contribute significantly to the final signal. It should be noted that the pull-down procedure described in this example includes all of the key steps in the multiplex assay described above.
  • Plasma samples were prepared by diluting 50 ⁇ L EDTA-plasma to 100 ⁇ L in SB18 with 0.05% Tween-20 (SB18T) and 2 ⁇ M Z-Block.
  • the plasma solution was equilibrated with 10 pmoles of a PBDC-aptamer in a final volume of 150 ⁇ L for 2 hours at 37° C.
  • complexes and unbound aptamer were captured with 133 ⁇ L of a 7.5% Streptavidin-agarose bead slurry by incubating with shaking for 5 minutes at RT in a Durapore filter plate.
  • the samples bound to beads were washed with biotin and with buffer under vacuum as described in Example 1.
  • This step will capture proteins bound to aptamers as well as proteins that may have dissociated from aptamers since the initial equilibration.
  • the beads were washed as described in Example 1. Proteins were eluted from the MyOne Streptavidin beads by incubating with 50 mM DTT in SB17T for 25 minutes at 37° C. with shaking. The eluate was then transferred to MyOne beads coated with a sequence complimentary to the 3′ fixed region of the aptamer and incubated for 25 minutes at 37° C. with shaking. This step captures all of the remaining aptamer. The beads were washed 2 ⁇ with 100 ⁇ L SB17T for 1 minute and 1 ⁇ with 100 ⁇ L SB19T for 1 minute.
  • Aptamer was eluted from these final beads by incubating with 45 ⁇ L 20 mM NaOH for 2 minutes with shaking to disrupt the hybridized strands. 40 ⁇ L of this eluate was neutralized with 10 ⁇ L 80 mM HCl containing 0.05% Tween-20. Aliquots representing 5% of the eluate from the first set of beads (representing all plasma proteins bound to the aptamer) and 20% of the eluate from the final set of beads (representing all plasma proteins remaining bound at the end of our clinical assay) were run on a NuPAGE 4-12% Bis-Tris gel (Invitrogen) under reducing and denaturing conditions. Gels were imaged on an Alpha Innotech FluorChem Q scanner in the Cy5 channel to image the proteins.
  • Pull-down gels for aptamers were selected against LBP ( ⁇ 1 ⁇ 10 ⁇ 7 M in plasma, polypeptide MW ⁇ 60 kDa), C9 ( ⁇ 1 ⁇ 10 ⁇ 6 M in plasma, polypeptide MW ⁇ 60 kDa), and IgM ( ⁇ 9 ⁇ 10 ⁇ 6 M in plasma, MW ⁇ 70 kDa and 23 kDa), respectively. (See FIG. 16 ).
  • lane 1 is the eluate from the Streptavidin-agarose beads
  • lane 2 is the final eluate
  • lane 3 is a MW marker lane (major bands are at 110, 50, 30, 15, and 3.5 kDa from top to bottom). It is evident from these gels that there is a small amount non-specific binding of plasma proteins in the initial equilibration, but only the target remains after performing the capture steps of the assay. It is clear that the single aptamer reagent is sufficient to capture its intended analyte with no up-front depletion or fractionation of the plasma. The amount of remaining aptamer after these steps is then proportional to the amount of the analyte in the initial sample.
  • tissue samples from surgical resections obtained from eight NSCLC patients were analyzed, All NSCLC patients were smokers, ranging in age from 47 to 75 years old and covering NSCLC stages 1A through 3B (Table 17). All tissue samples were obtained by freezing the tissue within 5-10 minutes of excision during surgery and after placing the tissues in OCT medium (10.24% polyvinyl alcohol, 4.26% polyethylene glycol, and 85.5% non-reactive ingredients). Three samples were obtained from each resection: tumor tissue sample, adjacent healthy tissue (within 1 cm of the tumor) and distant uninvolved lung tissue.
  • Sample total protein was adjusted to 16 ⁇ g/mL in SB17T buffer (SB17 buffer containing 0.05% tween 20) for proteomic profiling.
  • Samples prepared in this manner were run on the multiplex aptamer assay which, as noted above, measures over 800 proteins as described previously (Ostroff et al., Nature Precedings, http://precedings.nature.com/documents/4537/version/1 (2010)).
  • Most proteins were unchanged between tumor, adjacent tissue and distal tissue. However, some proteins were clearly suppressed ( FIG. 24 ) while others were elevated substantially in tumor tissues ( FIG. 23 ) compared to adjacent and distal tissues.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Medical Informatics (AREA)
  • Biophysics (AREA)
  • Theoretical Computer Science (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Evolutionary Biology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Pathology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Data Mining & Analysis (AREA)
  • General Engineering & Computer Science (AREA)
  • Databases & Information Systems (AREA)
  • Bioethics (AREA)
  • Computer Vision & Pattern Recognition (AREA)
  • Artificial Intelligence (AREA)
US13/808,751 2010-07-09 2011-07-11 Lung Cancer Biomarkers and Uses Thereof Abandoned US20130116150A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/808,751 US20130116150A1 (en) 2010-07-09 2011-07-11 Lung Cancer Biomarkers and Uses Thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US36312210P 2010-07-09 2010-07-09
US201161444947P 2011-02-21 2011-02-21
US13/808,751 US20130116150A1 (en) 2010-07-09 2011-07-11 Lung Cancer Biomarkers and Uses Thereof
PCT/US2011/043595 WO2012006632A2 (fr) 2010-07-09 2011-07-11 Biomarqueurs du cancer du poumon et leurs utilisations

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/043595 A-371-Of-International WO2012006632A2 (fr) 2008-09-09 2011-07-11 Biomarqueurs du cancer du poumon et leurs utilisations

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/993,132 Continuation US11221340B2 (en) 2010-07-09 2018-05-30 Lung cancer biomarkers and uses thereof

Publications (1)

Publication Number Publication Date
US20130116150A1 true US20130116150A1 (en) 2013-05-09

Family

ID=45441874

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/808,751 Abandoned US20130116150A1 (en) 2010-07-09 2011-07-11 Lung Cancer Biomarkers and Uses Thereof
US13/279,990 Abandoned US20120101002A1 (en) 2008-09-09 2011-10-24 Lung Cancer Biomarkers and Uses Thereof
US15/993,132 Active 2032-06-01 US11221340B2 (en) 2010-07-09 2018-05-30 Lung cancer biomarkers and uses thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US13/279,990 Abandoned US20120101002A1 (en) 2008-09-09 2011-10-24 Lung Cancer Biomarkers and Uses Thereof
US15/993,132 Active 2032-06-01 US11221340B2 (en) 2010-07-09 2018-05-30 Lung cancer biomarkers and uses thereof

Country Status (12)

Country Link
US (3) US20130116150A1 (fr)
EP (1) EP2591357A4 (fr)
JP (1) JP5905003B2 (fr)
KR (1) KR101870123B1 (fr)
CN (2) CN102985819B (fr)
AU (1) AU2011274422B2 (fr)
BR (1) BR112012032537B8 (fr)
CA (1) CA2801110C (fr)
IL (1) IL223295A (fr)
MX (2) MX2012014268A (fr)
SG (2) SG186953A1 (fr)
WO (1) WO2012006632A2 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100070191A1 (en) * 2008-09-09 2010-03-18 Somalogic, Inc. Lung Cancer Biomarkers and Uses Thereof
US20130282390A1 (en) * 2012-04-20 2013-10-24 International Business Machines Corporation Combining knowledge and data driven insights for identifying risk factors in healthcare
RU2538625C1 (ru) * 2014-01-20 2015-01-10 Денис Николаевич Бахмутов Способ диагностики рака
KR20180050303A (ko) * 2015-09-09 2018-05-14 소마로직, 인크. 개인별 약물 치료 계획의 개발 방법 및 단백질체 프로파일을 기반으로 하는 표적 약물 개발 방법
US9984147B2 (en) 2008-08-08 2018-05-29 The Research Foundation For The State University Of New York System and method for probabilistic relational clustering
WO2020018005A1 (fr) * 2018-07-17 2020-01-23 Limited Liability Company "Gero" Dispositifs, procédés, compositions et systèmes pour le traitement du vieillissement et de troubles liés à l'âge
WO2020092211A1 (fr) * 2018-10-30 2020-05-07 Somalogic, Inc. Procédés d'évaluation de qualité d'échantillon
WO2020243574A1 (fr) * 2019-05-31 2020-12-03 Delphinus Medical Technologies, Inc. Systèmes et procédés de caractérisation de lésion interactive

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2651185C (fr) * 2006-05-09 2022-08-30 The University Of British Columbia Proteine dissoutes marqueurs de l'arthrite
AU2008329529B2 (en) 2007-11-27 2015-02-19 The University Of British Columbia 14-3-3 eta antibodies and uses thereof for the diagnosis and treatment of arthritis
US20100221752A2 (en) * 2008-10-06 2010-09-02 Somalogic, Inc. Ovarian Cancer Biomarkers and Uses Thereof
US9495515B1 (en) 2009-12-09 2016-11-15 Veracyte, Inc. Algorithms for disease diagnostics
CN102414563B (zh) 2009-03-11 2014-12-03 奥古雷克斯生命科学公司 用于表征关节炎疾患的组合物和方法
CA2801110C (fr) 2010-07-09 2021-10-05 Somalogic, Inc. Biomarqueurs du cancer du poumon et leurs utilisations
AU2011289284B2 (en) 2010-08-13 2015-04-09 Somalogic Operating Co., Inc. Pancreatic cancer biomarkers and uses thereof
US20140235487A1 (en) * 2010-11-12 2014-08-21 William Marsh Rice University Oral cancer risk scoring
DK2768851T3 (en) 2011-10-21 2017-09-18 Augurex Life Sciences Corp ANTIGENS DERIVED BY CITRULLINATED 14-3-3 AND APPLICATIONS THEREOF IN THE DIAGNOSIS OF RHEUMATOID ARTHRITIS
EP2841603A4 (fr) * 2012-04-26 2016-05-25 Allegro Diagnostics Corp Procédés d'évaluation du statut de cancer du poumon
JP6022285B2 (ja) * 2012-09-28 2016-11-09 シスメックス株式会社 標本収納装置、標本収納方法、及び標本検査システム
BR112015009138A2 (pt) 2012-10-23 2020-10-20 Caris Life Sciences Switzerland Holdings, S.A.R.L. métodos para caracterizar um câncer
US10942184B2 (en) 2012-10-23 2021-03-09 Caris Science, Inc. Aptamers and uses thereof
US9103837B2 (en) 2012-11-07 2015-08-11 Somalogic, Inc. Chronic obstructive pulmonary disease (COPD) biomarkers and uses thereof
AU2013361323B2 (en) 2012-12-19 2018-09-06 Caris Science, Inc. Compositions and methods for aptamer screening
WO2014186036A1 (fr) 2013-03-14 2014-11-20 Allegro Diagnostics Corp. Procédés d'évaluation de l'état d'une maladie pulmonaire obstructive chronique (copd)
US11976329B2 (en) 2013-03-15 2024-05-07 Veracyte, Inc. Methods and systems for detecting usual interstitial pneumonia
EP3296747B1 (fr) 2013-03-15 2020-01-08 Somalogic, Inc. Biomarqueurs de la maladie du foie gras non alcoolique (nafld) et de la stéatohépatite non alcoolique (nash) et leurs utilisations
WO2014153327A1 (fr) 2013-03-18 2014-09-25 Barbeau James M Procédés de détermination de la carcinogenèse, d'identification de marqueurs pour le diagnostic précoce du cancer et d'identification de cibles d'une thérapie
EP2999797B1 (fr) * 2013-05-21 2018-07-04 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Isoformes spécifiques de gata6 et nkx2-1 en tant que nouveaux marqueurs pour les approches de diagnostic et de thérapie du cancer et en tant que cibles pour la thérapie anticancéreuse
KR101552014B1 (ko) 2013-10-11 2015-09-09 인제대학교 산학협력단 폐암 진단을 위한 nir의 신규한 용도
WO2015066564A1 (fr) * 2013-10-31 2015-05-07 Cancer Prevention And Cure, Ltd. Méthodes d'identification et de diagnostic de maladies pulmonaires à l'aide de systèmes de classification et trousses associées
BR112016010322A2 (pt) * 2013-11-07 2017-08-08 Medial Res Ltd Método computadorizado de avaliação de risco de câncer de pulmão, meio legível por computador, sistema de avaliação de câncer de pulmão, e método de geração de um classificador para avaliação de risco de câncer de pulmão
CN111540410B (zh) * 2013-12-16 2024-04-02 菲利普莫里斯生产公司 用于预测个体的吸烟状况的系统和方法
WO2015164616A1 (fr) 2014-04-24 2015-10-29 Somalogic, Inc. Biomarqueurs de détection de la tuberculose
WO2015164617A1 (fr) 2014-04-24 2015-10-29 Somalogic, Inc. Biomarqueurs de la tuberculose dans l'urine et leurs utilisations
WO2015164772A1 (fr) * 2014-04-25 2015-10-29 Rush University Medical Center Protéines associées au facteur de croissance analogue à l'insuline (igf) en circulation pour la détection du cancer du poumon
EP3169814B1 (fr) * 2014-07-14 2022-05-04 Veracyte, Inc. Procédés pour évaluer le stade d'un cancer du poumon
BR112017005730B1 (pt) 2014-09-26 2023-12-12 Somalogic Operating Co., Inc Método para triagem de um indivíduo quanto ao risco de um evento cardiovascular ou para predizer a probabilidade que um indivíduo tenha tal evento
EP3770274A1 (fr) 2014-11-05 2021-01-27 Veracyte, Inc. Systèmes et procédés de diagnostic de la fibrose pulmonaire idiopathique sur des biopsies transbronchiques à l'aide de l'apprentissage automatique et de données de transcription dimensionnelle élevée
WO2016123058A1 (fr) 2015-01-27 2016-08-04 Somalogic, Inc. Biomarqueurs pour la détection du risque de tuberculose
EP3295176A1 (fr) 2015-05-08 2018-03-21 Somalogic, Inc. Biomarqueurs pour la détection du risque de tuberculose
KR102078310B1 (ko) * 2015-09-11 2020-02-19 주식회사 압타머사이언스 비소세포성 폐암 진단용 단백질 바이오마커 패널 및 이를 이용한 비소세포성 폐암 진단 방법
US10546650B2 (en) 2015-10-23 2020-01-28 Google Llc Neural network for processing aptamer data
EP3414575A1 (fr) 2016-02-08 2018-12-19 Somalogic, Inc. Biomarqueurs de stéatose hépatique non-alcoolique (nafld) et de stéatohépatite non-alcoolique (shna) et utilisations associées
JP6889428B2 (ja) * 2016-02-26 2021-06-18 国立大学法人山口大学 情報処理装置と情報処理プログラム並びに情報処理方法
JP6041331B1 (ja) * 2016-02-26 2016-12-07 国立大学法人山口大学 情報処理装置と情報処理プログラム並びに情報処理方法
CN107435062B (zh) * 2016-05-25 2020-10-20 上海伯豪医学检验所有限公司 甄别肺部微小结节良恶性的外周血基因标志物及其用途
KR102531819B1 (ko) * 2016-06-08 2023-05-16 리서치 디벨럽먼트 파운데이션 혈관내 광 간섭 단층촬영법을 사용한 자동 관상동맥 플라크 특징화 및 위험 평가를 위한 시스템들 및 방법들
KR20190026769A (ko) 2016-06-21 2019-03-13 더 위스타 인스티튜트 오브 아나토미 앤드 바이올로지 유전자 발현 프로파일을 사용하여 폐암을 진단하기 위한 조성물 및 방법
JP2020522690A (ja) * 2017-06-02 2020-07-30 ベラサイト インコーポレイテッド 肺疾病の特定又はモニタリング方法及びシステム
CN109470855B (zh) * 2017-09-08 2022-02-11 广州市丹蓝生物科技有限公司 肺癌早期诊断用蛋白芯片及试剂盒
KR102028967B1 (ko) 2017-12-28 2019-10-07 한국원자력의학원 Rip1을 포함하는 폐암 전이 진단용 바이오 마커 및 이의 이용
WO2019202448A1 (fr) 2018-04-16 2019-10-24 University Of Cape Town Biomarqueur de protéomique à trois protéines pour la détermination prospective du risque de développement de tuberculose active
SG11202012472TA (en) * 2018-06-22 2021-01-28 Somalogic Inc Improved proteomic multiplex assays
KR102216645B1 (ko) * 2018-10-29 2021-02-17 사회복지법인 삼성생명공익재단 폐암의 분자 아형 결정을 위한 바이오마커 패널 및 이의 용도
CN109470859A (zh) * 2018-11-04 2019-03-15 华东医院 一种外泌体蛋白作为鉴别肺结节良恶性标志物及其应用
EP3671210A1 (fr) * 2018-12-21 2020-06-24 Biosystems International KFT Biomarqueurs épitomiques de protéines du cancer du poumon
US20210027890A1 (en) * 2019-07-24 2021-01-28 ConnSante BioTech, Inc. Detecting, evaluating and predicting system for cancer risk
EP4025916A1 (fr) 2019-09-03 2022-07-13 SomaLogic Operating Co., Inc. Prédiction d'évènement de risque cardio-vasculaire et utilisations associées
US20230048910A1 (en) 2020-01-10 2023-02-16 Somalogic Operating Co., Inc. Methods of Determining Impaired Glucose Tolerance
GB2603051B (en) 2020-01-30 2023-04-26 Prognomiq Inc Lung biomarkers and methods of use thereof
AU2021220787A1 (en) 2020-02-10 2022-09-01 Somalogic Operating Co., Inc. Nonalcoholic steatohepatitis (NASH) biomarkers and uses thereof
KR102260915B1 (ko) 2020-02-19 2021-06-04 한국원자력의학원 폐암의 방사선 치료 예후 예측을 위한 생체 표지자
CN111876478A (zh) * 2020-04-28 2020-11-03 中国科学院微生物研究所 肺结节诊断标志物及应用
WO2022035747A1 (fr) * 2020-08-14 2022-02-17 The Medical College Of Wisconsin, Inc. Signature d'expression génique pour prédire une réponse d'immunothérapie et procédés d'utilisation
JP2023545017A (ja) * 2020-10-02 2023-10-26 ボード オブ レジェンツ,ザ ユニバーシティ オブ テキサス システム 肺がんの検出及び治療のための方法
CN116381073A (zh) * 2020-10-10 2023-07-04 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) 生物标志物在制备肺癌检测试剂中的用途和方法
US20230393146A1 (en) 2020-10-20 2023-12-07 Somalogic Operating Co., Inc. Cardiovascular Event Risk Prediction
GB2607436A (en) * 2021-03-31 2022-12-07 Prognomiq Inc Multi-omic assessment
WO2023141248A1 (fr) 2022-01-21 2023-07-27 Somalogic Operating Co., Inc. Procédés d'évaluation de qualité d'échantillons
WO2023211770A1 (fr) 2022-04-24 2023-11-02 Somalogic Operating Co., Inc. Procédés d'évaluation de la qualité d'un échantillon
WO2023211771A1 (fr) 2022-04-24 2023-11-02 Somalogic Operating Co., Inc. Procédés d'évaluation de la qualité d'un échantillon
WO2023211769A1 (fr) 2022-04-24 2023-11-02 Somalogic Operating Co., Inc. Procédés pour évaluater la qualité d'un échantillon
WO2023211773A1 (fr) 2022-04-24 2023-11-02 Somalogic Operating Co., Inc. Procédés d'évaluation de qualité d'échantillons
WO2023244582A1 (fr) * 2022-06-13 2023-12-21 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Biomarqueurs associés au cancer du poumon et leurs procédés d'utilisation
WO2024010854A1 (fr) * 2022-07-07 2024-01-11 Diagnose Early, Inc. Génération rapide de rapports de santé basés sur la respiration et systèmes destinés à être utilisés dans ceux-ci
WO2024015485A1 (fr) 2022-07-14 2024-01-18 Somalogic Operating Co., Inc. Procédés d'évaluation du risque de démence
WO2024015486A1 (fr) 2022-07-14 2024-01-18 Somalogic Operating Co., Inc. Procédés d'évaluation de la qualité d'un échantillon
WO2024064322A2 (fr) 2022-09-23 2024-03-28 Somalogic Operating Co., Inc. Procédés d'évaluation de l'état d'utilisation de tabac

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120178111A1 (en) * 2009-09-23 2012-07-12 Diamandis Eleftherios P Methods and compositions for the detection of lung cancers

Family Cites Families (117)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5660985A (en) 1990-06-11 1997-08-26 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands containing modified nucleotides
US20040254101A1 (en) 1995-06-06 2004-12-16 Human Genome Sciences, Inc. Colon specific gene and protein and cancer
WO1998039721A1 (fr) 1997-03-07 1998-09-11 University Of Florida Methode de diagnostic et de stadification du cancer de la prostate
AU3316600A (en) 1999-02-22 2000-09-21 Torben F. Orntoft Gene expression in bladder tumors
ATE400813T1 (de) 1999-09-10 2008-07-15 Takashi Muramatsu Tumormarker für frühes krebsstadium
US20030144476A1 (en) 2000-03-06 2003-07-31 Pankaj Agarwal Novel compounds
US20030224501A1 (en) 2000-03-17 2003-12-04 Young Paul E. Bone morphogenic protein polynucleotides, polypeptides, and antibodies
US6596503B1 (en) * 2000-08-18 2003-07-22 East Carolina University Monoclonal antibody DS6, tumor-associated antigen CA6, and methods of use thereof
US6631330B1 (en) 2000-08-21 2003-10-07 Assistance Publique-Hopitaux De Paris (Ap-Hp) Diagnosis method of inflammatory, fibrotic or cancerous disease using biochemical markers
US6576423B2 (en) 2000-12-08 2003-06-10 The Board Of Regents Of The University Of Nebraska Specific mucin expression as a marker for pancreatic cancer
JP2005527180A (ja) * 2001-04-18 2005-09-15 プロテイン デザイン ラブス, インコーポレイテッド 肺がんの診断方法、肺がんの修飾因子の組成及びスクリーニングの方法
US7189507B2 (en) 2001-06-18 2007-03-13 Pdl Biopharma, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
US7622260B2 (en) 2001-09-05 2009-11-24 The Brigham And Women's Hospital, Inc. Diagnostic and prognostic tests
EP1444361A4 (fr) 2001-09-28 2006-12-27 Whitehead Biomedical Inst Classification de carcinomes pulmonaires par analyse de l'expression genique
WO2003058201A2 (fr) * 2001-12-31 2003-07-17 Quark Biotech, Inc. Procedes permettant d'identifier des genes marqueurs pour le cancer
WO2003078662A1 (fr) 2002-03-13 2003-09-25 Genomic Health Profilage d'expression genique dans des tissus tumoraux ponctionnes
DE60325628D1 (de) 2002-09-30 2009-02-12 Oncotherapy Science Inc Gene und polypeptide in verbindung mit menschlichen pankreaskarzinomen
US20050260639A1 (en) 2002-09-30 2005-11-24 Oncotherapy Science, Inc. Method for diagnosing pancreatic cancer
JP4606879B2 (ja) 2002-11-15 2011-01-05 ジェノミック ヘルス, インコーポレイテッド Egfr陽性癌の遺伝子発現プロファイリング
AU2003294828A1 (en) 2002-12-17 2004-07-09 Sinogenomax Co. Ltd. Chinese National Human Genomecenter Specific markers for pancreatic cancer
US8014952B2 (en) 2002-12-18 2011-09-06 Queen Elizabeth Hospital Serum biomarkers in lung cancer
US20050181375A1 (en) 2003-01-10 2005-08-18 Natasha Aziz Novel methods of diagnosis of metastatic cancer, compositions and methods of screening for modulators of metastatic cancer
US20040231909A1 (en) 2003-01-15 2004-11-25 Tai-Yang Luh Motorized vehicle having forward and backward differential structure
AU2003900747A0 (en) 2003-02-18 2003-03-06 Garvan Institute Of Medical Research Diagnosis and treatment of pancreatic cancer
JP4568716B2 (ja) 2003-02-20 2010-10-27 ジェノミック ヘルス, インコーポレイテッド 遺伝子発現を測定するためのイントロンrnaの使用
CA2516591A1 (fr) 2003-02-26 2004-09-10 Mount Sinai Hospital Dosage a marqueurs multiples utilise pour depister un cancer des ovaires
WO2004099432A2 (fr) 2003-05-02 2004-11-18 The Johns Hopkins University Identification de biomarqueurs permettant de detecter la presence d'un cancer du pancreas
CN1455257A (zh) * 2003-05-23 2003-11-12 北京师范大学 一种利用表面修饰蛋白芯片进行肺癌诊断方法
KR20060031809A (ko) 2003-06-09 2006-04-13 더 리젠츠 오브 더 유니버시티 오브 미시간 암 치료 및 진단용 조성물 및 방법
DK1641810T4 (en) 2003-06-24 2017-07-03 Genomic Health Inc Predicting the likelihood of cancer recurrence
ES2651849T3 (es) 2003-07-10 2018-01-30 Genomic Health, Inc. Algoritmo del perfil de expresión y test para el pronóstico del cáncer
EP1668360A4 (fr) 2003-08-15 2007-08-08 Univ Pittsburgh Analyse multifactorielle pour la detection du cancer
US7807392B1 (en) 2003-09-15 2010-10-05 Celera Corporation Lung disease targets and uses thereof
EP1682904A2 (fr) 2003-11-04 2006-07-26 Roche Diagnostics GmbH Methode pour distinguer des sous-types aml classes who
EP1723428A2 (fr) 2004-02-19 2006-11-22 Yale University Corporation Identification de biomarqueurs proteiques du cancer par des techniques proteomiques
JP2007527247A (ja) 2004-03-05 2007-09-27 ロゼッタ インファーマティクス エルエルシー 臨床的基準および情報提供性遺伝子セットの組合せを使用する乳癌患者の分類
ES2550614T3 (es) 2004-04-09 2015-11-11 Genomic Health, Inc. Marcadores de expresión génica para predecir la respuesta a la quimioterapia
ES2367311T3 (es) 2004-04-15 2011-11-02 University Of Florida Research Foundation, Inc. Productos de degradación proteolítica de map-2 como biomarcadores de diagnóstico para las lesiones neurales.
EP1743031A4 (fr) 2004-04-26 2008-05-28 Childrens Medical Center Biomarqueurs de plaquettes utilises dans le diagnostic de maladies
US20070275422A1 (en) 2004-07-26 2007-11-29 The Board Of Trustees Of The Leland Stanford Jr. U Methods and Compositions for Detecting Pancreatic Disease
WO2006022895A2 (fr) 2004-08-02 2006-03-02 Children's Medical Center Corporation Biomarqueurs de plaquettes servant a detecter le cancer
WO2006016697A1 (fr) 2004-08-10 2006-02-16 Oncotherapy Science, Inc. Gène relatif au cancer du poumon non a petites cellules, anln, et ses interactions avec le rhoa
JP2006053113A (ja) * 2004-08-16 2006-02-23 Medical Proteoscope Co Ltd 肺腺癌リンパ節転移診断方法及び診断キット
DK1815014T3 (da) 2004-11-05 2012-06-18 Genomic Health Inc Molekylære indikatorer for brystcancerprognose og forudsigelse af behandlingsrespons
EP1836629B1 (fr) 2004-11-05 2020-03-04 Genomic Health, Inc. Prediction de reaction a la chimiotherapie au moyen de marqueurs d'expression genique
US7862995B2 (en) 2004-12-10 2011-01-04 Targeted Molecular Diagnostics Methods and materials for predicting responsiveness to treatment with dual tyrosine kinase inhibitor
US8632983B2 (en) 2005-04-15 2014-01-21 Van Andel Research Institute Biomarkers for pancreatic cancer and diagnostic methods
WO2006135886A2 (fr) 2005-06-13 2006-12-21 The Regents Of The University Of Michigan Compositions et methodes pour le traitement et le diagnostic du cancer
US20090005268A1 (en) 2005-07-18 2009-01-01 Epigenomics Ag Compositions and Methods for Cancer Diagnostics Comprising Pan-Cancer Markers
US7521195B1 (en) 2005-07-21 2009-04-21 Celera Corporation Lung disease targets and uses thereof
JP2009502115A (ja) 2005-07-27 2009-01-29 オンコセラピー・サイエンス株式会社 小細胞肺癌の診断方法
US20080305962A1 (en) 2005-07-29 2008-12-11 Ralph Markus Wirtz Methods and Kits for the Prediction of Therapeutic Success, Recurrence Free and Overall Survival in Cancer Therapies
US7582441B1 (en) 2005-10-17 2009-09-01 Celera Corporation Methods and compositions for treating and diagnosing disease
EP1777523A1 (fr) 2005-10-19 2007-04-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthode pour pronostiquer la progression d'un cancer et les résultats du patient et les moyens pour performer la méthode
US8329408B2 (en) 2005-10-31 2012-12-11 Bayer Healthcare Llc Methods for prognosis and monitoring cancer therapy
CA2632190A1 (fr) * 2005-11-10 2007-07-12 University Of Kentucky Dosage de diagnostic de cancer du poumon
US8445198B2 (en) 2005-12-01 2013-05-21 Medical Prognosis Institute Methods, kits and devices for identifying biomarkers of treatment response and use thereof to predict treatment efficacy
US8014957B2 (en) 2005-12-15 2011-09-06 Fred Hutchinson Cancer Research Center Genes associated with progression and response in chronic myeloid leukemia and uses thereof
CA2821426A1 (fr) 2005-12-22 2007-07-05 Abbott Laboratories Procede et combinaisons de marqueurs permettant de depister une predisposition au cancer du poumon
MX2008008973A (es) 2006-01-11 2008-11-26 Genomic Health Inc Marcadores de expresion de genes para pronostico de cancer colorectal.
KR20080094803A (ko) 2006-01-27 2008-10-24 트리패스 이미징, 인코포레이티드 난소암의 발병 가능성이 높은 환자를 확인하는 방법 및 그의 조성물
WO2007092433A2 (fr) 2006-02-06 2007-08-16 Tethys Bioscience, Inc. Marqueurs associés à l'ostéoporose et leurs méthodes d'utilisation
WO2007109571A2 (fr) 2006-03-17 2007-09-27 Prometheus Laboratories, Inc. Procédés de prédiction et de suivi de la thérapie par l'inhibiteur de la tyrosine kinase
US7888019B2 (en) 2006-03-31 2011-02-15 Genomic Health, Inc. Genes involved estrogen metabolism
CA3086149A1 (fr) 2006-04-04 2007-10-11 Singulex, Inc. Systeme et procedes hautement sensibles destines a une analyse de la troponine
EP2030025A2 (fr) 2006-06-07 2009-03-04 Tethys Bioscience, Inc. Marqueurs associés à des événements artério-vasculaires et procédés d'utilisation de ces marqueurs
US20080090258A1 (en) 2006-06-28 2008-04-17 University Of Pittsburgh -Of The Commonwealth System Of Higher Education Method and composition for diagnosing endometrial cancer
EP2090665A2 (fr) 2006-10-20 2009-08-19 Exiqon A/S Nouveaux micro ARN humains associés au cancer
US20100184034A1 (en) 2006-11-13 2010-07-22 SOURCE PRECISION MEDICINE, INC d/b/a SOURCE MDX Gene Expression Profiling for Identification, Monitoring and Treatment of Lung Cancer
US7947447B2 (en) * 2007-01-16 2011-05-24 Somalogic, Inc. Method for generating aptamers with improved off-rates
US7964356B2 (en) 2007-01-16 2011-06-21 Somalogic, Inc. Method for generating aptamers with improved off-rates
KR100846354B1 (ko) * 2007-03-21 2008-07-15 (주) 프로탄바이오 혈청 당단백질부터 분리한 폐암 진단용 마커
PL2140269T3 (pl) 2007-03-27 2014-07-31 Immunovia Ab Podpis białkowy/znaczniki do wykrywania gruczolakoraka
RU2376372C2 (ru) 2007-04-03 2009-12-20 Государственное учреждение Научно-исследовательский институт медицинской генетики Томского научного центра Сибирского отделения Российской академии медицинских наук Способ генетической диагностики подверженности к сердечно-сосудистым заболеваниям
CA2681667A1 (fr) 2007-05-18 2008-11-27 Duke University Biomarqueurs seriques de detection precoce du cancer du poumon
WO2008153814A2 (fr) 2007-05-29 2008-12-18 President And Fellows Of Harvard College Molécules impliquées dans la régulation de l'activité des ostéoblastes et des ostéoclastes, et leurs méthodes d'utilisation
ES2647587T3 (es) * 2007-07-17 2017-12-22 Somalogic, Inc. Aptámeros con uridinas y/o timidinas sustituidas en la posición 5 con un grupo bencilo
EP2019318A1 (fr) 2007-07-27 2009-01-28 Erasmus University Medical Center Rotterdam Marqueurs de protéines pour événements cardio-vasculaires
GB0717637D0 (en) 2007-09-10 2007-10-17 Univ Leiden Future cardiac event biomarkers
US8541183B2 (en) * 2007-09-11 2013-09-24 Cancer Prevention And Cure, Ltd. Methods of identification, assessment, prevention and therapy of lung diseases and kits thereof
US7888051B2 (en) * 2007-09-11 2011-02-15 Cancer Prevention And Cure, Ltd. Method of identifying biomarkers in human serum indicative of pathologies of human lung tissues
US20100267041A1 (en) 2007-09-14 2010-10-21 Predictive Biosciences, Inc. Serial analysis of biomarkers for disease diagnosis
EP2201363A4 (fr) * 2007-09-18 2011-03-16 Univ Texas Détection dans la salive, de protéines modulées produites suite à un carcinome canalaire in situ du sein
CA2705486C (fr) 2007-11-19 2019-04-02 Celera Corporation Marqueurs de cancer du poumon et leurs utilisations
JP2011510297A (ja) 2008-01-18 2011-03-31 バトリックス・メディカル・インコーポレイテッド 動脈瘤のための診断バイオマーカー
GB0803192D0 (en) 2008-02-22 2008-04-02 Mubio Products Bv SCLC biomarker panel
US8067178B2 (en) 2008-03-14 2011-11-29 Genomic Health, Inc. Gene expression markers for prediction of patient response to chemotherapy
US10359425B2 (en) * 2008-09-09 2019-07-23 Somalogic, Inc. Lung cancer biomarkers and uses thereof
ES2559758T3 (es) * 2008-09-09 2016-02-15 Somalogic, Inc. Biomarcadores de cáncer de pulmón y usos de los mismos
US20120143805A1 (en) 2008-09-09 2012-06-07 Somalogic, Inc. Cancer Biomarkers and Uses Thereof
US20100221752A2 (en) 2008-10-06 2010-09-02 Somalogic, Inc. Ovarian Cancer Biomarkers and Uses Thereof
US20120165217A1 (en) 2008-10-06 2012-06-28 Somalogic, Inc. Cancer Biomarkers and Uses Thereof
CA2741117A1 (fr) 2008-10-30 2010-05-06 Centre De Recherche Public De La Sante Biomarqueurs d'insuffisance cardiaque
GB2512153B (en) 2008-11-17 2014-11-12 Veracyte Inc Methods and compositions of molecular profiling for disease diagnostics
WO2010059742A1 (fr) 2008-11-18 2010-05-27 Collabrx, Inc. Traitement du cancer individualisé
EP2370813A4 (fr) 2008-12-04 2012-05-23 Univ California Matériels et méthodes de diagnostic et de pronostic d'un cancer de la prostate
WO2010144358A1 (fr) 2009-06-08 2010-12-16 Singulex, Inc. Groupes de biomarqueurs très sensibles
PT2264183T (pt) 2009-06-09 2017-03-14 Gendiag Exe Sl Marcadores de risco para doença cardiovascular
CN102803951A (zh) 2009-06-15 2012-11-28 心脏Dx公司 冠状动脉疾病风险的测定
CA2771614A1 (fr) 2009-08-19 2011-02-24 Stanley Hazen Detection par marqueur pour caracteriser le risque de maladie cardiovasculaire ou de complications associees
EP2491396A4 (fr) 2009-10-22 2013-04-17 Univ California Estimation de la masse tumorale solide
WO2011059721A1 (fr) 2009-10-29 2011-05-19 Tethys Bioscience, Inc. Biomarqueurs de protéines et de lipides améliorant considérablement la prédiction du diabète de type 2
US20120245235A1 (en) 2009-12-01 2012-09-27 Compenda Bioscience, Inc Classification of cancers
CA2783536A1 (fr) 2009-12-09 2011-06-16 Aviir, Inc. Dosage de biomarqueurs pour le diagnostic et le classement des maladies cardiovasculaires
WO2011094483A2 (fr) 2010-01-29 2011-08-04 H. Lee Moffitt Cancer Center And Research Institute, Inc. Signatures géniques immunitaires dans le cancer
WO2011100472A1 (fr) 2010-02-10 2011-08-18 The Regents Of The University Of California Biomarqueurs transcriptomiques et protéomiques salivaires pour la détection du cancer du sein
CA2791905A1 (fr) 2010-03-01 2011-09-09 Caris Life Sciences Luxembourg Holdings, S.A.R.L. Biomarqueurs pour theranostique
JP2013526852A (ja) 2010-04-06 2013-06-27 カリス ライフ サイエンシズ ルクセンブルク ホールディングス 疾患に対する循環バイオマーカー
US20110256545A1 (en) * 2010-04-14 2011-10-20 Nancy Lan Guo mRNA expression-based prognostic gene signature for non-small cell lung cancer
CA2801110C (fr) 2010-07-09 2021-10-05 Somalogic, Inc. Biomarqueurs du cancer du poumon et leurs utilisations
AU2011289284B2 (en) 2010-08-13 2015-04-09 Somalogic Operating Co., Inc. Pancreatic cancer biomarkers and uses thereof
CA2809282C (fr) 2010-09-27 2017-09-12 Somalogic, Inc. Biomarqueurs de mesotheliome et utilisations de ceux-ci
CN110444287B (zh) * 2011-04-29 2024-02-06 癌症预防和治疗有限公司 使用分类系统及其试剂盒识别和诊断肺部疾病的方法
ES2777002T3 (es) 2011-09-30 2020-08-03 Somalogic Inc Predicción de eventos de riesgo cardiovascular y usos de la misma
ES2674318T3 (es) 2011-10-24 2018-06-28 Somalogic, Inc. Biomarcadores de cáncer de pulmón y usos de los mismos
US9103837B2 (en) 2012-11-07 2015-08-11 Somalogic, Inc. Chronic obstructive pulmonary disease (COPD) biomarkers and uses thereof
AU2014353102B2 (en) 2013-11-21 2019-05-16 Somalogic Operating Co., Inc. Cytidine-5-carboxamide modified nucleotide compositions and methods related thereto

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120178111A1 (en) * 2009-09-23 2012-07-12 Diamandis Eleftherios P Methods and compositions for the detection of lung cancers

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
Chen et al., Chem Med Chem, 2008, 3, pages 991-1001 *
Kettunen et al., Cancer Genetics and Cytogenetics, 2004, 149, pages 98-106 *
Okada et al., Biochemical and Biophysical Research Communications, 2008, Vol. 376, pages 370-376 *
Ostroff et al., Journal of Proteomics, published 9/2009, pp. 649-666 *
Patz et al. (Journal Clinical Oncology, 2007, 25, pages 5578-5583). *
Seder et al., Neoplasia, 2009, 11(4), pp. 388-396 *
Stearman et al., Cancer Research, 2008, 68(1), pp. 34-43 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9984147B2 (en) 2008-08-08 2018-05-29 The Research Foundation For The State University Of New York System and method for probabilistic relational clustering
US20100070191A1 (en) * 2008-09-09 2010-03-18 Somalogic, Inc. Lung Cancer Biomarkers and Uses Thereof
US10359425B2 (en) 2008-09-09 2019-07-23 Somalogic, Inc. Lung cancer biomarkers and uses thereof
US20130282390A1 (en) * 2012-04-20 2013-10-24 International Business Machines Corporation Combining knowledge and data driven insights for identifying risk factors in healthcare
US20130282393A1 (en) * 2012-04-20 2013-10-24 International Business Machines Corporation Combining knowledge and data driven insights for identifying risk factors in healthcare
RU2538625C1 (ru) * 2014-01-20 2015-01-10 Денис Николаевич Бахмутов Способ диагностики рака
KR20180050303A (ko) * 2015-09-09 2018-05-14 소마로직, 인크. 개인별 약물 치료 계획의 개발 방법 및 단백질체 프로파일을 기반으로 하는 표적 약물 개발 방법
KR102604025B1 (ko) * 2015-09-09 2023-11-21 소마로직 오퍼레이팅 컴퍼니, 인코포레이티드 개인별 약물 치료 계획의 개발 방법 및 단백질체 프로파일을 기반으로 하는 표적 약물 개발 방법
WO2020018005A1 (fr) * 2018-07-17 2020-01-23 Limited Liability Company "Gero" Dispositifs, procédés, compositions et systèmes pour le traitement du vieillissement et de troubles liés à l'âge
WO2020092211A1 (fr) * 2018-10-30 2020-05-07 Somalogic, Inc. Procédés d'évaluation de qualité d'échantillon
CN113167782A (zh) * 2018-10-30 2021-07-23 私募蛋白质体公司 用于样品质量评估的方法
WO2020243574A1 (fr) * 2019-05-31 2020-12-03 Delphinus Medical Technologies, Inc. Systèmes et procédés de caractérisation de lésion interactive

Also Published As

Publication number Publication date
MX2012014268A (es) 2013-02-12
JP5905003B2 (ja) 2016-04-20
MX355020B (es) 2018-04-02
US11221340B2 (en) 2022-01-11
CN104777313A (zh) 2015-07-15
CA2801110C (fr) 2021-10-05
CA2801110A1 (fr) 2012-01-12
US20180275143A1 (en) 2018-09-27
KR101870123B1 (ko) 2018-06-25
US20120101002A1 (en) 2012-04-26
CN102985819A (zh) 2013-03-20
SG186953A1 (en) 2013-02-28
BR112012032537B1 (pt) 2021-03-02
CN102985819B (zh) 2015-04-15
WO2012006632A2 (fr) 2012-01-12
IL223295A (en) 2016-11-30
KR20130129347A (ko) 2013-11-28
WO2012006632A3 (fr) 2012-05-18
BR112012032537A2 (pt) 2017-05-23
EP2591357A4 (fr) 2014-01-01
AU2011274422A1 (en) 2012-12-20
JP2013532295A (ja) 2013-08-15
EP2591357A2 (fr) 2013-05-15
BR112012032537B8 (pt) 2022-10-18
CN104777313B (zh) 2017-09-26
IL223295A0 (en) 2013-02-03
SG10201508656VA (en) 2015-11-27
AU2011274422B2 (en) 2016-02-11

Similar Documents

Publication Publication Date Title
US11221340B2 (en) Lung cancer biomarkers and uses thereof
US10359425B2 (en) Lung cancer biomarkers and uses thereof
EP2340506B1 (fr) Biomarqueurs de cancer du poumon et utilisations associées
US11041866B2 (en) Pancreatic cancer biomarkers and uses thereof
EP2771692B1 (fr) Biomarqueurs de cancer du poumon et leurs utilisations
US20100221752A2 (en) Ovarian Cancer Biomarkers and Uses Thereof
US20120143805A1 (en) Cancer Biomarkers and Uses Thereof
US20120165217A1 (en) Cancer Biomarkers and Uses Thereof
CA2737004A1 (fr) Biomarqueurs du cancer des ovaires et leurs utilisations
US20220065872A1 (en) Lung Cancer Biomarkers and Uses Thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: SOMALOGIC, INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WILCOX, SHERI K.;AYERS, DEBORAH;JANJIC, NEBOJSA;AND OTHERS;SIGNING DATES FROM 20110818 TO 20110908;REEL/FRAME:029581/0465

AS Assignment

Owner name: VISIUM HEALTHCARE PARTNERS, LP, NEW YORK

Free format text: SECURITY INTEREST;ASSIGNOR:SOMALOGIC, INC.;REEL/FRAME:037853/0211

Effective date: 20160225

AS Assignment

Owner name: MADRYN HEALTH PARTNERS, LP, NEW YORK

Free format text: CHANGE OF NAME;ASSIGNOR:VISIUM HEALTHCARE PARTNERS, LP;REEL/FRAME:041920/0957

Effective date: 20170217

STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION

AS Assignment

Owner name: SOMALOGIC, INC., COLORADO

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:MADRYN HEALTH PARTNERS, LP, FORMERLY VISIUM HEALTHCARE PARTNERS, LP;REEL/FRAME:055981/0884

Effective date: 20210416