US20170335396A1 - Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data - Google Patents

Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data Download PDF

Info

Publication number
US20170335396A1
US20170335396A1 US15/523,654 US201515523654A US2017335396A1 US 20170335396 A1 US20170335396 A1 US 20170335396A1 US 201515523654 A US201515523654 A US 201515523654A US 2017335396 A1 US2017335396 A1 US 2017335396A1
Authority
US
United States
Prior art keywords
seq
uip
transcripts
classifier
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
US15/523,654
Other languages
English (en)
Inventor
Giulia C. Kennedy
James Diggans
Jing Huang
Yoonha CHOI
Su Yeon KIM
Daniel Pankratz
Moraima Pagan
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.)
Veracyte Inc
Original Assignee
Veracyte 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 Veracyte Inc filed Critical Veracyte Inc
Priority to US15/523,654 priority Critical patent/US20170335396A1/en
Assigned to VERACYTE, INC. reassignment VERACYTE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PANKRATZ, Daniel, KIM, SU YEON, CHOI, Yoonha, HUANG, JING, KENNEDY, GIULIA C., PAGAN, MORAIMA
Assigned to VERACYTE, INC. reassignment VERACYTE, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: MADRYN HEALTH PARTNERS, LP
Publication of US20170335396A1 publication Critical patent/US20170335396A1/en
Assigned to VERACYTE, INC. reassignment VERACYTE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PANKRATZ, DAN, KIM, SU YEON, DIGGANS, JAMES, HUANG, JING, KENNEDY, GIULIA C., PAGAN, MORAIMA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • 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
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]
    • 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
    • C12Q1/6869Methods for sequencing
    • C12Q1/6874Methods for sequencing involving nucleic acid arrays, e.g. sequencing by hybridisation
    • 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
    • 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
    • C12Q2600/156Polymorphic 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
    • C12Q2600/158Expression markers

Definitions

  • Interstitial lung diseases are a heterogeneous group of acute and chronic bilateral parenchymal pulmonary disorders with similar clinical manifestations, but a wide spectrum of severity and outcome 1,2 .
  • IPF idiopathic pulmonary fibrosis
  • Most patients diagnosed with IPF die within five years of their initial diagnosis 7,8 .
  • the recent availability of two new drugs and other therapeutics in development may change this picture 9-11 , and accurate diagnosis is critical for appropriate therapeutic intervention 5,12 .
  • IPF can be challenging to diagnose.
  • the diagnostic approach to IPF requires exclusion of other interstitial pneumonias, as well as connective tissue disease and environmental and occupational exposures 3-6 .
  • Patients suspected of having IPF usually undergo high-resolution computed tomography (HRCT), which confirms the disease with high specificity only if the pattern of usual interstitial pneumonia (UIP) is clearly evident 5,13 .
  • HRCT high-resolution computed tomography
  • UIP interstitial pneumonia
  • diagnosis necessitates an invasive surgical lung biopsy (SLB) to clarify the histopathologic features of interstitial pneumonia and/or UIP pattern 5,14 and the typical length of time to diagnose IPF from the onset of symptoms may be 1-2 years 15 .
  • SLB invasive surgical lung biopsy
  • the present invention provides a method and/or system for detecting whether a lung tissue sample is positive for usual interstitial pneumonia (UIP) or non-usual interstitial pneumonia (non-UIP).
  • a method is provided for: assaying the expression level of each of a first group of transcripts and a second group of transcripts in a test sample of a subject, wherein the first group of transcripts includes any one or more of the genes overexpressed in UIP and listed in any of Tables 5, 7, 9, 10, 11, and 12 and the second group of transcripts includes any one or more of the genes under-expressed in UIP and listed in any of Tables 5, 8, 9, 10, 11 or 12.
  • the method further provides for comparing the expression level of each of the first group of transcripts and the second group of transcripts with reference expression levels of the corresponding transcripts to (1) classify said lung tissue as usual interstitial pneumonia (UIP) if there is (a) an increase in an expression level corresponding to the first group or (b) a decrease in an expression level corresponding to the second group as compared to the reference expression levels, or (2) classify the lung tissue as non-usual interstitial pneumonia (non-UIP) if there is (c) an increase in the expression level corresponding to the second group or (d) a decrease in the expression level corresponding to the first group as compared to the reference expression levels.
  • the method further provides for determining and/or comparing sequence variants for any of the one or more genes listed in tables 5, 8, 9, 11, and/or 12.
  • the present invention provides a method and/or system for detecting whether a lung tissue sample is positive for usual interstitial pneumonia (UIP) or non-usual interstitial pneumonia (non-UIP).
  • the method and/or system is used to assay by sequencing, array hybridization, or nucleic acid amplification the expression level of each of a first group of transcripts and a second group of transcripts in a test sample from a lung tissue of a subject, wherein the first group of transcripts includes any one or more of the genes over-expressed in UIP and listed in Tables 5, 7, 9, 10, 11 or 12 and the second group of transcripts includes any one or more of the genes under-expressed in UIP and listed in Tables 5, 8, 9, 10, 11 or 12.
  • the method and/or system further compares the expression level of each of the first group of transcripts and the second group of transcripts with reference expression levels of the corresponding transcripts to (1) classify said lung tissue as usual interstitial pneumonia (UIP) if there is (a) an increase in an expression level corresponding to the first group or (b) a decrease in an expression level corresponding to the second group as compared to the reference expression levels, or (2) classify the lung tissue as non-usual interstitial pneumonia (non-UIP) if there is (c) an increase in the expression level corresponding to the second group or (d) a decrease in the expression level corresponding to the first group as compared to the reference expression levels.
  • UIP interstitial pneumonia
  • the present invention provides a method and/or system for detecting whether a test sample is positive for UIP or non-UIP by
  • test sample is a biopsy sample or a bronchoalveolar lavage sample. In some embodiments, the test sample is fresh-frozen or fixed.
  • the transcript expression levels are determined by RT-PCR, DNA microarray hybridization, RNASeq, or a combination thereof. In some embodiments, one or more of the transcripts is labeled.
  • the method comprises detecting cDNA produced from RNA expressed in the test sample, wherein, optionally, the cDNA is amplified from a plurality of cDNA transcripts prior to the detecting step.
  • the methods of the present invention further comprise measuring the expression level of at least one control nucleic acid in the test sample.
  • the methods of the present invention classify the lung tissue as any one of interstitial lung diseases (ILD), a particular type of ILD, a non-ILD, or non-diagnostic.
  • methods of the present invention classify the lung tissue as either idiopathic pulmonary fibrosis (IPF) or Nonspecific interstitial pneumonia (NSIP).
  • the method and/or system of the present invention comprises assaying the test sample for the expression level of one or more transcripts of any one of SEQ ID NOS: 1-22. In some embodiments, the method further comprises assaying the test sample for the expression level of from 1 to 20 other genes. In some embodiments, the other genes comprise one or more, or optionally all of HMCN2, ADAMTSL1, CD79B, KEL, KLHL14, MPP2, NMNAT2, PLXDC1, CAPN9, TALDO1, PLK4, IGHV3-72, IGKV1-9, and CNTN4.
  • the method and/or systems of the present invention further comprise using smoking status as a covariate during training of a UIP vs. non-UIP classifier disclosed herein, wherein, optionally, the smoking status is determined by detecting an expression profile indicative of the subject's smoker status. In some embodiments, such a classifier is used to determine whether a test sample is UIP or non-UIP.
  • the method and/or systems of the present invention comprises training a UIP vs. non-UIP classifier, wherein genes that are susceptible to smoker-status bias are excluded or weighed differently than genes that are not susceptible to smoker-status bias during the classifier training.
  • the present invention provides a method and/or system for detecting whether a lung tissue sample is positive for usual interstitial pneumonia (UIP) or non-usual interstitial pneumonia (non-UIP), as described herein, wherein the method comprises a first classification of a test sample as smoker or non-smoker using a first classifier trained to recognize gene signatures that distinguish smokers from non-smokers; and wherein the method further comprises a second classification of the test sample a UIP or non-UIP, wherein the second classification step uses a second or third classifier, which second and third classifiers are trained to distinguish UIP vs.
  • UIP interstitial pneumonia
  • non-UIP non-usual interstitial pneumonia
  • non-UIP in smokers in smokers
  • non-smoker-specific classifier non-smokers
  • the second classification uses either (i) the smoker-specific classifier if the test sample is classified as smoker in the first classification or (ii) the non-smoker-specific classifier if the test sample is classified as non-smoker in the first classification.
  • the present invention provides a method and/or system for detecting whether a lung tissue sample is positive for usual interstitial pneumonia (UIP) or non-usual interstitial pneumonia (non-UIP), wherein the methods comprise implementing a classifier trained using one or more feature selected from gene expression, variants, mutations, fusions, loss of heterozygoxity (LOH), and biological pathway effect.
  • the classifier is trained using features comprising gene expression, sequence variants, mutations, fusions, loss of heterozygoxity (LOH), and biological pathway effect.
  • the present invention provides for assaying 2 or more different transcripts, or 3 or more, 4 or more, 5 or more, 10 or more, 15 or more, 20 or more, or more than 20 different transcripts in the first group and/or 2 or more different transcripts, or 3 or more, 4 or more, 5 or more, 10 or more, 15 or more, 20 or more, or more than 20 different transcripts in the second group.
  • the method provides for detecting 2 or more different transcripts of any one of SEQ ID NOS: 1-22, or 3 or more, 4 or more, 5 or more, 10 or more, 15 or more, 20 or more, or more than 20 different transcripts of any one of SEQ ID NOS: 1-22.
  • the current methods provide for assaying the test sample for the expression level of all of the transcripts of SEQ ID NOS: 1-22.
  • the method further comprising assaying the test sample for the expression level of from 1 to 20 other genes.
  • the method provides for assaying one or more of HMCN2, ADAMTSL1, CD79B, KEL, KLHL14, MPP2.
  • FIG. 1 Pairwise correlation on explant samples obtained from three patients diagnosed with IPF (Patients P1, P2, and P3). Locations (upper or lower, central or peripheral) are indicated for each sample. The top 200 differentially expressed genes separating IPF samples from normal lung samples were used to compute pairwise Pearson correlation coefficients and plotted as a heatmap with higher correlation represented in magenta color, and lower correlation represented in green color. Correlation between and with normal lung samples are in the 0-7 range (not shown).
  • FIGS. 2A-2D Performance of a classifier built using microarray data.
  • ROC curves were used to characterize performance in the training set using leave-one-patient-out (LOPO) cross-validation ( FIG. 2A ) and in the independent test set by scoring the samples with a fixed model ( FIG. 2C ).
  • Scores for individual samples are shown across patients in the training set ( FIG. 2B ), and across patients in an independent test set ( FIG. 2D ).
  • Patient-level pathology diagnosis is shown on the x-axis. Samples with UIP pathology labels are indicated by closed circles, and non-UIP samples by pathology are shown in open triangles. A dotted horizontal line is drawn to indicate the threshold that corresponds to 92% specificity and 64% sensitivity ( FIG. 2B ) and 92% specificity and 82% sensitivity ( FIG. 2D ).
  • FIGS. 3A-3D Performance of classifiers built using RNASeq ( FIG. 3A and FIG. 3B ) and microarray on the matched set ( FIG. 3C and FIG. 3D ).
  • Leave-one-patient-out (LOPO) cross-validation was performed and receiver operator characteristic (ROC) curves were produced for RNASeq ( FIG. 3A ) and microarray ( FIG. 3C ) classifiers.
  • Scores for individual samples in the training sets are shown for RNASeq ( FIG. 3B ), and microarray ( FIG. 3D ) classification.
  • Patient-level pathology diagnosis is shown on the x-axis. Samples with UIP pathology labels are indicated by closed circles, and non-UIP samples by pathology are shown in open triangles.
  • a score threshold corresponding to 95% specificity is indicated as a horizontal line in FIG. 3B and FIG. 3D .
  • FIG. 4 Simulation study assessing the impact of mislabeling on the classification performance.
  • individual samples' classification labels were swapped to another class label with a weight accounting for the disagreement level of three expert pathology diagnoses.
  • Each boxplot was drawn using LOPO CV performances (AUC) from 100 repeated simulations.
  • AUC LOPO CV performances
  • FIG. 6 Location of lung samplings from three normal organ donors (top) and three patients diagnosed with IPF (bottom). Donors N1-N3 and P3 were female. Donors P1 and P2 were male.
  • FIG. 7A Illustration of a computer system usable for implementing aspects disclosed herein.
  • FIG. 7B Detailed illustration of the processor of the computer system of FIG. 7A .
  • FIG. 7C Detailed illustration of one non-limiting method of the present invention, wherein gene product expression data for known UIP and non-UIP samples are used to train a classifier (e.g., using a classifier training module) for differentiating UIP vs. non-UIP, wherein the classifier optionally considers smoker status as a covariant, and wherein gene product expression data from unknown samples are input into the trained classifier to identify the unknown samples as either UIP or non-UIP, and wherein the results of the classification via the classifier are defined and output via a report.
  • a classifier e.g., using a classifier training module
  • the classifier optionally considers smoker status as a covariant
  • gene product expression data from unknown samples are input into the trained classifier to identify the unknown samples as either UIP or non-UIP, and wherein the results of the classification via the classifier are defined and output via a report.
  • FIG. 8 Differential gene expression in UIP and Non-UIP samples between smokers and non-smokers. The number of genes differentially expressed between UIP and Non UIP samples differs drastically between smokers and non-smokers.
  • FIG. 9 Shows differential gene expression between UIP and Non-UIP samples is susceptible to smoker-status bias.
  • Direction i.e., over- vs. under-expression
  • magnitude circle size
  • FIGS. 10A-10D Examples of genes that are differentially expressed in UIP vs. Non-UIP and the effect of smoking status on expression levels.
  • FIG. 10A differential expression of IGHV3-72 in UIP vs Non-UIP smokers vs. non-smokers.
  • FIG. 10B differential expression of CPXM1 in UIP vs Non-UIP smokers vs. non-smokers.
  • FIG. 10C differential expression of BPIFA1 in UIP vs Non-UIP smokers vs. non-smokers.
  • FIG. 10D differential expression of HLA-U in UIP vs Non-UIP smokers vs. non-smokers.
  • Interstitial lung disease or “ILD” (also known as diffuse parenchymal lung disease (DPLD)) as used herein refers to a group of lung diseases affecting the interstitium (the tissue and space around the air sacs of the lungs). ILD can be classified according to a suspected or known cause, or can be idiopathic.
  • DPLD diffuse parenchymal lung disease
  • ILD can be classified as caused by inhaled substances (inorganic or organic), drug induced (e.g., antibiotics, chemotherapeutic drugs, antiarrhythmic agents, statins), associated with connective tissue disease (e.g., systemic sclerosis, polymyositis, dermatomyositis, systemic lupus erythematous, rheumatoid arthritis), associated with pulmonary infection (e.g., atypical pneumonia, Pneumocystis pneumonia (PCP), tuberculosis, Chlamydia trachomatis , Respiratory Syncytial Virus), associated with a malignancy (e.g., Lymphangitic carcinomatosis), or can be idiopathic (e.g., sarcoidosis, idiopathic pulmonary fibrosis, Hamman-Rich syndrome, antisynthetase syndrome).
  • connective tissue disease e.g., systemic sclerosis
  • ILD Inflammation refers to an analytical grouping of inflammatory ILD subtypes characterized by underlying inflammation. These subtypes can be used collectively as a comparator against IPF and/or any other non-inflammation lung disease subtype. “ILD inflammation” can include HP, NSIP, sarcoidosis, and/or organizing pneumonia.
  • Idiopathic interstitial pneumonia or “IIP” (also referred to as noninfectious pneumonia” refers to a class of ILDs which includes, for example, desquamative interstitial pneumonia, nonspecific interstitial pneumonia, lymphoid interstitial pneumonia, cryptogenic organizing pneumonia, and idiopathic pulmonary fibrosis.
  • IPF pulmonary fibrosis
  • IPF interstitial pneumonia
  • Nonspecific interstitial pneumonia or “NSIP” is a form of idiopathic interstitial pneumonia generally characterized by a cellular pattern defined by chronic inflammatory cells with collagen deposition that is consistent or patchy, and a fibrosing pattern defined by a diffuse patchy fibrosis. In contrast to UIP, there is no honeycomb appearance nor fibroblast foci that characterize usual interstitial pneumonia.
  • “Hypersensitivity pneumonitis” or “HP” refers to also called extrinsic allergic alveolitis, (EAA) refers to an inflammation of the alveoli within the lung caused by an exaggerated immune response and hypersensitivity to as a result of an inhaled antigen (e.g., organic dust).
  • EAA extrinsic allergic alveolitis
  • “Pulmonary sarcoidosis” or “PS” refers to a syndrome involving abnormal collections of chronic inflammatory cells (granulomas) that can form as nodules.
  • the inflammatory process for HP generally involves the alveoli, small bronchi, and small blood vessels. In acute and subacute cases of HP, physical examination usually reveals dry rales.
  • microarray refers to an ordered arrangement of hybridizable array elements, preferably polynucleotide probes, on a substrate.
  • polynucleotide when used in singular or plural, generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • polynucleotides as defined herein include, without limitation, single- and double-stranded DNA, DNA including single- and double-stranded regions, single- and double-stranded RNA, and RNA including single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or include single- and double-stranded regions.
  • polynucleotide refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • the strands in such regions may be from the same molecule or from different molecules.
  • the regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules.
  • One of the molecules of a triple-helical region often is an oligonucleotide.
  • polynucleotide can also include DNAs (e.g., cDNAs) and RNAs that contain one or more modified bases (e.g., to provide a detectable signal, such as a fluorophore).
  • DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein.
  • DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritiated bases are included within the term “polynucleotides” as defined herein.
  • polynucleotide embraces all chemically, enzymatically and/or metabolically modified forms of unmodified polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells.
  • oligonucleotide refers to a relatively short polynucleotide (e.g., 100, 50, 20 or fewer nucleotides) including, without limitation, single-stranded deoxyribonucleotides, single- or double-stranded ribonucleotides, RNA:DNA hybrids and double-stranded DNAs. Oligonucleotides, such as single-stranded DNA probe oligonucleotides, are often synthesized by chemical methods, for example using automated oligonucleotide synthesizers that are commercially available. However, oligonucleotides can be made by a variety of other methods, including in vitro recombinant DNA-mediated techniques and by expression of DNAs in cells and organisms.
  • RNA transcript RNA transcription products
  • a gene product can be, for example, a polynucleotide gene expression product (e.g., an unspliced RNA, an mRNA, a splice variant mRNA, a microRNA, a fragmented RNA, and the like) or a protein expression product (e.g., a mature polypeptide, a post-translationally modified polypeptide, a splice variant polypeptide, and the like).
  • the gene expression product may be a sequence variant including mutations, fusions, loss of heterozygoxity (LOH), and.or biological pathway effects.
  • normalized expression level refers to a level of the gene product normalized relative to one or more reference (or control) gene expression products.
  • a “reference expression level” as applied to a gene expression product refers to an expression level for one or more reference (or control) gene expression products.
  • a “reference normalized expression level” as applied to a gene expression product refers to a normalized expression level value for one or more reference (or control) gene expression products (i.e., a normalized reference expression level).
  • a reference expression level is an expression level for one or more gene product in normal sample, as described herein. In some embodiments, a reference expression level is determined experimentally.
  • a reference expression level is a historical expression level, e.g., a database value of a reference expression level in a normal sample, which sample indicates a single reference expression level, or a summary of a plurality of reference expression levels (such as, e.g., (i) an average of two or more, preferably three or more reference expression levels from replicate analysis of the reference expression level from a single sample; (ii) an average of two or more, preferably three or more reference expression levels from analysis of the reference expression level from a plurality of different samples (e.g., normal samples); (iii) and a combination of the above mentioned steps (i) and (ii) (i.e., average of reference expression levels analyzed from a plurality of samples, wherein at least one of the reference expression levels are analyzed in replicate).
  • the “reference expression level” is an expression level of sequence variants, for example, in a sample that has been definitively determined to be UIP or non-UIP by other means (i.
  • a “reference expression level value” as applied to a gene expression product refers to an expression level value for one or more reference (or control) gene expression products.
  • a “reference normalized expression level value” as applied to a gene expression product refers to a normalized expression level value for one or more reference (or control) gene expression products.
  • “Stringency” of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured DNA to re-anneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, (Wiley Interscience, 1995).
  • “Stringent conditions” or “high stringency conditions”, as defined herein, typically: (1) employ low ionic strength solutions and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5 ⁇ SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 ⁇ Denhardt's solution, sonicated salmon sperm DNA (50 ⁇ g/ml), 0.1% SDS, and 10% dextran sulfate
  • Modely stringent conditions may be identified as described by Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Press, 1989), and include the use of washing solution and hybridization conditions (e.g., temperature, ionic strength and % SDS) less stringent that those described above.
  • washing solution and hybridization conditions e.g., temperature, ionic strength and % SDS
  • An example of moderately stringent condition is overnight incubation at 37° C.
  • “Sensitivity” as used herein refers to the proportion of true positives of the total number tested that actually have the target disorder (i.e., the proportion of patients with the target disorder who have a positive test result). “Specificity” as used herein refers to the proportion of true negatives of all the patients tested who actually do not have the target disorder (i.e., the proportion of patients without the target disorder who have a negative test result).
  • references to “at least one,” “at least two.” “at least five,” etc. of the genes listed in any particular gene set means any one or any and all combinations of the genes listed.
  • splicing and “RNA splicing” are used interchangeably and refer to RNA processing that removes introns and joins exons to produce mature mRNA with continuous coding sequence that moves into the cytoplasm of a eukaryotic cell.
  • exon refers to any segment of an interrupted gene that is represented in a mature RNA product (B. Lewin, Genes 7V (Cell Press, 1990)).
  • intron refers to any segment of DNA that is transcribed but removed from within the transcript by splicing together the exons on either side of it.
  • exon sequences occur in the mRNA sequence of a gene as defined by Ref. SEQ ID numbers.
  • intron sequences are the intervening sequences within the genomic DNA of a gene, bracketed by exon sequences and usually having GT and AG splice consensus sequences at their 5′ and 3 ′ boundaries.
  • a “computer-based system” refers to a system of hardware, software, and data storage medium used to analyze information.
  • Hardware of a patient computer-based system can include a central processing unit (CPU), and hardware for data input, data output (e.g., display), and data storage.
  • the data storage medium can include any manufacture comprising a recording of the present information as described above, or a memory access device that can access such a manufacture.
  • module refers to any assembly and/or set of operatively-coupled electrical components that can include, for example, a memory, a processor, electrical traces, optical connectors, software (executing in hardware), and/or the like.
  • a module executed in the processor can be any combination of hardware-based module (e.g., a field-programmable gate array (FPGA), an application specific integrated circuit (ASIC), a digital signal processor (DSP)) and/or software-based module (e.g., a module of computer code stored in memory and/or executed at the processor) capable of performing one or more specific functions associated with that module.
  • FPGA field-programmable gate array
  • ASIC application specific integrated circuit
  • DSP digital signal processor
  • Record data programming or other information on a computer readable medium refers to a process for storing information, using any such methods as known in the art. Any convenient data storage structure may be chosen, based on the means used to access the stored information. A variety of data processor programs and formats can be used for storage, e.g. word processing text file, database format, etc.
  • a “processor” or “computing means” references any hardware and/or software combination that will perform the functions required of it.
  • a suitable processor may be a programmable digital microprocessor such as available in the form of an electronic controller, mainframe, server or personal computer (desktop or portable).
  • suitable programming can be communicated from a remote location to the processor, or previously saved in a computer program product (such as a portable or fixed computer readable storage medium, whether magnetic, optical or solid state device based).
  • a magnetic medium or optical disk may carry the programming, and can be read by a suitable reader communicating with each processor at its corresponding station.
  • test sample is a sample of one or more cells, preferable a tissue sample (e.g., a lung tissue sample such as a transbronchial biopsy (TBB) sample) obtained from a subject.
  • a test sample is a biopsy sample obtained by any means known in the art.
  • the test sample is a sample obtained by a video-assisted thoracoscopic surgery (VATS); a bronchoalveolar lavage (BAL); a transbronchial biopsy (TBB); or a cryo-transbronchial biopsy.
  • the test sample is obtained from a patient suspected of having a lung disease, e.g., an ILD, based on clinical signs and symptoms with which the patient presents (e.g., shortness of breath (generally aggravated by exertion), dry cough), and, optionally the results of one or more of an imaging test (e.g., chest X-ray, computerized tomography (CT)), a pulmonary function test (e.g., spirometry, oximetry, exercise stress test), lung tissue analysis (e.g., histological and/or cytological analysis of samples obtained by bronchoscopy, bronchoalveolar lavage, surgical biopsy).
  • an imaging test e.g., chest X-ray, computerized tomography (CT)
  • CT computerized tomography
  • pulmonary function test e.g., spirometry, oximetry, exercise stress test
  • lung tissue analysis e.g., histological and/or cytological analysis of samples obtained by bronchoscopy,
  • a “gene signature” is a gene expression pattern (i.e., expression level of one or more gene, or fragments thereof), which is indicative of some characteristic or phenotype.
  • gene signature refers to the expression (and/or lack of expression) of a gene, a plurality of genes, a fragment of a gene or a plurality fragments of one or more genes, which expression and/or lack of expression is indicative of UIP, Non-UIP, smoker-status, or Non-smoker-status.
  • a smoker is meant to refer to a subject who currently smokes cigarettes or a person who has smoked cigarettes in the past or a person who has the gene signature of a person who currently smokes cigarettes or has smoked cigarettes in the past.
  • variant when used to describe a feature used during training of a classifier of the present invention, refers to an alternative splice variant.
  • mutation when used to describe a feature used during training of a classifier of the present invention, refers to a sequence deviation from a known normal reference sequence.
  • the deviation is a deviation from an accepted native gene sequence according to a publically accessible database such as the UniGene database (Pontius J U, Wagner L, Schuler G D. UniGene: a unified view of the transcriptome. In: The NCBI Handbook. Bethesda (Md.): National Center for Biotechnology Information; 2003, incorporated herein), RefSeq (The NCBI handbook [Internet].
  • the mutation includes an addition, deletion, or substitution of a sequence residue present in the reference sequence.
  • Abbreviations include: HRCT, high-resolution computed tomography; VATS, video-assisted thorascopic surgery; SLB, surgical lung biopsy; TBB, transbronchial biopsy; RB, respiratory bronchiolitis; OP, organizing pneumonia, DAD, diffuse alveolar damage, CIF/NOC, chronic interstitial fibrosis not otherwise classified; MDT, multidisciplinary team; CV, cross-validation; LOPO, leave-one-patient-out; ROC, receiver operator characteristic; AUC, area under the curve; RNASeq, RNA sequencing by next-generation sequencing technology; NGS, next-generation sequencing technology; H&E, hematoxylin and eosin; FDR, false discovery rate; IRB, Institutional Review Board; ATS, American Thoracic Society; COPD, chronic obstructive pulmonary disease; KEGG, Kyoto Encyclopedia of Genes and Genomes; CI, confidence interval
  • the accurate diagnosis of UIP from samples where expert pathology is not available stands to benefit ILD patients by accelerating diagnosis, thus facilitating treatment decisions and reducing surgical risk to patients and costs to the healthcare system.
  • the methods and/or systems disclosed herein provide classifiers which can differentiate UIP from non-UIP patterns based on high-dimensional transcriptional data without prior knowledge of clinical or demographic information.
  • the present invention provides methods for differentiating UIP from non-UIP using a classifier that comprises or consists of one or more sequences or fragments thereof presented in any of Tables 5, 7, 8, 9, 10, 11, or 12 or at least one sequence or fragment thereof from each of Tables 5, 7, 8, 9, 10, 11 and 12.
  • the present invention provides such methods that use a classifier comprising or consisting of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more of the sequences provided in any one or more or all of Tables 5, 7, 8, 9, 10, 11 and 12.
  • the present invention provides such methods that use classifiers comprising or consisting of at least 11, 12, 13, 14, 15, 20, 30, 50, 100, 150, 200, 250, 300, or more sequences provided in any one or more or all of Tables 5, 7, 8, 9, 10, 11 and 12, including all integers (e.g., 16, 17, 18, 19, 21, 22, 23, 24, 25 sequences, etc.) and ranges (e.g., from about 1-10 sequences from any one or more or all of Tables 5, 7, 8, 9, 10, 11, and 12, from about 10-15 sequences, 10-20 sequences, 5-30 sequences, 5-50 sequences, 10-100 sequences, 50-200 sequences, etc.) between.
  • classifiers comprising or consisting of at least 11, 12, 13, 14, 15, 20, 30, 50, 100, 150, 200, 250, 300, or more sequences provided in any one or more or all of Tables 5, 7, 8, 9, 10, 11 and 12, including all integers (e.g., 16, 17, 18, 19, 21, 22, 23, 24, 25 sequences, etc.) and ranges (e.g., from about 1-10 sequences from any one
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of one or more of the following sequences or fragments thereof: 1) HLA-F (SEQ ID NO.:1), 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:13), 14) PTCHD4 (SEQ ID NO.:14
  • the present invention provides a method and/or system for differentiating UIP from non-UIP using a classifier that comprises or consists of 2; 3; 4; 5; 6; 7; 8; 9; 10; 11; 12; 13; 14; 15; 16; 17; 18; 19; 20; or 21 of the following sequences: 1) HLA-F (SEQ ID NO.:1), 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.: 11), 12) DZIP1 (SEQ ID NO.:1)
  • the present invention provides a method and/or system for differentiating UIP from non-UIP using a classifier that comprises or consists of all of the following sequences: 1) HLA-F (SEQ ID NO.:1), 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.: 11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:13), 14) PTCHD4 (SEQ ID NO.:14), 15) PDL
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of one or more of the following sequences or fragments thereof: 1) HLA-F (SEQ ID NO.:1), 2) HMCN2, 3) ADAMTSL1, 4) CD79B, 5) KEL, 6) KLHL14, 7) MPP2, 8) NMNAT2, 9) PLXDC1, 10) CAPN9, 11) TALDO1, 12) PLK4, 13) IGHV3-72, 14) IGKV1-9, and 15) CNTN4.
  • the classifier may contain 1, 2, 3, 4, 5, 6, 7, 8, or more additional genes.
  • the classifier may omit 1, 2, 3, 4, 5, 6, 7, 8, or more, of these genes, while optionally including other genes.
  • the present invention provides a method and/or system for differentiating UIP from non-UIP using a classifier that comprises or consists of 2; 3; 4; 5; 6; 7; 8; 9; 10; 11; 12; 13; or 14 of the following sequences: 1) HLA-F (SEQ ID NO.:1), 2) HMCN2, 3) ADAMTSL1, 4) CD79B, 5) KEL, 6) KLHL14, 7) MPP2, 8) NMNAT2, 9) PLXDC1, 10) CAPN9, 11) TALDO1, 12) PLK4, 13) IGHV3-72, 14) IGKV1-9, and 15) CNTN4.
  • the classifier may contain 1, 2, 3, 4, 5, 6, 7, 8, or more additional genes.
  • the classifier may omit 1, 2, 3, 4, 5, 6, 7, 8, or more, of these genes, while optionally including other genes.
  • the present invention provides a method and/or system for differentiating UIP from non-UIP using a classifier that comprises or consists of the following sequences: 1) HLA-F (SEQ ID NO.:1), 2) HMCN2, 3) ADAMTSL1, 4) CD79B, 5) KEL, 6) KLHL14, 7) MPP2, 8) NMNAT2, 9) PLXDC1, 10) CAPN9, 11) TALDO1, 12) PLK4, 13) IGHV3-72, 14) IGKV1-9, and 15) CNTN4.
  • the classifier may contain 1, 2, 3, 4, 5, 6, 7, 8, or more additional genes.
  • the classifier may omit 1, 2, 3, 4, 5, 6, 7, 8, or more, of these genes, while optionally including other genes.
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of HLA-F (SEQ ID NO.:1) or fragments thereof.
  • the method uses a classifier comprising 1) HLA-F (SEQ ID NO.:1) and at least one of 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1) and
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of HMCN2 or fragments thereof.
  • the method uses a classifier comprising HMCN2 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of ADAMTSL1 or fragments thereof.
  • the method uses a classifier comprising ADAMTSL1 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1)
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of CD79B or fragments thereof.
  • the method uses a classifier comprising CD79B and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1) HLA-F (SEQ
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of KEL or fragments thereof.
  • the method uses a classifier comprising KEL and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.: 11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:13),
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of KLHL14 or fragments thereof.
  • the method uses a classifier comprising KLHL14 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of MPP2 or fragments thereof.
  • the method uses a classifier comprising MPP2 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1)) HLA-F (S
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of NMNAT2 or fragments thereof.
  • the method uses a classifier comprising NMNAT2 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of PLXDC1 or fragments thereof.
  • the method uses a classifier comprising PLXDC1 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1)
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of CAPN9 or fragments thereof.
  • the method uses a classifier comprising CAPN9 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1)) HLA-F (S
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of TALDO1 or fragments thereof.
  • the method uses a classifier comprising TALDO1 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of PLK4 or fragments thereof.
  • the method uses a classifier comprising PLK4 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1)) HLA-F (S
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of IGHV3-72 or fragments thereof.
  • the method uses a classifier comprising IGHV3-72 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1) (
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of IGKV1-9 or fragments thereof.
  • the method uses a classifier comprising IGKV1-9 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:1)
  • the present invention provides methods and/or systems for differentiating UIP from non-UIP using a classifier that comprises or consists of CNTN4 or fragments thereof.
  • the method uses a classifier comprising CNTN4 and at least one of 1) HLA-F (SEQ ID NO.:1) 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.
  • the present invention provides a method and/or system for differentiating UIP from non-UIP using a classifier that comprises or consists of 2; 3; 4; 5; 6; 7; 8; 9; 10; 11; 12; 13; 14; 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 of the following sequences: 1) HLA-F (SEQ ID NO.:1), 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11
  • the present invention provides a method and/or system for differentiating UIP from non-UIP using a classifier that comprises or consists of all of the following sequences: 1) HLA-F (SEQ ID NO.:1), 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.: 11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:13), 14) PTCHD4 (SEQ ID NO.:14), 15) PDL
  • the classifier may contain 1, 2, 3, 4, 5, 6, 7, 8, or more additional genes. In other aspects, the classifier may omit 1, 2, 3, 4, 5, 6, 7, 8, or more, of these genes, while optionally including other genes.
  • the present invention provides a method and/or system for differentiating UIP from non-UIP using a classifier described herein, wherein the method further comprises implementing a classifier that classifies the subject as a smoker or non-smoker.
  • a smoker status classification can optionally be implemented prior to implementing a UIP vs.
  • Non-UIP classifier, or a smoker status classification step can be built in as a covariate used during the training (e.g., using a classifier training module) of a UIP vs.
  • Non-UIP classifier of the present invention can be built in as a covariate used during the training (e.g., using a classifier training module) of a UIP vs.
  • Non-UIP classifier of the present invention can be built
  • the method of and/or system for differentiating UIP from non-UIP using a classifier described herein further comprises a step of excluding or assigning differential weight to certain genes or variants thereof that are susceptible to smoker-status bias during the training (e.g., using a classifier training module) or implementation of the UIP vs. Non-UIP classifier.
  • smoke status bias refers to genes or variants thereof, which in non-smoker patients are differentially expressed in UIP vs. non-UIP patients, but which are not detectably differentially expressed in UIP vs. non-UIP patients that are (or have been) smokers.
  • the method of and/or system for the present invention comprises a tiered classifier comprising at least a first and a second classifier, wherein the first classifier is trained (e.g., using a classifier training module) to recognize gene signatures that distinguish smokers from non-smokers, and a second classifier is trained (e.g., using a classifier training module) to distinguish UIP vs. Non UIP in smokers or non-smokers, respectively.
  • a tiered classifier comprising at least a first and a second classifier, wherein the first classifier is trained (e.g., using a classifier training module) to recognize gene signatures that distinguish smokers from non-smokers, and a second classifier is trained (e.g., using a classifier training module) to distinguish UIP vs. Non UIP in smokers or non-smokers, respectively.
  • the method and/or systems of the present invention comprises: extracting nucleic acids (e.g., RNA, such as, e.g., total RNA) from a test sample (e.g. lung tissue;
  • RNA nucleic acids
  • test sample e.g. lung tissue
  • the method and/or system of the present invention further comprises incorporating smoker status into the training exercise.
  • smoker status is optionally incorporated in one of the following ways:
  • Non-UIP classification is performed using a classifier trained (e.g., using a classifier training module) with UIP and Non-UIP samples from smokers. Conversely, if the pre-classifier determines that the test sample is from a non-smoker, a UIP vs. Non-UIP classification is performed using a classifier trained (e.g., using a classifier training module) with UIP and Non-UIP samples from non-smokers. In some embodiments, such smoker- or non-smoker-specific classifiers provide improved diagnostic performance due, at least in part, to a reduction in background noise caused by the inclusion of genes susceptible to smoker-status bias in the classifier training.
  • the present invention also provides suitable classifiers for use in methods of differentiating UIP from non-UIP, as disclosed herein.
  • the present invention provides a classifier suitable for differentiating UIP from non-UIP, wherein the classifier is trained (e.g., using a classifier training module) using microarray or sequencing data from a sample corresponding to one or more histopathology label determined by an expert pathologist.
  • the sample is labelled UIP or Non-UIP.
  • the present invention presents a classifier comprising or consisting of one or more sequences or fragments thereof presented in any of Tables 5, 7, 8, 9, 10, 11, or 12, or at least one sequence or fragment thereof from each of Tables 5, 7, 8, 9, 10, 11, or 12.
  • the present invention provides a classifier comprising or consisting of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more of the sequences provided in any one or more or all of Tables 5, 7, 8, 9, 10, 11 and 12.
  • the present invention provides a classifier comprising or consisting of at least 11, 12, 13, 14, 15, 20, 30, 50, 100, 150, 200, 250, 300, or more sequences provided in any one or more or all of Tables 5, 7, 8, 9, 10, 11, or 12, including all integers (e.g., 16, 17, 18, 19, 21, 22, 23, 24, 25 sequences, etc.) and ranges (e.g., from about 1-10 sequences from any one or more or all of Tables 5, 7, 8, 9, 10, 11, or 12, from about 10-15 sequences, 10-20 sequences, 5-30 sequences, 5-50 sequences, 10-100 sequences, 50-200 sequences from any one or more or all of Tables 5, 7, 8, 9, 10, 11, or 12, etc.) between.
  • a classifier comprising or consisting of at least 11, 12, 13, 14, 15, 20, 30, 50, 100, 150, 200, 250, 300, or more sequences provided in any one or more or all of Tables 5, 7, 8, 9, 10, 11, or 12, including all integers (e.g., 16, 17, 18, 19, 21, 22, 23, 24, 25 sequences, etc.) and ranges
  • the present invention provides a classifier that comprises or consists of all sequences provided in Table 5, all sequences provided in Table 7, all sequences provided in Table 8, all sequences provided in Table 9, all sequences provided in table 10, all sequences provided in Table 11, or all sequences provided in Table 12. In one embodiment, the present invention provides a classifier that comprises or consists of all sequences provided in each of Tables 5, 7, 8, 9, 10, 11, or 12.
  • the present invention provides a classifier for differentiating UIP from non-UIP, wherein the classifier comprises or consists of one or more of the following sequences or fragments thereof: 1) HLA-F (SEQ ID NO.:1), 2) CDKL2 (SEQ ID NO.:2), 3) GPR98 (SEQ ID NO.:3), 4) PRKCQ (SEQ ID NO.:4), 5) HLA-G (SEQ ID NO.:5), 6) PFKFB3 (SEQ ID NO.:6), 7) CEACAM1 (SEQ ID NO.:7), 8) RABGAP1L (SEQ ID NO.:8), 9) CD274 (SEQ ID NO.:9), 10) PRUNE2 (SEQ ID NO.:10), 11) ARAP2 (SEQ ID NO.:11), 12) DZIP1 (SEQ ID NO.:12), 13) MXRA7 (SEQ ID NO.:13), 14) PTCHD4 (SEQ ID NO.:14),
  • the classifier comprises or consists of all 22 of the above mentioned sequences.
  • the present invention provides a classifier for differentiating UIP from non-UIP, wherein the classifier comprises or consists of 2; 3; 4; 5; 6; 7; 8; 9; 10; 11; 12; 13; 14; 15; 16; 17; 18; 19; 20; or 21 of the abovementioned 22 sequences.
  • the classifier contains 1, 2, 3, 4, 5, 6, 7, 8, or more additional genes or fragments thereof.
  • the classifier omits 1, 2, 3, 4, 5, 6, 7, 8, or more, of the abovementioned 22 sequences, while optionally including other genes.
  • each of the 22 genes may be used in combination with any 1 or more, up to 20 more, of the other genes.
  • a lung tissue sample for use in a subject analytical or diagnostic method can be a biopsy sample (e.g., a biopsy sample obtained by video-assisted thoracoscopic surgery; VATS); a bronchoalveolar lavage (BAL) sample; a transbronchial biopsy; a cryo-transbronchial biopsy; and the like.”
  • Lung tissue samples for analysis can be provided in a suitable preservation solution.
  • Tissue samples can be obtained from a patient suspected of having a lung disease, e.g., an ILD, based on clinical signs and symptoms with which the patient presents (e.g., shortness of breath (generally aggravated by exertion), dry cough), and, optionally the results of one or more of an imaging test (e.g., chest X-ray, computerized tomography (CT)), a pulmonary function test (e.g., spirometry, oximetry, exercise stress test), lung tissue analysis (e.g., histological and/or cytological analysis of samples obtained by bronchoscopy, bronchoalveolar lavage, surgical biopsy).
  • an imaging test e.g., chest X-ray, computerized tomography (CT)
  • CT computerized tomography
  • pulmonary function test e.g., spirometry, oximetry, exercise stress test
  • lung tissue analysis e.g., histological and/or cytological analysis of samples obtained by bronchoscopy
  • the lung tissue sample can be processed in any of a variety of ways.
  • the lung tissue sample can be subjected to cell lysis.
  • the lung tissue sample can be preserved in RNAprotect solution (a solution that inhibits RNA degradation, e.g., that inhibits nuclease digestion of RNA) and subsequently subjected to cell lysis.
  • RNAprotect solution a solution that inhibits RNA degradation, e.g., that inhibits nuclease digestion of RNA
  • Components such as nucleic acids and/or proteins can be enriched or isolated from the lung tissue sample, and the enriched or isolated component can be used in a subject method.
  • Methods of enriching for and isolating components such nucleic acids and proteins are known in the art; and any known method can be used. Methods of isolating RNA for expression analysis have been described in the art.
  • the general methods for determining gene expression product levels are known to the art and may include but are not limited to one or more of the following: additional cytological assays, assays for specific proteins or enzyme activities, assays for specific expression products including protein or RNA or specific RNA splice variants, in situ hybridization, whole or partial genome expression analysis, microarray hybridization assays, serial analysis of gene expression (SAGE), enzyme linked immunoabsorbance assays, mass-spectrometry, immunohistochemistry, blotting, sequencing, RNA sequencing, DNA sequencing (e.g., sequencing of cDNA obtained from RNA); Next-Gen sequencing, nanopore sequencing, pyrosequencing, or Nanostring sequencing.
  • additional cytological assays assays for specific proteins or enzyme activities
  • assays for specific expression products including protein or RNA or specific RNA splice variants
  • in situ hybridization whole or partial genome expression analysis
  • microarray hybridization assays serial analysis of gene expression (SAGE), enzyme linked immunoabsorbance assay
  • gene expression product levels can be determined according to the methods described in Kim, et. al. (Lancet Respir Med. 2015 June; 3(6):473-82, incorporated herein in its entirety, including all supplements).
  • the terms “assaying” or “detecting” or “determining” are used interchangeably in reference to determining gene expression product levels, and in each case, it is contemplated that the above-mentioned methods of determining gene expression product levels are suitable for detecting or assaying gene expression product levels.
  • Gene expression product levels may be normalized to an internal standard such as total mRNA or the expression level of a particular gene including but not limited to glyceraldehyde 3 phosphate dehydrogenase, or tubulin.
  • a sample comprises cells harvested from a tissue sample (e.g., a lung tissue sample such as a TBB sample).
  • Cells can be harvested from a sample using standard techniques known in the art or disclosed herein. For example, in one embodiment, cells are harvested by centrifuging a cell sample and resuspending the pelleted cells. The cells can be resuspended in a buffered solution such as phosphate-buffered saline (PBS). After centrifuging the cell suspension to obtain a cell pellet, the cells can be lysed to extract nucleic acid, e.g, messenger RNA. All samples obtained from a subject, including those subjected to any sort of further processing, are considered to be obtained from the subject.
  • PBS phosphate-buffered saline
  • the sample in one embodiment, is further processed before detection of the gene expression products is performed as described herein.
  • mRNA in a cell or tissue sample can be separated from other components of the sample.
  • the sample can be concentrated and/or purified to isolate mRNA in its non-natural state, as the mRNA is not in its natural environment.
  • studies have indicated that the higher order structure of mRNA in vivo differs from the in vitro structure of the same sequence (see, e.g., Rouskin et al. (2014). Nature 505, pp. 701-705, incorporated herein in its entirety for all purposes).
  • mRNA from the sample in one embodiment, is hybridized to a synthetic DNA probe, which in some embodiments, includes a detection moiety (e.g., detectable label, capture sequence, barcode reporting sequence). Accordingly, in these embodiments, a non-natural mRNA-cDNA complex is ultimately made and used for detection of the gene expression product.
  • mRNA from the sample is directly labeled with a detectable label, e.g., a fluorophore.
  • the non-natural labeled-mRNA molecule is hybridized to a cDNA probe and the complex is detected.
  • cDNA complementary DNA
  • cDNA-mRNA hybrids are synthetic and do not exist in vivo.
  • cDNA is necessarily different than mRNA, as it includes deoxyribonucleic acid and not ribonucleic acid.
  • the cDNA is then amplified, for example, by the polymerase chain reaction (PCR) or other amplification method known to those of ordinary skill in the art.
  • LCR ligase chain reaction
  • Genomics 4:560 (1989)
  • Landegren et al. Science, 241:1077 (1988)
  • transcription amplification Kwoh et al., Proc. Natl. Acad. Sci. USA, 86:1173 (1989), incorporated by reference in its entirety for all purposes
  • self-sustained sequence replication Guatelli et al., Proc. Nat. Acad. Sci.
  • RNA based sequence amplification RNA based sequence amplification
  • NASBA nucleic acid based sequence amplification
  • the product of this amplification reaction i.e., amplified cDNA is also necessarily a non-natural product.
  • cDNA is a non-natural molecule.
  • the amplification process serves to create hundreds of millions of cDNA copies for every individual cDNA molecule of starting material. The number of copies generated are far removed from the number of copies of mRNA that are present in vivo.
  • cDNA is amplified with primers that introduce an additional DNA sequence (e.g., adapter, reporter, capture sequence or moiety, barcode) onto the fragments (e.g., with the use of adapter-specific primers), or mRNA or cDNA gene expression product sequences are hybridized directly to a cDNA probe comprising the additional sequence (e.g., adapter, reporter, capture sequence or moiety, barcode).
  • Amplification and/or hybridization of mRNA to a cDNA probe therefore serves to create non-natural double stranded molecules from the non-natural single stranded cDNA, or the mRNA, by introducing additional sequences and forming non-natural hybrids.
  • amplification procedures have error rates associated with them. Therefore, amplification introduces further modifications into the cDNA molecules.
  • a detectable label e.g., a fluorophore
  • a detectable label is added to single strand cDNA molecules.
  • Amplification therefore also serves to create DNA complexes that do not occur in nature, at least because (i) cDNA does not exist in vivo, (i) adapter sequences are added to the ends of cDNA molecules to make DNA sequences that do not exist in vivo, (ii) the error rate associated with amplification further creates DNA sequences that do not exist in vivo, (iii) the disparate structure of the cDNA molecules as compared to what exists in nature and (iv) the chemical addition of a detectable label to the cDNA molecules.
  • the expression of a gene expression product of interest is detected at the nucleic acid level via detection of non-natural cDNA molecules.
  • the gene expression products described herein include RNA comprising the entire or partial sequence of any of the nucleic acid sequences of interest, or their non-natural cDNA product, obtained synthetically in vitro in a reverse transcription reaction.
  • fragment is intended to refer to a portion of the polynucleotide that generally comprise at least 10, 15, 20, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 800, 900, 1,000, 1,200, or 1,500 contiguous nucleotides, or up to the number of nucleotides present in a fulllength gene expression product polynucleotide disclosed herein.
  • a fragment of a gene expression product polynucleotide will generally encode at least 15, 25, 30, 50, 100, 150, 200, or 250 contiguous amino acids, or up to the total number of amino acids present in a full-length gene expression product protein of the invention.
  • a gene expression profile may be obtained by whole transcriptome shotgun sequencing (“WTSS” or “RNAseq”; see, e.g., Ryan et al BioTechniques 45 : 81-94), which makes the use of high-throughput sequencing technologies to sequence cDNA in order to about information about a sample's RNA content.
  • WTSS whole transcriptome shotgun sequencing
  • RNAseq RNAseq
  • cDNA is made from RNA, the cDNA is amplified, and the amplification products are sequenced.
  • the cDNA may be sequenced using any convenient method.
  • the fragments may be sequenced using Illumina's reversible terminator method, Roche's pyrosequencing method (454), Life Technologies' sequencing by ligation (the SOLiD platform) or Life Technologies' Ion Torrent platform. Examples of such methods are described in the following references: Margulies et al (Nature 2005 437: 376-80); Ronaghi et al (Analytical Biochemistry 1996 242: 84-9); Shendure (Science 2005 309: 1728); Imelfort et al (Brief Bioinform. 2009 10:609-18); Fox et al (Methods Mol Biol.
  • the products may be sequenced using nanopore sequencing (e.g. as described in Soni et al Clin Chem 53: 1996-2001 2007, or as described by Oxford Nanopore Technologies).
  • Nanopore sequencing is a single-molecule sequencing technology whereby a single molecule of DNA is sequenced directly as it passes through a nanopore.
  • a nanopore is a small hole, of the order of 1 nanometer in diameter. Immersion of a nanopore in a conducting fluid and application of a potential (voltage) across it results in a slight electrical current due to conduction of ions through the nanopore. The amount of current which flows is sensitive to the size and shape of the nanopore.
  • the gene expression product of the subject methods is a protein
  • the amount of protein in a particular biological sample is analyzed using a classifier derived from protein data obtained from cohorts of samples.
  • the amount of protein can be determined by one or more of the following: enzyme-linked immunosorbent assay (ELISA), mass spectrometry, blotting, or immunohistochemistry.
  • gene expression product markers and alternative splicing markers may be determined by microarray analysis using, for example, Affymetrix arrays, cDNA microarrays, oligonucleotide microarrays, spotted microarrays, or other microarray products from Biorad, Agilent, or Eppendorf.
  • Microarrays provide particular advantages because they may contain a large number of genes or alternative splice variants that may be assayed in a single experiment.
  • the microarray device may contain the entire human genome or transcriptome or a substantial fraction thereof allowing a comprehensive evaluation of gene expression patterns, genomic sequence, or alternative splicing. Markers may be found using standard molecular biology and microarray analysis techniques as described in Sambrook Molecular Cloning a Laboratory Manual 2001 and Baldi, P., and Hatfield, W. G., DNA Microarrays and Gene Expression 2002.
  • Microarray analysis generally begins with extracting and purifying nucleic acid from a biological sample, (e.g. a biopsy or fine needle aspirate) using methods known to the art.
  • a biological sample e.g. a biopsy or fine needle aspirate
  • RNA e.g. DNA
  • niRNA RNA from other forms of RNA such as tRNA and rRNA.
  • Purified nucleic acid may further be labeled with a fluorescent label, radionuclide, or chemical label such as biotin, digoxigenin, or digoxin for example by reverse transcription, polymerase chain reaction (PCR), ligation, chemical reaction or other techniques.
  • the labeling can be direct or indirect which may further require a coupling stage.
  • the coupling stage can occur before hybridization, for example, using aminoallyl-UTP and NHS amino-reactive dyes (like cyanine dyes) or after, for example, using biotin and labelled streptavidin.
  • modified nucleotides e.g.
  • the aaDNA may then be purified with, for example, a column or a diafiltration device.
  • the aminoallyl group is an amine group on a long linker attached to the nucleobase, which reacts with a reactive label (e.g. a fluorescent dye).
  • the labeled samples may then be mixed with a hybridization solution which may contain sodium dodecyl sulfate (SDS), SSC, dextran sulfate, a blocking agent (such as COT1 DNA, salmon sperm DNA, calf thymus DNA, PolyA or PolyT), Denhardt's solution, formamine, or a combination thereof.
  • SDS sodium dodecyl sulfate
  • SSC dextran sulfate
  • a blocking agent such as COT1 DNA, salmon sperm DNA, calf thymus DNA, PolyA or PolyT
  • Denhardt's solution formamine, or a combination thereof.
  • a hybridization probe is a fragment of DNA or RNA of variable length, which is used to detect in DNA or RNA samples the presence of nucleotide sequences (the DNA target) that are complementary to the sequence in the probe.
  • the probe thereby hybridizes to single-stranded nucleic acid (DNA or RNA) whose base sequence allows probe-target base pairing due to complementarity between the probe and target.
  • the labeled probe is first denatured (by heating or under alkaline conditions) into single DNA strands and then hybridized to the target DNA.
  • the probe is tagged (or labeled) with a molecular marker; commonly used markers are 32P or Digoxigenin, which is nonradioactive antibody-based marker.
  • DNA sequences or RNA transcripts that have moderate to high sequence complementarity (e.g. at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or more complementarity) to the probe are then detected by visualizing the hybridized probe via autoradiography or other imaging techniques.
  • Hybridization probes used in DNA microarrays refer to DNA covalently attached to an inert surface, such as coated glass slides or gene chips, and to which a mobile cDNA target is hybridized.
  • a mix comprising target nucleic acid to be hybridized to probes on an array may be denatured by heat or chemical means and added to a port in a microarray.
  • the holes may then be sealed and the microarray hybridized, for example, in a hybridization oven, where the microarray is mixed by rotation, or in a mixer. After an overnight hybridization, non-specific binding may be washed off (e.g. with SDS and SSC).
  • the microarray may then be dried and scanned in a machine comprising a laser that excites the dye and a detector that measures emission by the dye.
  • the image may be overlaid with a template grid and the intensities of the features (e.g. a feature comprising several pixels) may be quantified.
  • kits can be used for the amplification of nucleic acid and probe generation of the subject methods.
  • kit that can be used in the present invention include but are not limited to Nugen WT-Ovation FFPE kit, cDNA amplification kit with Nugen Exon Module and Frag/Label module.
  • the NuGEN WT-OvationTM. FFPE System V2 is a whole transcriptome amplification system that enables conducting global gene expression analysis on the vast archives of small and degraded RNA derived from FFPE samples.
  • the system is comprised of reagents and a protocol required for amplification of as little as 50 ng of total FFPE RNA.
  • the protocol can be used for qPCR, sample archiving, fragmentation, and labeling.
  • the amplified cDNA can be fragmented and labeled in less than two hours for GeneChipTM. 3′ expression array analysis using NuGEN's FL-OvationTM. cDNA Biotin Module V2. For analysis using Affymetrix GeneChipTM. Exon and Gene ST arrays, the amplified cDNA can be used with the WT-Ovation Exon Module, then fragmented and labeled using the FL-OvationTM. cDNA Biotin Module V2. For analysis on Agilent arrays, the amplified cDNA can be fragmented and labeled using NuGEN's FL-Ovation®. cDNA Fluorescent Module.
  • Ambion WT-expression kit can be used.
  • Ambion WT-expression kit allows amplification of total RNA directly without a separate ribosomal RNA (rRNA) depletion step.
  • rRNA ribosomal RNA
  • samples as small as 50 ng of total RNA can be analyzed on AffymetrixTM. GeneChip® Human, Mouse, and Rat Exon and Gene 1.0 ST Arrays.
  • the AmbionTM. WT Expression Kit provides a significant increase in sensitivity. For example, a greater number of probe sets detected above background can be obtained at the exon level with the AmbionTM.
  • AmbionTM-expression kit may be used in combination with additional Affymetrix labeling kit.
  • AmpTec Trinucleotide Nano mRNA Amplification kit (6299-A15) can be used in the subject methods.
  • the ExpressArtTM TRinucleotide mRNA amplification Nano kit is suitable for a wide range, from 1 ng to 700 ng of input total RNA. According to the amount of input total RNA and the required yields of aRNA, it can be used for 1-round (input >300 ng total RNA) or 2-rounds (minimal input amount 1 ng total RNA), with aRNA yields in the range of >10 ⁇ g.
  • AmpTec's proprietary TRinucleotide priming technology results in preferential amplification of mRNAs (independent of the universal eukaryotic 3′-poly(A)-sequence), combined with selection against rRNAs. More information on AmpTec Trinucleotide Nano mRNA Amplification kit can be obtained at www.amp-tec.com/products.htm. This kit can be used in combination with cDNA conversion kit and Affymetrix labeling kit.
  • the raw data may then be normalized, for example, by subtracting the background intensity and then dividing the intensities making either the total intensity of the features on each channel equal or the intensities of a reference gene and then the t-value for all the intensities may be calculated. More sophisticated methods, include z-ratio, loess and lowess regression and RMA (robust multichip analysis), such as for Affymetrix chips.
  • the above described methods may be used for determining transcript expression levels for training (e.g., using a classifier training module) a classifier to differentiate whether a subject is a smoker or non-smoker. In some embodiments, the above described methods may be used for determining transcript expression levels for training (e.g., using a classifier training module) a classifier to differentiate whether a subject has UIP or non-UIP.
  • results of molecular profiling performed on a sample from a subject may be compared to a biological sample that is known or suspected to be normal (“normal sample”).
  • a normal sample is a sample that does not comprise or is expected to not comprise an ILD, or conditions under evaluation, or would test negative in the molecular profiling assay for the one or more ILDs under evaluation.
  • a normal sample is that which is or is expected to be free of any ILD, or a sample that would test negative for any ILD in the molecular profiling assay.
  • the normal sample may be from a different subject from the subject being tested, or from the same subject.
  • the normal sample is a lung tissue sample obtained from a subject such as the subject being tested for example.
  • the normal sample may be assayed at the same time, or at a different time from the test sample.
  • a normal sample is a sample that is known or suspected to be from a non-smoker.
  • the normal sample is a sample that has been confirmed by at least two expert pathologists to be Non-UIP.
  • the normal sample is a sample that has been confirmed by at least two expert pathologists to be Non-IPF.
  • the results of an assay on the test sample may be compared to the results of the same assay on a sample having a known disease state (e.g., normal, affected by a selected ILD (e.g., IPF, NSIP, etc.), smoker, non-smoker).
  • a known disease state e.g., normal, affected by a selected ILD (e.g., IPF, NSIP, etc.), smoker, non-smoker.
  • the results of the assay on the normal sample are from a database, or a reference.
  • the results of the assay on the normal sample are a known or generally accepted value or range of values by those skilled in the art.
  • the comparison is qualitative. In other cases the comparison is quantitative.
  • qualitative or quantitative comparisons may involve but are not limited to one or more of the following: comparing fluorescence values, spot intensities, absorbance values, chemiluminescent signals, histograms, critical threshold values, statistical significance values, gene product expression levels, gene product expression level changes, alternative exon usage, changes in alternative exon usage, protein levels, DNA polymorphisms, copy number variations, indications of the presence or absence of one or more DNA markers or regions, or nucleic acid sequences.
  • the molecular profiling results are evaluated using methods known to the art for correlating gene product expression levels or alternative exon usage with specific phenotypes such as a particular ILD, or normalcy (e.g. disease or condition free).
  • a specified statistical confidence level may be determined in order to provide a diagnostic confidence level. For example, it may be determined that a confidence level of greater than 90% may be a useful predictor of the presence of an ILD or of a smoker or non-smoker status. In other embodiments, more or less stringent confidence levels may be chosen.
  • a confidence level of about or at least about 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99%, 99.5%, or 99.9% may be chosen as a useful phenotypic predictor.
  • the confidence level provided may in some cases be related to the quality of the sample, the quality of the data, the quality of the analysis, the specific methods used, and/or the number of gene expression products analyzed.
  • the specified confidence level for providing a diagnosis may be chosen on the basis of the expected number of false positives or false negatives and/or cost.
  • Methods for choosing parameters for achieving a specified confidence level or for identifying markers with diagnostic power include but are not limited to Receiver Operating Characteristic (ROC) curve analysis, binormal ROC, principal component analysis, partial least squares analysis, singular value decomposition, least absolute shrinkage and selection operator analysis, least angle regression, and the threshold gradient directed regularization method.
  • ROC Receiver Operating Characteristic
  • Raw gene expression level and alternative splicing data may in some cases be improved through the application of methods and/or processes designed to normalize and or improve the reliability of the data.
  • the data analysis requires a computer or other device, machine or apparatus for application of the various methods and/or processes described herein due to the large number of individual data points that are processed.
  • a “machine learning classifier” refers to a computational-based prediction data structure or method, employed for characterizing a gene expression profile. The signals corresponding to certain expression levels, which are obtained by, e.g., microarray-based hybridization assays, are typically subjected to the classifier to classify the expression profile.
  • Supervised learning generally involves “training” a classifier to recognize the distinctions among classes and then “testing” the accuracy of the classifier on an independent test set. For new, unknown samples the classifier can be used to predict the class in which the samples belong. In various embodiments, such training is be achieved, e.g., using a classifier training module.
  • the robust multi-array average (RMA) method may be used to normalize raw data.
  • the RMA method begins by computing background-corrected intensities for each matched cell on a number of microarrays.
  • the background corrected values are restricted to positive values as described by Irizarry et al. Biostatistics 2003 Apr. 4 (2): 249-64.
  • the base-2 logarithm of each background corrected matched-cell intensity is then obtained.
  • the back-ground corrected, log-transformed, matched intensity on each microarray is then normalized using the quantile normalization method in which for each input array and each probe expression value, the array percentile probe value is replaced with the average of all array percentile points, this method is more completely described by Bolstad et al.
  • the normalized data may then be fit to a linear model to obtain an expression measure for each probe on each microarray.
  • Tukey's median polish algorithm (Tukey, J. W., Exploratory Data Analysis. 1977) may then be used to determine the log-scale expression level for the normalized probe set data.
  • feature selection and model estimation may be performed by logistic regression with lasso penalty using glmnet (Friedman J, Hastie T, Tibshirani R. Regularization Paths for Generalized Linear Models via Coordinate Descent. Journal of statistical software 2010; 33(1): 1-22).
  • Raw reads may be aligned using TopHat (Trapnell C, Pachter L, Salzberg S L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 2009; 25(9): 1105-11.).
  • Gene counts may be obtained using HTSeq (Anders S, Pyl P T, Huber W.
  • Confidence intervals may be computed using the pROC package (Robin X, Turck N, Hainard A, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC bioinformatics 2011; 12: 77)
  • data may be filtered to remove data that may be considered suspect.
  • data deriving from microarray probes that have fewer than about 4, 5, 6, 7 or 8 guanosine+cytosine nucleotides may be considered to be unreliable due to their aberrant hybridization propensity or secondary structure issues.
  • data deriving from microarray probes that have more than about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 guanosine+cytosine nucleotides may be considered unreliable due to their aberrant hybridization propensity or secondary structure issues.
  • unreliable probe sets may be selected for exclusion from data analysis by ranking probe-set reliability against a series of reference datasets.
  • RefSeq or Ensembl are considered very high quality reference datasets.
  • Data from probe sets matching RefSeq or Ensembl sequences may in some cases be specifically included in microarray analysis experiments due to their expected high reliability.
  • data from probe-sets matching less reliable reference datasets may be excluded from further analysis, or considered on a case by case basis for inclusion.
  • the Ensembl high throughput cDNA (HTC) and/or mRNA reference datasets may be used to determine the probe-set reliability separately or together. In other cases, probe-set reliability may be ranked.
  • probes and/or probe-sets that match perfectly to all reference datasets such as for example RefSeq, HTC, HTSeq, and mRNA, may be ranked as most reliable (1).
  • probes and/or probe-sets that match two out of three reference datasets may be ranked as next most reliable (2), probes and/or probe-sets that match one out of three reference datasets may be ranked next (3) and probes and/or probe sets that match no reference datasets may be ranked last (4). Probes and or probe-sets may then be included or excluded from analysis based on their ranking.
  • probe-sets may be ranked by the number of base pair mismatches to reference dataset entries. It is understood that there are many methods understood in the art for assessing the reliability of a given probe and/or probe-set for molecular profiling and the methods of the present disclosure encompass any of these methods and combinations thereof.
  • probe-sets may be excluded from analysis if they are not expressed or expressed at an undetectable level (not above background).
  • a probe-set is judged to be expressed above background if for any group:
  • T0 Sqr(GroupSize)(T ⁇ P)/Sqr(Pvar);
  • GroupSize Number of CEL files in the group,
  • T Average of probe scores in probe-set,
  • P Average of Background probes averages of GC content, and
  • Pvar Sum of Background probe variances/(Number of probes in probe-set) 2
  • probe-sets that exhibit no, or low variance may be excluded from further analysis.
  • Low-variance probe-sets are excluded from the analysis via a Chi-Square test.
  • a probe-set is considered to be low-variance if its transformed variance is to the left of the 99 percent confidence interval of the Chi-Squared distribution with (N ⁇ 1) degrees of freedom. (N ⁇ 1)*Probe-set Variance/(Gene Probe-set Variance). about.Chi-Sq(N ⁇ 1) where N is the number of input CEL files, (N ⁇ 1) is the degrees of freedom for the Chi-Squared distribution, and the “probe-set variance for the gene” is the average of probe-set variances across the gene.
  • probe-sets for a given gene or transcript cluster may be excluded from further analysis if they contain less than a minimum number of probes that pass through the previously described filter steps for GC content, reliability, variance and the like.
  • probe-sets for a given gene or transcript cluster may be excluded from further analysis if they contain less than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or less than about 20 probes.
  • Methods of data analysis of gene expression levels or of alternative splicing may further include the use of a feature selection method and/or process as provided herein.
  • feature selection is provided by use of the LIMMA software package (Smyth, G. K. (2005). Limma: linear models for microarray data. In: Bioinformatics and Computational Biology Solutions using R and Bioconductor, R. Gentleman, V. Carey, S. Dudoit, R. Irizarry, W. Huber (eds.), Springer, New York, pages 397-420).
  • Methods of data analysis of gene expression levels and or of alternative splicing may further include the use of a pre-classifier method and/or process (e.g., implemented by a pre-classifier analysis module).
  • a method and/or process may use a cell-specific molecular fingerprint to pre-classify the samples according to their composition and then apply a correction/normalization factor. This data/information may then be fed in to a final classification method and/or process which would incorporate that information to aid in the final diagnosis.
  • the methods of the present invention include the use of a pre-classifier method and/or process (e.g., implemented by a pre-classifier analysis module) that uses a molecular fingerprint to pre-classify the samples as smoker or non-smoker prior to application of a UIP/non-UIP classifier of the present invention.
  • a pre-classifier method and/or process e.g., implemented by a pre-classifier analysis module
  • Methods of data analysis of gene expression levels and/or of alternative splicing may further include the use of a classifier method and/or process (e.g., implemented by a classifier analysis module) as provided herein.
  • a classifier method and/or process e.g., implemented by a classifier analysis module
  • a diagonal linear discriminant analysis, k-nearest neighbor classifier, support vector machine (SVM) classifier, linear support vector machine, random forest classifier, or a probabilistic model-based method or a combination thereof is provided for classification of microarray data.
  • identified markers that distinguish samples e.g. first ILD from second ILD, normal vs. ILD
  • distinguish subtypes e.g. IPF vs. NSIP
  • FDR Benjamin Hochberg or another correction for false discovery rate
  • the classifier may be supplemented with a meta-analysis approach such as that described by Fishel and Kaufman et al. 2007 Bioinformatics 23(13): 1599-606.
  • the classifier may be supplemented with a meta-analysis approach such as a repeatability analysis.
  • the repeatability analysis selects markers that appear in at least one predictive expression product marker set.
  • the posterior probabilities may be used to rank the markers provided by the classifier.
  • markers may be ranked according to their posterior probabilities and those that pass a chosen threshold may be chosen as markers whose differential expression is indicative of or diagnostic for samples that are for example IPF or NSIP.
  • Illustrative threshold values include prior probabilities of 0.7, 0.75, 0.8, 0.85, 0.9, 0.925, 0.95, 0.975, 0.98, 0.985, 0.99, 0.995 or higher.
  • a statistical evaluation of the results of the molecular profiling may provide, but is not limited to providing, a quantitative value or values indicative of one or more of the following: the likelihood of diagnostic accuracy; the likelihood of an ILD; the likelihood of a particular ILD; the likelihood of the success of a particular therapeutic intervention, the likelihood the subject is a smoker, and the likelihood the subject is a non-smoker.
  • a physician who is not likely to be trained in genetics or molecular biology, need not understand the raw data. Rather, the data is presented directly to the physician in its most useful form to guide patient care.
  • results of the molecular profiling can be statistically evaluated using a number of methods known to the art including, but not limited to: the students T test, the two sided T test, pearson rank sum analysis, hidden markov model analysis, analysis of q-q plots, principal component analysis, one way ANOVA, two way ANOVA, LIMMA and the like.
  • the use of molecular profiling alone or in combination with cytological analysis may provide a classification, identification, or diagnosis that is between about 85% accurate and about 99% or about 100% accurate.
  • the molecular profiling process and/or cytology provide a classification, identification, diagnosis of an ILD that is about, or at least about 85%, 86%, 87%, 88%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 97.5%, 98%, 98.5%, 99%, 99.5%, 99.75%, 99.8%, 99.85%, or 99.9% accurate.
  • the molecular profiling process and/or cytology provide a classification, identification, or diagnosis of the presence of a particular ILD type (e.g.
  • IPF IPF; NSIP; HP
  • IPF NSIP; HP
  • NSIP NSIP
  • HP 85%, 86%, 87%, 88%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 97.5%, 98%, 98.5%, 99%, 99.5%, 99.75%, 99.8%, 99.85%, or 99.9% accurate.
  • accuracy may be determined by tracking the subject over time to determine the accuracy of the original diagnosis. In other cases, accuracy may be established in a deterministic manner or using statistical methods. For example, receiver operator characteristic (ROC) analysis may be used to determine the optimal assay parameters to achieve a specific level of accuracy, specificity, positive predictive value, negative predictive value, and/or false discovery rate.
  • ROC receiver operator characteristic
  • gene expression products and compositions of nucleotides encoding for such products which are determined to exhibit the greatest difference in expression level or the greatest difference in alternative splicing between a first ILD and a second ILD (e.g., between IPF and NSIP), between ILD and normal, and/or between smoker and non-smoker may be chosen for use as molecular profiling reagents of the present disclosure.
  • Such gene expression products may be particularly useful by providing a wider dynamic range, greater signal to noise, improved diagnostic power, lower likelihood of false positives or false negative, or a greater statistical confidence level than other methods known or used in the art.
  • the use of molecular profiling alone or in combination with cytological analysis may reduce the number of samples scored as non-diagnostic by about, or at least about 100%, 99%, 95%, 90%, 80%, 75%, 70%, 65%, or about 60% when compared to the use of standard cytological techniques known to the art.
  • the methods of the present invention may reduce the number of samples scored as intermediate or suspicious by about, or at least about 100%, 99%, 98%, 97%, 95%, 90%, 85%, 80%, 75%, 70%, 65%, or about 60%, when compared to the standard cytological methods used in the art.
  • results of the molecular profiling assays are entered into a database for access by representatives or agents of a molecular profiling business, the individual, a medical provider, or insurance provider.
  • assay results include sample classification, identification, or diagnosis by a representative, agent or consultant of the business, such as a medical professional.
  • a computer analysis of the data is provided automatically.
  • the molecular profiling business may bill the individual, insurance provider, medical provider, researcher, or government entity for one or more of the following: molecular profiling assays performed, consulting services, data analysis, reporting of results, or database access.
  • the results of the molecular profiling are presented as a report on a computer screen or as a paper record.
  • the report may include, but is not limited to, such information as one or more of the following: the number of genes differentially expressed, the suitability of the original sample, the number of genes showing differential alternative splicing, a diagnosis, a statistical confidence for the diagnosis, the likelihood the subject is a smoker, the likelihood of an ILD, and indicated therapies.
  • the results of the molecular profiling may be classified into one of the following: smoker, non-smoker, ILD, a particular type of ILD, a non-ILD, or non-diagnostic (providing inadequate information concerning the presence or absence of an ILD).
  • the results of the molecular profiling may be classified into IPF versus NSIP categories.
  • the results may be classified as UIP or non-UIP.
  • results are classified using a trained classifier.
  • Trained classifiers of the present invention implement methods and/or processes that have been developed using a reference set of known ILD and normal samples, known smoker and non-smoker samples, or combinations of known ILD and normal samples from smokers and/or non-smokers including, but not limited to, samples with one or more histopathologies.
  • training e.g., using a classifier training module
  • training comprises comparison of gene expression product levels in a first set biomarkers from a first ILD that is non-UIP to gene expression product levels in a second set of biomarkers from a second ILD that is UIP, where the first set of biomarkers includes at least one biomarker that is not in the second set.
  • training further comprises comparison of gene expression product levels in a first set biomarkers from a first subject that is a smoker to gene expression product levels in a second set of biomarkers from a second subject that is a non-smoker, where the first set of biomarkers includes at least one biomarker that is not in the second set.
  • either the entire classifier or portions of the classifier can be trained (e.g., using a classifier training module) using comparisons of expression levels of biomarker panels within a classification panel against all other biomarker panels (or all other biomarker signatures) used in the classifier.
  • Classifiers suitable for categorization of samples include but are not limited to k-nearest neighbor classifiers, support vector machines, linear discriminant analysis, diagonal linear discriminant analysis, updown, naive Bayesian classifiers, neural network classifiers, hidden Markov model classifiers, genetic classifiers, or any combination thereof.
  • trained classifiers of the present invention may incorporate data other than gene expression or alternative splicing data such as but not limited to DNA polymorphism data, sequencing data, scoring or diagnosis by cytologists or pathologists of the present invention, information provided by the pre-classifier method and/or process of the present disclosure, or information about the medical history of the subject of the present disclosure.
  • n is a negative classifier output, such as no ILD, or absence of a particular disease tissue as described herein
  • false negative is when the prediction outcome is n while the actual value is p.
  • a diagnostic test that seeks to determine whether a person has a certain disease. A false positive in this case occurs when the person tests positive, but actually does not have the disease. A false negative, on the other hand, occurs when the person tests negative, suggesting they are healthy, when they actually do have the disease.
  • a Receiver Operator Characteristic (ROC) curve assuming real-world prevalence of subtypes can be generated by re-sampling errors achieved on available samples in relevant proportions.
  • the positive predictive value is the proportion of patients with positive test results who are correctly diagnosed. It is the most important measure of a diagnostic method as it reflects the probability that a positive test reflects the underlying condition being tested for. Its value does however depend on the prevalence of the disease, which may vary. In one example, FP (false positive) TN (true negative); TP (true positive); FN (false negative).
  • False positive rate ( ⁇ ) FP/(FP+TN)—specificity
  • False negative rate (p) FN/(TP+FN)—sensitivity
  • Likelihood-ratio positive sensitivity/(1 ⁇ specificity)
  • Likelihood-ratio negative (1 ⁇ sensitivity)/specificity.
  • the negative predictive value is the proportion of patients with negative test results who are correctly diagnosed.
  • PPV and NPV measurements can be derived using appropriate disease subtype prevalence estimates.
  • An estimate of the pooled disease prevalence can be calculated from the pool of indeterminates which roughly classify into B vs M by surgery.
  • disease prevalence may sometimes be incalculable because there are not any available samples. In these cases, the subtype disease prevalence can be substituted by the pooled disease prevalence estimate.
  • the level of expression products or alternative exon usage is indicative of one or the following: IPF, NSIP, or HP.
  • the level of expression products or alternative exon usage is indicative that the subject is a smoker or a non-smoker.
  • the results of the expression analysis of the subject methods provide a statistical confidence level that a given diagnosis is correct.
  • such statistical confidence level is at least about, or more than about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% 99.5%, or more.
  • a subject method and/or system may include generating a report that provides an indication that a sample (a lung tissue sample) is an ILD sample (e.g., using a report module).
  • a subject diagnostic method can include generating a report that provides an indication as to whether an individual being tested has an ILD.
  • a subject diagnostic method can include generating a report that provides an indication as to whether an individual being tested is, or is not a smoker.
  • a subject method (or report module) can include generating a report that provides an indication as to whether an individual being tested has IPF (and not, e.g., an ILD other than IPF; e.g., the report can indicate that the individual has IPF and not NSIP).
  • a subject method of diagnosing an ILD involves generating a report (e.g., using a report module).
  • a report can include information such as a likelihood that the patient has an ILD; a likelihood that the patient is a smoker; a recommendation regarding further evaluation; a recommendation regarding therapeutic drug and/or device intervention; and the like.
  • the methods disclosed herein can further include a step of generating or outputting a report providing the results of a subject diagnostic method, which report can be provided in the form of an electronic medium (e.g., an electronic display on a computer monitor), or in the form of a tangible medium (e.g., a report printed on paper or other tangible medium).
  • An assessment as to the results of a subject diagnostic method e.g., a likelihood that an individual has an ILD; a likelihood that an individual has IPF; a likelihood that an individual is a smoker
  • a person or entity that prepares a report (“report generator”) may also perform steps such as sample gathering, sample processing, and the like.
  • a diagnostic assessment report can be provided to a user.
  • a “user” can be a health professional (e.g., a clinician, a laboratory technician, a physician (e.g., a cardiologist), etc.).
  • a subject report can further include one or more of: 1) service provider information; 2) patient data; 3) data regarding the expression level of a given gene product or set of gene products, a score or classifier decision; 4) follow-up evaluation recommendations; 5) therapeutic intervention or recommendations; and 6) other features.
  • a physician or other qualified medical personnel can determine whether further evaluation of the test subject (the patient) is required. Further evaluation can include, e.g., spirometry.
  • a physician or other qualified medical personnel can determine whether appropriate therapeutic intervention is advised.
  • Therapeutic intervention includes drug-based therapeutic intervention, device-based therapeutic intervention, and surgical intervention. Where a report indicates a likelihood that an individual has IPF, drug-based therapeutic intervention includes, e.g., administering to the individual an effective amount of pirfenidone, prednisone, azathioprine, or N-acetylcysteine. Surgical intervention includes, e.g., arterial bypass surgery.
  • the methods of the present disclosure can be computer-implemented, such that method steps (e.g., assaying, comparing, calculating, and the like) are be automated in whole or in part.
  • the present disclosure provides methods, computer systems, devices and the like in connection with computer-implemented methods of facilitating a diagnosis of an interstitial lung disease (e.g., a diagnosis of IPF, NSIP, HP, etc.), including differential diagnosis.
  • an interstitial lung disease e.g., a diagnosis of IPF, NSIP, HP, etc.
  • the present disclosure further provides methods, computer systems, devices and the like in connection with computer-implemented methods of facilitating determination of smoker status (e.g., smoker vs. non-smoker).
  • the present disclosure further provides methods, computer systems, devices and the like in connection with computer-implemented methods of facilitating a diagnosis of an interstitial lung disease (e.g., a diagnosis of IPF, NSIP, HP, etc.), including differential diagnosis, wherein the methods further comprise determining a subjects smoker status (smoker vs. non-smoker) and incorporating smoker status into the determination of the subjects interstitial lung disease diagnosis.
  • smoker status is incorporated into the interstitial lung disease diagnosis as a covariate in the model used during training (e.g., using a classifier training module).
  • (ii) smoker status is incorporated into the interstitial lung disease diagnosis by identifying one or more genes that are susceptible to smoker status bias and excluding such genes or weighing such genes differently than other genes that are not susceptible to smoker-status during interstitial lung disease diagnosis classifier training.
  • (iii) smoker status is incorporated into the interstitial lung disease diagnosis by constructing a tiered classification in which an initial classifier is trained to recognize the gene signatures that distinguish smokers from non-smokers (e.g., using a classifier training module).
  • Such methods comprising the step of incorporating smoker status into the determination of the subjects interstitial lung disease diagnosis include a combination of one or more of the above mentioned means of such incorporation (i.e., a combination of two or more of embodiments (i) to (iii) in the instant paragraph.
  • the method steps including obtaining values for biomarker levels, comparing normalized biomarker (gene) expression levels to a control level, calculating the likelihood of an ILD (and optionally the likelihood a subject is a smoker), generating a report, and the like, can be completely or partially performed by a computer program product.
  • Values obtained can be stored electronically, e.g., in a database, and can be subjected to a classifier executed by a programmed computer (e.g., using a classifier analysis module).
  • the methods and/or systems of the present disclosure can involve inputting a biomarker level (e.g., a normalized expression level of a gene product) into a classifier analysis module to execute a method and/or process to perform the comparing and calculating step(s) described herein, and generate a report (e.g., using a report module) as described herein, e.g., by displaying or printing a report to an output device at a location local or remote to the computer.
  • a biomarker level e.g., a normalized expression level of a gene product
  • a report module e.g., by displaying or printing a report to an output device at a location local or remote to the computer.
  • the output to the report can be a score (e.g., numerical score (representative of a numerical value) or a non-numerical score (e.g., non-numerical output (e.g., “IPF”, “No evidence of IPF”) representative of a numerical value or range of numerical values.
  • the output may indicate “UIP” vs. “non-UIP.”
  • the output may indicate “Smoker” vs. “Non-smoker”
  • the present disclosure thus provides a computer program product including a computer readable storage medium having software and/or hardware modules stored on it.
  • the software and/or hardware modules can, when executed by a processor, execute relevant calculations based on values obtained from analysis of one or more biological sample (e.g., lung tissue sample) from an individual.
  • the computer program product has stored therein a computer program for performing the calculation(s).
  • the present disclosure provides systems for executing the program described above, which system generally includes: a) a central computing environment or processor executing software and/or hardware modules; b) an input device, operatively connected to the computing environment, to receive patient data, wherein the patient data can include, for example, biomarker level or other value obtained from an assay using a biological sample from the patient, as described above; c) an output device, connected to the computing environment, to provide information to a user (e.g., medical personnel); and d) a method and/or process executed by the central computing environment (e.g., a processor), where the method and/or process is executed based on the data received by the input device, and wherein the method and/or process calculates a value, which value is indicative of the likelihood the subject has an ILD, as described herein.
  • a central computing environment or processor executing software and/or hardware modules
  • an input device operatively connected to the computing environment, to receive patient data, wherein the patient data can include, for example, biomarker
  • the present disclosure also provides systems for executing the program described above, which system generally includes: a) a central computing environment or processor executing software and/or hardware modules; b) an input device, operatively connected to the computing environment, to receive patient data, wherein the patient data can include, for example, biomarker level or other value obtained from an assay using a biological sample from the patient, as described above; c) an output device, connected to the computing environment, to provide information to a user (e.g., medical personnel); and d) a method and/or process executed by the central computing environment (e.g., a processor), where the method and/or process is executed based on the data received by the input device, wherein the method and/or process calculates a value, which value is indicative of the likelihood the subject has an ILD, as described herein, and wherein the method and/or process uses smoking status (smoker vs.
  • a central computing environment or processor executing software and/or hardware modules
  • an input device operatively connected to the computing
  • the method and/or process excludes or weighs one or more gene that is susceptible to smoker status bias differently during classifier training to enrich the feature space used for training with genes that are not confounded or affected by smoking status.
  • the present disclosure provides systems for executing the program described above, which system generally includes: a) a central computing environment or processor executing software and/or hardware modules; b) an input device, operatively connected to the computing environment, to receive patient data, wherein the patient data can include, for example, biomarker level or other value obtained from an assay using a biological sample from the patient, as described above; c) an output device, connected to the computing environment, to provide information to a user (e.g., medical personnel); and d) a first method and/or process executed by the central computing environment (e.g., a processor), where the first method and/or process is executed based on the data received by the input device, wherein the first method and/or process calculates a value, which value is indicative of the likelihood a subject is a smoker or a non-smoker, as described herein, wherein the subject's status as a smoker or non-smoker causes the first method and/or process to apply a second method and
  • Non UIP in smokers or non-smokers respectively and e) wherein the second method and/or process is executed by the central computing environment (e.g., a processor), where the second method and/or process is executed based on the data received by the input device, and wherein the second method and/or process calculates a value, which value is indicative of the likelihood the subject has an ILD, as described herein,
  • the central computing environment e.g., a processor
  • FIG. 7A illustrates a processing system 100 including at least one processor 102 , or processing unit or plurality of processors, memory 104 , at least one input device 106 and at least one output device 108 , coupled together via a bus or group of buses 110 .
  • Processing system can be implemented on any suitable device, such as, for example, a host device, a personal computer, a handheld or laptop device, a personal digital assistant, a multiprocessor system, a microprocessor-based system, a programmable consumer electronic device, a minicomputer, a server computer, a web server computer, a mainframe computer, and/or a distributed computing environment that includes any of the above systems or devices
  • input device 106 and output device 108 can be the same device.
  • An interface 112 can also be provided for coupling the processing system 100 to one or more peripheral devices, for example interface 112 can be a PCI card or PC card.
  • At least one storage device 114 which houses at least one database 116 can also be provided.
  • the memory 104 can be any form of memory device, for example, volatile or nonvolatile memory, solid state storage devices, magnetic devices, etc.
  • the memory 104 can be a random access memory (RAM), a memory buffer, a hard drive, a read-only memory (ROM), an erasable programmable read-only memory (EPROM), a database, and/or the like.
  • the processor 102 can include more than one distinct processing device, for example to handle different functions within the processing system 100 .
  • the processor 100 can be any suitable processing device configured to run or execute a set of instructions or code (e.g., stored in the memory) such as a general-purpose processor (GPP), a central processing unit (CPU), an accelerated processing unit (APU), a graphics processor unit (GPU), an Application Specific Integrated Circuit (ASIC), and/or the like.
  • a processor 100 can run or execute a set of instructions or code stored in the memory associated with using a personal computer application, a mobile application, an internet web browser, a cellular and/or wireless communication (via a network), and/or the like. More specifically, the processor can execute a set of instructions or code stored in the memory 104 associated with analyzing and classifying data, as described herein.
  • Input device 106 receives input data 118 and can comprise, for example, a keyboard, a pointer device such as a pen-like device or a mouse, audio receiving device for voice controlled activation such as a microphone, data receiver or antenna such as a modem or wireless data adaptor, data acquisition card, etc.
  • Input data 118 can come from different sources, for example keyboard instructions in conjunction with data received via a network.
  • Output device 108 produces or generates output data 120 and can comprise, for example, a display device or monitor in which case output data 120 is visual, a printer in which case output data 120 is printed, a port for example a USB port, a peripheral component adaptor, a data transmitter or antenna such as a modem or wireless network adaptor, etc.
  • Output data 120 can be distinct and derived from different output devices, for example a visual display on a monitor in conjunction with data transmitted to a network. A user can view data output, or an interpretation of the data output, on, for example, a monitor or using a printer.
  • the input device 106 and/or the output device 108 can be a communication interface configured to send and/or receive data via a network. More specifically, in such embodiments, the processing system 100 can act as a host device to one or more client devices (not shown in FIG. 7A ). As such, the processing system 100 can send data to (e.g., output data 120 ) and receive data from (e.g., input data 118 ) the client devices.
  • client devices e.g., input data 118
  • Such a communication interface can be any suitable module and/or device that can place the processing system 100 in communication with a client device such as one or more network interface cards or the like.
  • Such a network interface card can include, for example, an Ethernet port, a WiFi® radio, a Bluetooth® radio, a near field communication (NFC) radio, and/or a cellular radio that can place the client device 150 in communication with the host device 110 via a network or the like.
  • a network interface card can include, for example, an Ethernet port, a WiFi® radio, a Bluetooth® radio, a near field communication (NFC) radio, and/or a cellular radio that can place the client device 150 in communication with the host device 110 via a network or the like.
  • the storage device 114 can be any form of data or information storage means, for example, volatile or non-volatile memory, solid state storage devices, magnetic devices, etc.
  • the storage device 114 can be a random access memory (RAM), a memory buffer, a hard drive, a read-only memory (ROM), an erasable programmable read-only memory (EPROM), a database, and/or the like.
  • the processing system 100 is adapted to allow data or information to be stored in and/or retrieved from, via wired or wireless communication means, at least one database 116 .
  • the interface 112 may allow wired and/or wireless communication between the processing unit 102 and peripheral components that may serve a specialized purpose.
  • the processor 102 can receive instructions as input data 118 via input device 106 and can display processed results or other output to a user by utilizing output device 108 . More than one input device 106 and/or output device 108 can be provided.
  • the processing system 100 may be any suitable form of terminal, server, specialized hardware, or the like.
  • the processing system 100 may be a part of a networked communications system.
  • Processing system 100 can connect to a network, for example, a local area network (LAN), a virtual network such as a virtual local area network (VLAN), a wide area network (WAN), a metropolitan area network (MAN), a worldwide interoperability for microwave access network (WiMAX), a cellular network, the Internet, and/or any other suitable network implemented as a wired and/or wireless network.
  • LAN local area network
  • VLAN virtual local area network
  • WAN wide area network
  • MAN metropolitan area network
  • WiMAX worldwide interoperability for microwave access network
  • the modem which may be internal or external, may be connected to a system bus via a user input interface, or via another appropriate mechanism.
  • program modules depicted relative to the computing system environment 100 may be stored in a remote memory storage device. It is to be appreciated that the illustrated network connections of FIG. 7 are examples and other means of establishing a communications link between multiple computers may be used.
  • Input data 118 and output data 120 can be communicated to other devices via the network.
  • the transfer of information and/or data over the network can be achieved using wired communications means or wireless communications means.
  • a server can facilitate the transfer of data between the network and one or more databases.
  • a server and one or more databases provide an example of an information source.
  • the processing computing system environment 100 illustrated in FIG. 7A may operate in a networked environment using logical connections to one or more remote computers.
  • the remote computer may be a personal computer, a server, a router, a network PC, a peer device, or other common network node, and typically includes many or all of the elements described above.
  • FIG. 7B illustrates the processor 102 of FIG. 7A in greater detail.
  • the processor 102 can be configured to execute specific modules.
  • the modules can be, for example, hardware modules, software modules stored in the memory 104 and/or executed in the processor 102 , and/or any combination thereof.
  • the processor 102 includes and/or executes a pre-classifier analysis module 130 , a classifier training module 132 , a classifier analysis module 134 and a report module 136 .
  • the pre-classifier analysis module 130 , the classifier training module 132 , the classifier analysis module 134 and the report module 136 can be connected and/or electrically coupled. As such, signals can be sent between the pre-classifier analysis module 130 , the classifier training module 132 , the classifier analysis module 134 and the report module 136 .
  • the classifier training module 132 can be configured to receive a corpora of data (e.g. gene expression data, sequencing data) and train a classifier.
  • a corpora of data e.g. gene expression data, sequencing data
  • clinical annotation data from samples previously identified as UIP and non-UIP e.g., by an expert
  • expert TBB histopathology labels i.e., UIP or Non UIP
  • expert HRCT labels i.e., HRCT labels
  • expert patient-level clinical outcome labels can be obtained and used alone or in combination to train the classifier using microarray and/or sequencing data.
  • the feature space used can include gene expression, variants, mutations, fusions, loss of heterozygoxity (LOH), biological pathway effect and/or any other dimension of the data that can be extracted as a feature for the purposes of training a machine-learning algorithm.
  • the feature space used for training a UIP vs. Non-UIP classifier, a smoker vs. Non-smoker classifier, or a UIP vs. Non-UIP and smoker vs. Non-smoker classifier includes gene expression, variants, mutations, fusions, loss of heterozygoxity (LOH), and biological pathway effect.
  • the feature space used for training a UIP vs. Non-UIP classifier, a smoker vs. Non-smoker classifier, or a UIP vs. Non-UIP and smoker vs. Non-smoker classifier includes gene expression and variant dimensions.
  • the classifier training module 132 can train a smoker classifier and a non-smoker classifier based on an indication associated with whether a received sample is associated with a smoker or non-smoker.
  • the smoker/non-smoker can be used as an attribute (a model covariate) to train a single classifier. After the classifier is trained, it can be used to identify and/or classify newly received and unknown samples as described herein.
  • the pre-classifier analysis module 130 can identify whether a sample is associated with a smoker or a non-smoker. Specifically, the pre-classifier analysis module 130 can use any suitable method to identify and/or classify a sample as coming from an individual that smokes (or has a past history of heavy smoking) versus an individual that does not smoke (or has no smoking history). The classification can be done in any suitable manner such as, receiving an indication from a user, identification of genes that are susceptible to smoker-status bias, using a machine-learning classifier, and/or any other suitable method described herein.
  • the classifier analysis module 134 can input the sample into the classifier to identify and/or classify the received sample as associated with UIP and non-UIP. Specifically, the classifier analysis module 134 can use a trained classifier to identify whether the sample indicates UIP or non-UIP. In some embodiments, the classifier analysis module 134 can indicate a percentage or confidence score of the sample being associated with UIP or non-UIP. In some embodiments, the classifier analysis module 134 can execute two separate classifiers: one for smoker samples and the other for non-smoker samples (as determined by the pre-classifier analysis module 130 ). In other embodiments, a single classifier is executed for both smoker and non-smoker samples with an input for smoker status.
  • the report module 136 can be configured to generate any suitable report based on the outcome of the classifier analysis module 134 as described in further detail herein.
  • the report may include, but is not limited to, such information as one or more of the following: the number of genes differentially expressed, the suitability of the original sample, the number of genes showing differential alternative splicing, a diagnosis, a statistical confidence for the diagnosis, the likelihood the subject is a smoker, the likelihood of an ILD, and indicated therapies.
  • FIG. 7C illustrates a flow chart of one non-limiting embodiment of the present invention wherein gene product expression data for known UIP and non-UIP samples are used to train (e.g., using a classifier training module) a classifier for differentiating UIP vs. non-UIP, wherein the classifier optionally considers smoker status as a covariant, and wherein gene product expression data from unknown samples are input into the trained classifier to identify the unknown samples as either UIP or non-UIP, and wherein the results of the classification via the classifier are defined and output via a report.
  • a classifier training module e.g., a classifier training module
  • Embodiments may be implemented with numerous other general-purpose or special-purpose computing devices and computing system environments or configurations.
  • Examples of other computing systems, environments, and configurations that may be suitable for use with an embodiment include, but are not limited to, personal computers, handheld or laptop devices, personal digital assistants, multiprocessor systems, microprocessor-based systems, programmable consumer electronics, network, minicomputers, server computers, web server computers, mainframe computers, and distributed computing environments that include any of the above systems or devices.
  • Embodiments may be described in a general context of computer-executable instructions, such as hardware and/or software modules. An embodiment may also be practiced in a distributed computing environment where tasks are performed by remote processing devices that are linked through a communications network. In a distributed computing environment, program modules may be located in both local and remote computer storage media including memory storage devices.
  • the present disclosure provides computer program products that, when executed on a programmable computer such as that described above with reference to FIG. 7 , can carry out the methods of the present disclosure.
  • the subject matter described herein may be embodied in systems, apparatus, methods, and/or articles depending on the desired configuration.
  • These various implementations may include implementation in one or more computer programs that are executable and/or interpretable on a programmable system including at least one programmable processor, which may be special or general purpose, coupled to receive data and instructions from, and to transmit data and instructions to, a storage system, at least one input device (e.g. video camera, microphone, joystick, keyboard, and/or mouse), and at least one output device (e.g. display monitor, printer, etc.).
  • at least one input device e.g. video camera, microphone, joystick, keyboard, and/or mouse
  • at least one output device e.g. display monitor, printer, etc.
  • Computer programs include instructions for a programmable processor, and may be implemented in a high-level procedural and/or object-oriented programming language, and/or in assembly/machine language.
  • machine-readable medium refers to any computer program product, apparatus and/or device (e.g., magnetic discs, optical disks, memory, etc.) used to provide machine instructions and/or data to a programmable processor, including a machine-readable medium that receives machine instructions as a machine-readable signal.
  • processors such as a microprocessor, executing sequences of instructions stored in memory or other computer-readable medium including any type of ROM, RAM, cache memory, network memory, floppy disks, hard drive disk (HDD), solid-state devices (SSD), optical disk, CD-ROM, and magnetic-optical disk, EPROMs, EEPROMs, flash memory, or any other type of media suitable for storing instructions in electronic format.
  • processors such as a microprocessor, executing sequences of instructions stored in memory or other computer-readable medium including any type of ROM, RAM, cache memory, network memory, floppy disks, hard drive disk (HDD), solid-state devices (SSD), optical disk, CD-ROM, and magnetic-optical disk, EPROMs, EEPROMs, flash memory, or any other type of media suitable for storing instructions in electronic format.
  • processor(s) may be, or may include, one or more programmable general-purpose or special-purpose microprocessors, digital signal processors (DSPs), programmable controllers, application specific integrated circuits (ASICs), programmable logic devices (PLDs), trusted platform modules (TPMs), or the like, or a combination of such devices.
  • DSPs digital signal processors
  • ASICs application specific integrated circuits
  • PLDs programmable logic devices
  • TPMs trusted platform modules
  • special-purpose hardware such as logic circuits or other hardwired circuitry may be used in combination with software instructions to implement the techniques described herein.
  • a subject array can comprise a plurality of nucleic acids, each of which hybridizes to a gene differentially expressed in a cell present in a tissue sample obtained from an individual being tested for an ILD.
  • a subject array can comprise a plurality of nucleic acids, each of which hybridizes to a gene differentially expressed in a cell present in a tissue sample obtained from an individual being tested for smoker status.
  • a subject array can comprise a plurality of nucleic acids, each of which hybridizes to a gene differentially expressed in a cell present in a tissue sample obtained from an individual being tested for both smoker status and an ILD.
  • a subject array can comprise a plurality of member nucleic acids, each of which member nucleic acids hybridizes to a different gene product. In some cases, two or more member nucleic acids hybridize to the same gene product; e.g., in some cases 2, 3, 4, 5, 6, 7, 8, 9, 10, or more member nucleic acids hybridize to the same gene product.
  • a member nucleic acid can have a length of from about 5 nucleotides (nt) to about 100 nt, e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 18, 19, 20, 20-25, 25-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, or 90-100 nt.
  • a nucleic acid can have one or more phosphate backbone modifications.
  • a subject array can include from about 10 to about 10 5 unique member nucleic acids, or more than 10 5 unique member nucleic acids.
  • a subject array can include from about 10 to about 10 2 , from about 10 2 to about 10 3 , from about 10 to about 10 4 , from about 10 4 to about 10 5 , or more than 10 5 , unique member nucleic acids.
  • adj.P.Value.edgeR False discovery rate adjusted p value of RNAseq gene expression data using edgeR analysis.
  • adj.P.Value.microarray False discovery rate adjusted p value of RNAseq gene expression data using microarray analysis
  • adj.P.Value.npSeq False discovery rate adjusted p value of RNAseq gene expression data using npSeq analysis
  • BRONCH Broncholitis
  • Classified edgeR an R package for the significance analysis of sequencing data
  • Ensembl ID Gene Identifier from Ensembl Genome Browser database
  • FDR False Discovery Rate, an adjusted p value that limits the possibility that the results are random due to the large number of genes simultaneously evaluated.
  • Gene Symbol Gene Identifier from HUGO Gene Nomenclature Committee logFC.edgeR: Log2 fold change of RNAseq gene expression data using edgeR analysis logFC.microarray: Log2 fold change of RNAseq gene expression data using LIMMA microarray analysis logFC.npSeq: Log2 fold change of RNAseq gene expression data using npSeq analysis microarray: Gene expression analysis using gene arrays such as from Affymetrix.
  • NML Normal Lung, usually obtained from human lung donor tissue that was ultimately never transplanted
  • npSeq an R package for the significance analysis of sequencing data
  • NSIP Non Specific Interstitial Pneumonia OP: Organizing Pneumonia
  • P.value.edgeR p value of RNAseq gene expression data using edgeR analysis
  • P.value.microarray p value of RNAseq gene expression data using LIMMA microarray analysis
  • P.value.npSeqp value of RNAseq gene expression data using npSeq analysis
  • RB Respiratory Broncholitis REST: A combination of all other ILDs except the subtype it is being compared to.
  • TCID Squamous Cell Carcinoma
  • TCID Transcript Cluster Identifier
  • TCID numbers thus refer to a gene product(s) of a specific gene, and can be found, e.g., at the following world wide web address: affymetrix.com/the sequences of which probes and gene products are hereby incorporation herein in their entirety.
  • UIP Usual Interstitial Pneumonia; the HRCT or histopathology pattern observed in IPF
  • LIMMA Linear Models for Microarray Data; an R package for the significance analysis of microarray data.
  • ENSEMBL ID refers to a gene identifier number from the Ensembl Genome Browser database (see world wide web address: ensembl.org/index.html, incorporate herein). Each identifier begins with the letters ENSG to denote “Ensembl Gene”. Each ENSEMBL ID number (i.e., each “gene” in the Ensembl database) refers to a gene defined by a specific start and stop position on a particular human chromosome, and therefore defines a specific locus of the human genome. As one of average skill in the art would fully appreciate, all of the gene symbols disclosed herein refer to gene sequences, which are readily available on publically available databases, e.g., UniGene database (Pontius J U, Wagner L, Schuler G D.
  • VATS Video-assisted thoracoscopic surgery
  • IRB Institutional Review Board
  • BRonchial sAmple collection for a noVel gEnomic test sponsored by Veracyte, Inc. (South San Francisco, Calif.). Additional VATS and surgical lung biopsy specimens were obtained from banked sources.
  • a patient can have more than one sample-level diagnosis (i.e. one per VATS sample per patient, most often one from each of the lower and upper lobes of the right lung), but can only have one patient-level diagnosis.
  • sample-level pathology diagnoses were converted into binary class labels (UIP and non-UIP).
  • UIP binary class labels
  • non-UIP pathology diagnosis categories
  • the ‘UIP’ class includes (1) UIP, (2) Classic UIP, (3) Difficult UIP, and (4) CIF/NOC, Favor UIP. All other pathology diagnoses except Non-diagnostic (ND) were assigned to the ‘non-UIP’ class.
  • Tissue-Tek O.C.T. medium Sakura Finetek U.S.A.
  • 2 ⁇ 20 ⁇ m sections generated using a CM1800 cryostat (Leica Biosystems, Buffalo Grove, Ill.).
  • Tissue curls were immediately immersed in RNAprotect (QIAGEN, Valencia, Calif.), incubated overnight at 4° C. and stored at ⁇ 80° C. until extraction.
  • RNAprotect QIAGEN, Valencia, Calif.
  • adjacent 5 ⁇ m tissue curls were mounted onto glass slides and processed for hematoxylin and eosin (H&E) staining following standard procedures.
  • RNA yield and quality was determined using Quant-it (Invitrogen) and Pico BioAnalyzer kits (Agilent). Fifteen nanograms of total RNA were amplified using Ovation FFPE WTA System (NuGEN, San Carlos, Calif.), hybridized to GeneChip Gene ST 1.0 (Affymetrix, Santa Clara, Calif.) microarrays, processed and scanned according to the manufacturer's protocols. Expression data was normalized by Robust Multi-array Average (RMA).
  • RMA Robust Multi-array Average
  • RNA sequencing was performed on select samples at a targeted minimum read depth of 80 million paired-end reads per sample. Briefly, 10 ng of total RNA was amplified using the Ovation RNASeq System v2 (NuGEN, San Carlos, Calif.) and TruSeq (Illumina, San Diego, Calif.) sequencing libraries were prepared and sequenced on an Illumina HiSeq according to manufacturer's instructions. Raw reads were aligned to the hg19 genome assembly using TopHat2. Gene counts were obtained using HTSeq and normalized in Bioconductor using the varianceStabilizingTransformation function in the DESeq2 package. Raw counts and normalized expression levels were obtained for 55,097 transcripts.
  • CIF/NOC samples CIF/NOC samples were not excluded. Only one BRAVE cohort sample was omitted, due to missing central pathology diagnosis.
  • 125 samples (86 patients) were available for microarray classification.
  • the 86 patients were randomized into training and test sets while controlling for patient-level pathology subtype bias (Table 1).
  • the microarray training set consists of 77 samples (39 UIP and 38 non-UIP) from 54 patients.
  • the microarray test set consists of 48 samples (22 UIP vs. 26 non-UIP) from 32 patients.
  • RNASeq data was generated for a subset of 36 samples (17 UIP and 19 non-UIP) from 29 patients (Table 1), representing a spectrum of ILD subtypes.
  • 36 samples 17 UIP and 19 non-UIP
  • 22 overlap with the microarray training set
  • 14 overlap with the microarray test set. Due to the small sample size of this dataset, classification performance was evaluated by cross-validation (CV) only.
  • CV cross-validation
  • Classifier performance was evaluated by CV and, when available, by an independent test set. To minimize over-fitting, a single patient was maintained as the smallest unit when defining the training/test set and the CV partition; i.e. all samples belonging to the same patient were held together as a group in the training/test set or in CV partitions.
  • the CV methods used include leave-one-patient-out (LOPO) and 10-fold patient-level CV.
  • Performance was reported as the area under the curve (AUC), and specificity (1.0—false positive rate) and sensitivity (1.0—false negative rate) at a given score threshold.
  • AUC area under the curve
  • specificity 1.0—false positive rate
  • sensitivity 1.0—false negative rate
  • score threshold was set to require at least >90% specificity.
  • 95% confidence intervals were computed using 2000 stratified bootstrap replicates and the pROC package and reported as [CI lower-upper].
  • Gene expression was evaluated in seven normal and 53 IPF explant lung samples. Genes differentially expressed between normal and IPF patient explant samples were identified and ranked by false discovery rate (FDR) using the R limma package (Smyth, G. K. (2005)). The top 200 genes differentially expressed between UIP and non-UIP classes in the microarray training set are shown in Table 12. Using the top 200 genes with the lowest FDR adjusted P-values ( ⁇ 1.45e-07), the Pearson correlation coefficient was calculated for all pairs of 53 UIP samples.
  • FDR false discovery rate
  • the number and location of the samplings are indicated in FIG. 6 and IPF patient clinical characteristics in Table 4.
  • the results for the three patients diagnosed with IPF are shown in FIG. 1 . Although correlation across all IPF samples is high, three distinct patterns emerge in the correlation structure among IPF samples.
  • One patient (P1) shows substantial differences in upper vs. lower lobe gene expression i.e. lower correlation in gene signals.
  • Pathology at transplant demonstrated end-stage lung disease, diffuse fibrosis, temporal heterogeneity and fibroblastic foci suggestive of UIP.
  • Pre-transplant SLB demonstrated UIP pattern with fibroblastic foci consistent with IPF.
  • Pathology at transplant showed interstitial fibrosis, giant cell reaction, reorganization, bronchiectasis and reactive lymph node.
  • Expression data was normalized by Robust Multi-array Average (RMA). Feature selection and model estimation were performed by logistic regression with lasso penalty using glmnet3. Raw reads were aligned using TopHat. Gene counts were obtained using HTSeq and normalized using DESeq. The top features (N ranging from 10 to 200) were used to train a linear support vector machine (SVM) using the e1071 library. Confidence intervals were computed using the pROC package.
  • RMA Robust Multi-array Average
  • LOPO CV performance is summarized as a receiver operating characteristic (ROC) curve ( FIG. 2A ).
  • the AUC is 0.9 [CI 0.82-0.96], with 92% [CI 84%-100%] specificity and 64% [CI 49%-79%] sensitivity.
  • Individual LOPO CV classification scores are shown for all patients ( FIG. 2B ).
  • the threshold (0-86 and 1.30)
  • the latter sample with the high score was diagnosed as an ‘unclassifiable fibrotic ILD’ at both the sample- and patient-level.
  • UIP samples fifteen (36%) have a score below the threshold (false negatives) but none of those samples have a large negative score. Since LOPO CV in certain cases has the potential to overestimate performance, we also evaluated 10-fold patient-level CV (i.e., 10% of patients are left out in each loop) which gives very similar performance (the median AUC from five repeated 10-fold CVs is 0.88).
  • FIG. 2C Independent test set performance is shown in FIG. 2C showing an AUC of 0.94 [CI 0-86-0-99], with 92% [CI 81%-100%] specificity and 82% [CI 64%-95%] sensitivity.
  • the individual classification score distribution shows good separation between UIP and non-UIP classes ( FIG. 2D ).
  • the two misclassified non-UIP samples have both patient- and sample-level expert diagnoses of ‘unclassifiable fibrotic ILD’ indicating uncertainty in the diagnosis.
  • the score range observed in the test set ( FIG. 2D ) is narrower than the range seen in LOPO CV scores ( FIG. 2B ), likely due to the larger variability inherent in applying a series of sub-classifiers within each CV loop, compared to scores obtained by applying a single model.
  • Classification performance including 95% confidence intervals are summarized in Table 6.
  • RB respiratory bronchiolitis
  • DAD diffuse alveolar damage
  • ORA over-representation analysis
  • a simulation study swapping binary classification labels (UIP or non-UIP) was performed on the microarray training set. Samples were selected at random for label permutation, at total proportions per simulation set ranging from 1% to 40%.
  • Sample labels were changed to the other class with a weight proportional to the probability accounting for the disagreement level in the blinded review of the three expert pathologists: 5% for 3/3 or 2/2 agreement, 50% for 2/3 agreement, and 90% for 1/3 agreement. Simulations were repeated 100 times at each proportion.
  • Interstitial lung diseases are more prevalent in persons that smoke, or have had a long history of smoking prior to quitting, than in persons who never smoked.
  • Transbronchial biopsy samples were prepared [according to the methods described in Examples 1 and 2, and RNA sequencing analysis was performed according to the method described in Example 3].
  • smoking status (smoker vs. non-smoker) is used as a covariate in the model during training.
  • This simple approach boosts signal-to-noise ratio, particularly in data derived from smokers (were noise is higher) and allows data derived from smokers and non-smokers to be combined and used simultaneously.
  • genes that are susceptible to smoker-status bias are identified and excluded, or optionally weighted differently than genes that are not susceptible to such bias, during classifier training. This method enriches the feature space used for training with genes that are not confounded or affected by smoking status.
  • a tiered classification effort is utilized in which an initial classifier is trained to recognize gene signatures that distinguish smokers from non-smokers. Once patient samples are pre-classified as “smoker” or “non-smoker”, distinct classifiers that were each trained to distinguish UIP vs. Non UIP in smokers or non-smokers, respectively, are implemented. Such smoker or non-smoker-specific classifiers provide improved diagnostic performance.
  • Positive log2 fold change value indicates over-expression in UIP relative to Non UIP; negative log2 value indicates under-expression in UIP relative to Non UIP.
  • smoking history status of the patients involved was not evaluated, and the cohort harbored both smokers and non-smokers.
  • Some embodiments described herein relate to a computer storage product with a non-transitory computer-readable medium (also can be referred to as a non-transitory processor-readable medium) having instructions or computer code thereon for performing various computer-implemented operations.
  • the computer-readable medium or processor-readable medium
  • the media and computer code may be those designed and constructed for the specific purpose or purposes.
  • non-transitory computer-readable media include, but are not limited to, magnetic storage media such as hard disks, floppy disks, and magnetic tape; optical storage media such as Compact Disc/Digital Video Discs (CD/DVDs), Compact Disc-Read Only Memories (CD-ROMs), and holographic devices; magneto-optical storage media such as optical disks; carrier wave signal processing modules; and hardware devices that are specially configured to store and execute program code, such as Application-Specific Integrated Circuits (ASICs), Programmable Logic Devices (PLDs), Read-Only Memory (ROM) and Random-Access Memory (RAM) devices.
  • ASICs Application-Specific Integrated Circuits
  • PLDs Programmable Logic Devices
  • ROM Read-Only Memory
  • RAM Random-Access Memory
  • Other embodiments described herein relate to a computer program product, which can include, for example, the instructions and/or computer code discussed herein.
  • Hardware modules may include, for example, a general-purpose processor, a field programmable gate array (FPGA), and/or an application specific integrated circuit (ASIC).
  • Software modules (executed on hardware) can be expressed in a variety of software languages (e.g., computer code), including C, C++, JavaTM, Ruby, Visual BasicTM, R, and/or other object-oriented, procedural, statistical, or other programming language and development tools.
  • Examples of computer code include, but are not limited to, micro-code or micro-instructions, machine instructions, such as produced by a compiler, code used to produce a web service, and files containing higher-level instructions that are executed by a computer using an interpreter.
  • embodiments may be implemented using imperative programming languages (e.g., C, Fortran, etc.), functional programming languages (e.g., Haskell, Erlang, etc.), logical programming languages (e.g., Prolog), object-oriented programming languages (e.g., Java, C++, etc.), statistical programming languages and/or environments (e.g., R, etc.) or other suitable programming languages and/or development tools.
  • Additional examples of computer code include, but are not limited to, control signals, encrypted code, and compressed code.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pathology (AREA)
  • Pulmonology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
US15/523,654 2014-11-05 2015-11-05 Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data Abandoned US20170335396A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/523,654 US20170335396A1 (en) 2014-11-05 2015-11-05 Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201462075328P 2014-11-05 2014-11-05
US201562130800P 2015-03-10 2015-03-10
PCT/US2015/059309 WO2016073768A1 (en) 2014-11-05 2015-11-05 Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data
US15/523,654 US20170335396A1 (en) 2014-11-05 2015-11-05 Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/059309 A-371-Of-International WO2016073768A1 (en) 2014-11-05 2015-11-05 Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US201916551645A Continuation 2014-11-05 2019-08-26

Publications (1)

Publication Number Publication Date
US20170335396A1 true US20170335396A1 (en) 2017-11-23

Family

ID=55909824

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/523,654 Abandoned US20170335396A1 (en) 2014-11-05 2015-11-05 Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data
US16/840,009 Abandoned US20210079471A1 (en) 2014-11-05 2020-04-03 Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data
US17/218,125 Active US11639527B2 (en) 2014-11-05 2021-03-30 Methods for nucleic acid sequencing
US18/181,535 Pending US20240110242A1 (en) 2014-11-05 2023-03-09 Systems and methods of diagnosing idiopathic pulmonary fibrosis

Family Applications After (3)

Application Number Title Priority Date Filing Date
US16/840,009 Abandoned US20210079471A1 (en) 2014-11-05 2020-04-03 Systems and methods of diagnosing idiopathic pulmonary fibrosis on transbronchial biopsies using machine learning and high dimensional transcriptional data
US17/218,125 Active US11639527B2 (en) 2014-11-05 2021-03-30 Methods for nucleic acid sequencing
US18/181,535 Pending US20240110242A1 (en) 2014-11-05 2023-03-09 Systems and methods of diagnosing idiopathic pulmonary fibrosis

Country Status (5)

Country Link
US (4) US20170335396A1 (de)
EP (2) EP3770274A1 (de)
JP (2) JP7356788B2 (de)
CN (2) CN114606309A (de)
WO (1) WO2016073768A1 (de)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10526655B2 (en) 2013-03-14 2020-01-07 Veracyte, Inc. Methods for evaluating COPD status
CN110819700A (zh) * 2018-08-10 2020-02-21 杭州米天基因科技有限公司 一种构建肺部小结节计算机辅助检测模型的方法
US11479802B2 (en) 2017-04-11 2022-10-25 Regeneron Pharmaceuticals, Inc. Assays for screening activity of modulators of members of the hydroxy steroid (17-beta) dehydrogenase (HSD17B) family
WO2022261437A3 (en) * 2021-06-11 2023-01-05 Acceleron Pharma Inc. Actrii proteins and uses thereof
US11639527B2 (en) 2014-11-05 2023-05-02 Veracyte, Inc. Methods for nucleic acid sequencing
US11670417B2 (en) * 2021-10-05 2023-06-06 Neumora Therapeutics, Inc. Machine learning systems for processing multi-modal patient data
US11702700B2 (en) 2017-10-11 2023-07-18 Regeneron Pharmaceuticals, Inc. Inhibition of HSD17B13 in the treatment of liver disease in patients expressing the PNPLA3 I148M variation
US11753628B2 (en) 2017-01-23 2023-09-12 Regeneron Pharmaceuticals, Inc. HSD17B13 variants and uses thereof
US11976329B2 (en) * 2013-03-15 2024-05-07 Veracyte, Inc. Methods and systems for detecting usual interstitial pneumonia

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2017324949B2 (en) * 2016-09-07 2024-05-23 Veracyte, Inc. Methods and systems for detecting usual interstitial pneumonia
CA3036597C (en) * 2016-09-14 2023-03-28 Philip Morris Products S.A. Systems, methods, and gene signatures for predicting a biological status of an individual
WO2018223066A1 (en) * 2017-06-02 2018-12-06 Veracyte, Inc. Methods and systems for identifying or monitoring lung disease
WO2019202767A1 (ja) * 2018-04-17 2019-10-24 国立大学法人大阪大学 抗線維化剤及び線維症のバイオマーカー
US20200058399A1 (en) * 2018-08-16 2020-02-20 Htc Corporation Control method and reinforcement learning for medical system
EP3844757A1 (de) * 2018-08-30 2021-07-07 Life Technologies Corporation Maschinenlernsystem zur genotypisierung von pcr-tests
CN111401214B (zh) * 2020-03-12 2023-04-18 四川大学华西医院 一种基于深度学习的多分辨率集成her2判读方法
WO2022060842A1 (en) * 2020-09-15 2022-03-24 The Trustees Of Columbia University In The City Of New York Systems and methods for predicting graft dysfunction with exosome proteins
CN113607941A (zh) * 2020-10-22 2021-11-05 广州中医药大学顺德医院(佛山市顺德区中医院) 一种新型冠状病毒肺炎重症区分与疗效评价系统

Family Cites Families (329)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3640268A (en) 1965-10-23 1972-02-08 Hugh J Davis Method and device for biopsy specimen collecting and handling
AT277936B (de) 1968-02-27 1970-01-12 Guenter Dipl Ing Knapp Verfahren zur quantitativen Bestimmung von Jod und von Schilddrüsenhormonen sowie Vorrichtung zur Durchführung des Verfahrens
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4641662A (en) 1984-09-28 1987-02-10 Jaicks John R Endocervical curette system
US4800896A (en) 1985-11-08 1989-01-31 Jalowayski Alfredo A Cell sample collector probe
US5130238A (en) 1988-06-24 1992-07-14 Cangene Corporation Enhanced nucleic acid amplification process
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
CA2020958C (en) 1989-07-11 2005-01-11 Daniel L. Kacian Nucleic acid sequence amplification methods
US6013431A (en) 1990-02-16 2000-01-11 Molecular Tool, Inc. Method for determining specific nucleotide variations by primer extension in the presence of mixture of labeled nucleotides and terminators
US5494810A (en) 1990-05-03 1996-02-27 Cornell Research Foundation, Inc. Thermostable ligase-mediated DNA amplifications system for the detection of genetic disease
US5798257A (en) 1990-07-09 1998-08-25 Research Corporation Technologies, Inc. Nucleic acid encoding human MTS-1 protein
EP0562025B1 (de) 1990-12-06 2001-02-07 Affymetrix, Inc. (a Delaware Corporation) Verbindungen und ihre Verwendung in einer binären Synthesestrategie
US5455166A (en) 1991-01-31 1995-10-03 Becton, Dickinson And Company Strand displacement amplification
US5994069A (en) 1996-01-24 1999-11-30 Third Wave Technologies, Inc. Detection of nucleic acids by multiple sequential invasive cleavages
US5846717A (en) 1996-01-24 1998-12-08 Third Wave Technologies, Inc. Detection of nucleic acid sequences by invader-directed cleavage
US5726060A (en) 1991-09-17 1998-03-10 Bridges; Michael Anthony Method for culturing mammalian respiratory epithelial cells
CA2124087C (en) 1991-11-22 2002-10-01 James L. Winkler Combinatorial strategies for polymer synthesis
US5384261A (en) 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US6204370B1 (en) 1992-03-11 2001-03-20 Institute Of Virology, Slovak Academy Of Sciences MN gene and protein
US6027887A (en) 1992-03-11 2000-02-22 Institute Of Virology, Solvak Academy Of Sciences MN gene and protein
US5541061A (en) 1992-04-29 1996-07-30 Affymax Technologies N.V. Methods for screening factorial chemical libraries
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5422273A (en) 1993-03-23 1995-06-06 Baal Medical Products, Inc. Cell collection apparatus
US5876978A (en) 1993-04-06 1999-03-02 Medical College Of Ohio Method for quantitative measurement of gene expression using multiplex competitive reverse transcriptase-polymerase chain reaction
US5477863A (en) 1993-04-14 1995-12-26 Grant; Michael A. Collection kit with a sample collector
US5538848A (en) 1994-11-16 1996-07-23 Applied Biosystems Division, Perkin-Elmer Corp. Method for detecting nucleic acid amplification using self-quenching fluorescence probe
US5529925A (en) 1993-12-03 1996-06-25 St. Jude Children's Research Hospital Nucleic acid sequences and fusion proteins present in human t(2;5) lymphoma
US5440942A (en) 1994-02-02 1995-08-15 Hubbard; Stephen H. Biological sample collecting and holding device
US5989815A (en) 1994-03-18 1999-11-23 University Of Utah Research Foundation Methods for detecting predisposition to cancer at the MTS gene
US5648211A (en) 1994-04-18 1997-07-15 Becton, Dickinson And Company Strand displacement amplification using thermophilic enzymes
US7625697B2 (en) 1994-06-17 2009-12-01 The Board Of Trustees Of The Leland Stanford Junior University Methods for constructing subarrays and subarrays made thereby
US5830753A (en) 1994-09-30 1998-11-03 Ludwig Institute For Cancer Research Isolated nucleic acid molecules coding for tumor rejection antigen precursor dage and uses thereof.
US5795782A (en) 1995-03-17 1998-08-18 President & Fellows Of Harvard College Characterization of individual polymer molecules based on monomer-interface interactions
US5710029A (en) 1995-06-07 1998-01-20 Gen-Probe Incorporated Methods for determining pre-amplification levels of a nucleic acid target sequence from post-amplification levels of product
US5705365A (en) 1995-06-07 1998-01-06 Gen-Probe Incorporated Kits for determining pre-amplification levels of a nucleic acid target sequence from post-amplification levels of product
JP2001520636A (ja) 1995-08-30 2001-10-30 ザ リージェンツ オブ ザ ユニバーシティー オブ カリフォルニア 慢性炎症性疾患における細胞蓄積の治療
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US5985557A (en) 1996-01-24 1999-11-16 Third Wave Technologies, Inc. Invasive cleavage of nucleic acids
WO1997039354A1 (fr) 1996-04-16 1997-10-23 Kishimoto, Tadamitsu Procede de detection de cellules solides cancereuses et d'heterotypie histologique et procede d'examen du tissu dans le but d'effectuer une transplantation de moelle osseuse et une transplantation de cellules souches du sang peripherique
US6136182A (en) 1996-06-07 2000-10-24 Immunivest Corporation Magnetic devices and sample chambers for examination and manipulation of cells
GB9711040D0 (en) 1997-05-29 1997-07-23 Duff Gordon W Prediction of inflammatory disease
AU7799498A (en) 1997-06-10 1998-12-30 Quadrivium, L.L.C. System and method for detection of a biological condition
US20020076735A1 (en) 1998-09-25 2002-06-20 Williams Lewis T. Diagnostic and therapeutic methods using molecules differentially expressed in cancer cells
US6085907A (en) 1998-05-08 2000-07-11 Institute Of Legal Medicine, University Of Bern Foldable cardboard box for contact-free drying and long-term storage of biological evidence recovered on cotton swabs and forensic evidence collection kit including same
EP1082459A4 (de) 1998-05-21 2002-09-25 Diadexus Inc Neue methode zur diagnose, überwachung und bestimmung des stadiums von lungenkrebs
AU752642B2 (en) 1998-06-08 2002-09-26 Fuso Pharmaceutical Industries, Ltd. Antibody against LAR phosphatase subunit
US6204375B1 (en) 1998-07-31 2001-03-20 Ambion, Inc. Methods and reagents for preserving RNA in cell and tissue samples
US6468476B1 (en) 1998-10-27 2002-10-22 Rosetta Inpharmatics, Inc. Methods for using-co-regulated genesets to enhance detection and classification of gene expression patterns
EP1873260B1 (de) 1998-11-09 2009-03-18 Eiken Kagaku Kabushiki Kaisha Prozess zur Synthetisierung von Nukleinsäure
US20040241728A1 (en) 1999-01-06 2004-12-02 Chondrogene Limited Method for the detection of lung disease related gene transcripts in blood
US6436642B1 (en) 1999-04-20 2002-08-20 Curagen Corporation Method of classifying a thyroid carcinoma using differential gene expression
US7258838B2 (en) 1999-06-22 2007-08-21 President And Fellows Of Harvard College Solid state molecular probe device
EP1192453B1 (de) 1999-06-22 2012-02-15 President and Fellows of Harvard College Bestimmung von biopolymeren entsprechend der molekularen oder atomaren skala
US6667154B1 (en) 1999-06-30 2003-12-23 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
US6746846B1 (en) 1999-06-30 2004-06-08 Corixa Corporation Methods for diagnosing lung cancer
DE60009323T2 (de) 1999-09-13 2005-02-10 Nugen Technologies, Inc., San Carlos Methoden und zusammensetzungen zur linearen isothermalen amplifikation von polynukleotidsequenzen
US7211390B2 (en) 1999-09-16 2007-05-01 454 Life Sciences Corporation Method of sequencing a nucleic acid
US7244559B2 (en) 1999-09-16 2007-07-17 454 Life Sciences Corporation Method of sequencing a nucleic acid
EP1223866A1 (de) 1999-10-18 2002-07-24 Denis Marshall-Fraser Gerät zur entnahme einer probe
WO2001052789A2 (en) 2000-01-20 2001-07-26 The Brigham And Women's Hospital, Inc. PAX8-PPARη NUCLEIC ACID MOLECULES AND POLYPEPTIDES AND USES THEREOF
US7361488B2 (en) 2000-02-07 2008-04-22 Illumina, Inc. Nucleic acid detection methods using universal priming
US6383804B1 (en) 2000-07-13 2002-05-07 International Bioproducts, Inc. Sampling device with snap-off head and method of use
AU2001281136A1 (en) 2000-08-04 2002-02-18 Board Of Regents, The University Of Texas System Detection and diagnosis of smoking related cancers
US20020160388A1 (en) 2000-11-20 2002-10-31 Roberto Macina Compositions and methods relating to lung specific genes and proteins
AU2002236507A1 (en) 2000-11-29 2002-06-11 Cangen International Dap-kinase and hoxa9, two human genes associated with genesis, progression, and aggressiveness of non-small cell lung cancer
US6988040B2 (en) 2001-01-11 2006-01-17 Affymetrix, Inc. System, method, and computer software for genotyping analysis and identification of allelic imbalance
US6972181B2 (en) 2001-01-12 2005-12-06 Mount Sinai School Of Medicine Of New York University Differential diagnosis of cancer and other conditions based on expression of p63
WO2003040325A2 (en) 2001-02-01 2003-05-15 Curagen Corporation Novel proteins and nucleic acids encoding same
US20020169137A1 (en) 2001-02-09 2002-11-14 Active Pass Pharmaceuticals, Inc. Regulation of amyloid precursor protein expression by modification of ABC transporter expression or activity
US7026163B1 (en) 2001-02-23 2006-04-11 Mayo Foundation For Medical Education And Research Sulfotransferase sequence variants
IL157872A0 (en) 2001-03-12 2004-03-28 Monogen Inc A panel for detecting a generic disease state containing a plurality of probes and using cell-based diagnosis
US20030190602A1 (en) 2001-03-12 2003-10-09 Monogen, Inc. Cell-based detection and differentiation of disease states
AU2002250285A1 (en) 2001-03-14 2002-09-24 Medical College Of Ohio Method for quantitative measurement of gene expression for identifying individuals at risk for bronchogenic carcinoma
CA2444691A1 (en) 2001-04-18 2002-10-31 Protein Design Labs, Inc. Methods of diagnosis of lung cancer, compositions and methods of screening for modulators of lung cancer
US6494845B2 (en) 2001-05-04 2002-12-17 Oralscan Laboratories, Inc. Retractable brush for use with endoscope for brush biopsy
EP1453977B1 (de) 2001-08-16 2009-11-18 THE UNITED STATES OF AMERICA, as represented by the Secretary of the Department of Health and Human Services Molekulare eigenschaften von nichtkleinzelligem lungenkrebs
JP4291143B2 (ja) 2001-08-31 2009-07-08 ジェン−プロウブ インコーポレイテッド 分析物ポリヌクレオチドを定量するためのアフィニティーシフトしたプローブ
US6824520B2 (en) 2001-09-21 2004-11-30 Pulmonary Data Services, Inc. Method and apparatus for tracking usage of a respiratory measurement device
WO2003029273A2 (en) 2001-09-28 2003-04-10 Whitehead Institute For Biomedical Research Classification of lung carcinomas using gene expression analysis
WO2003051388A2 (en) * 2001-12-18 2003-06-26 Mondobiotech Laboratories Anstalt Pharmaceutical composition of interferon gamma or pirfenidone with molecular diagnostics for the improved treatment of interstitial lung diseases
US7141372B2 (en) 2002-01-18 2006-11-28 Health Research Incorporated Universal RT-coupled PCR method for the specific amplification of mRNA
US20040005294A1 (en) 2002-02-25 2004-01-08 Ho-Young Lee IGFBP-3 in the diagnosis and treatment of cancer
CA2480045C (en) 2002-03-28 2013-09-24 Medical College Of Ohio Method and compositions for the diagnosis and treatment of non-small cell lung cancer using gene expression profiles
US20030186248A1 (en) 2002-03-29 2003-10-02 Erlander Mark G. Interpreting cytological specimens via molecular histological signatures
US20030194734A1 (en) 2002-03-29 2003-10-16 Tim Jatkoe Selection of markers
US20050143933A1 (en) 2002-04-23 2005-06-30 James Minor Analyzing and correcting biological assay data using a signal allocation model
DE10219117C1 (de) 2002-04-29 2003-10-30 Adnagen Ag Verfahren zur Stabilisierung von RNS und Verwendungen von Stabilisierungspuffern
WO2003097666A2 (en) 2002-05-16 2003-11-27 Euro-Celtique, S.A. Use of gastrin releasing peptide (grp) and its receptor in cancer screening
WO2004005891A2 (en) 2002-07-10 2004-01-15 The Regents Of The University Of Michigan Expression profile of lung cancer
EP2385137A1 (de) 2002-07-31 2011-11-09 University of Southern California Polymorphismen zur Vorhersage des Krankheits- und Behandlungsausgangs
US7282328B2 (en) 2002-09-20 2007-10-16 New England Biolabs, Inc. Helicase dependent amplification of nucleic acids
EP1403638A1 (de) 2002-09-25 2004-03-31 Mondobiotech SA Molekulare diagnostische Methoden zur Erkennung interstitieller Lungenerkrankungen
ATE411996T1 (de) 2002-09-30 2008-11-15 Bayer Healthcare Ag Kondensierte azolpyrimidinderivate
US7300788B2 (en) 2002-10-08 2007-11-27 Affymetrix, Inc. Method for genotyping polymorphisms in humans
US20050266443A1 (en) 2002-10-11 2005-12-01 Thomas Jefferson University Novel tumor suppressor gene and compositions and methods for making and using the same
US20060127907A1 (en) 2002-11-07 2006-06-15 Daiichi Pure Chemicals Co., Ltd. Method of detecting gene mutation
EP2159285B1 (de) 2003-01-29 2012-09-26 454 Life Sciences Corporation Verfahren zur Verstärkung und Sequenzierung von Nukleinsäuren
US20050266409A1 (en) 2003-02-04 2005-12-01 Wyeth Compositions and methods for diagnosing, preventing, and treating cancers
PT1597391E (pt) 2003-02-20 2008-12-19 Genomic Health Inc Utilização de arn intrónico para medir a expressão genética
US20060265138A1 (en) 2003-03-14 2006-11-23 David Bowtell Expression profiling of tumours
US20040241725A1 (en) 2003-03-25 2004-12-02 Wenming Xiao Lung cancer detection
WO2004091383A2 (en) 2003-04-01 2004-10-28 Genzyme Corporation Breast endothelial cell expression patterns
WO2004091511A2 (en) 2003-04-10 2004-10-28 Genzyme Corporation Compositions and methods to diagnose and treat lung cancer
US7378233B2 (en) 2003-04-12 2008-05-27 The Johns Hopkins University BRAF mutation T1796A in thyroid cancers
WO2005001132A2 (en) 2003-05-30 2005-01-06 The Board Of Trustees Of The University Of Illinois Gene expression profiles that identify genetically elite ungulate mammals
US20060019256A1 (en) 2003-06-09 2006-01-26 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
WO2004111197A2 (en) 2003-06-10 2004-12-23 Trustees Of Boston University Gene expression signatures, methods and compositions for diagnosing disorders of the lung
EP2327795B1 (de) 2003-06-10 2017-08-09 The Trustees Of Boston University Nachweismethoden für Lungenerkrankungen
ES2651849T3 (es) 2003-07-10 2018-01-30 Genomic Health, Inc. Algoritmo del perfil de expresión y test para el pronóstico del cáncer
WO2005007830A2 (en) 2003-07-14 2005-01-27 Mayo Foundation For Medical Education And Research Methods and compositions for diagnosis, staging and prognosis of prostate cancer
WO2005017652A2 (en) 2003-08-05 2005-02-24 Rosetta Inpharmatics, Llc Computer systems and methods for inferring causality from cellular constituent abundance data
US7678061B2 (en) 2003-09-18 2010-03-16 Cardiac Pacemakers, Inc. System and method for characterizing patient respiration
CA2552686A1 (en) 2003-11-12 2005-05-26 Trustees Of Boston University Isolation of nucleic acid from mouth epithelial cells
JP2005168432A (ja) 2003-12-12 2005-06-30 Nara Institute Of Science & Technology 甲状腺濾胞癌と甲状腺濾胞腺腫を判別するためのデータを収集するための方法及び該方法のためのキット
US20050130177A1 (en) 2003-12-12 2005-06-16 3M Innovative Properties Company Variable valve apparatus and methods
WO2005059108A2 (en) 2003-12-12 2005-06-30 Bayer Pharmaceuticals Corporation Gene expression profiles and methods of use
WO2005062770A2 (en) 2003-12-19 2005-07-14 Novakoff James L Method for conducting pharmacogenomics-based studies
US20090020433A1 (en) 2003-12-31 2009-01-22 Microfabrica Inc. Electrochemical Fabrication Methods for Producing Multilayer Structures Including the use of Diamond Machining in the Planarization of Deposits of Material
US7407755B2 (en) 2004-01-15 2008-08-05 Lubinski Jan Determining a predisposition to cancer
CA2554818A1 (en) 2004-02-09 2005-08-25 Thomas Jefferson University Diagnosis and treatment of cancers with microrna located in or near cancer-associated chromosomal features
WO2005085471A2 (de) 2004-03-03 2005-09-15 Universität Leipzig Verfahren und mittel zur differentiellen diagnose von schilddrüsentumoren
WO2005086679A2 (en) 2004-03-05 2005-09-22 Medical College Of Ohio Methods and compositions for assessing nucleic acids and alleles
US8426126B2 (en) 2004-03-18 2013-04-23 Applied Biosystems, Llc Modified surfaces as solid supports for nucleic acid purification
WO2006028508A2 (en) 2004-03-23 2006-03-16 President And Fellows Of Harvard College Methods and apparatus for characterizing polynucleotides
US20100075384A1 (en) 2004-03-25 2010-03-25 New England Biolabs, Inc. Helicase-dependent amplification of circular nucleic acids
JP2005304497A (ja) 2004-03-25 2005-11-04 Joji Inasawa 特定の癌関連遺伝子を用いる癌の検出方法及び癌の抑制方法
US7319011B2 (en) 2004-04-08 2008-01-15 Duke University Method for distinguishing follicular thyroid adenoma (FTA) from follicular thyroid carcinoma (FTC)
ATE491043T1 (de) 2004-04-09 2010-12-15 Us Gov Health & Human Serv Diagnosegerät zur diagnose gutartiger gegenüber bösartigen schilddrüsenläsionen
US20050240357A1 (en) 2004-04-26 2005-10-27 Minor James M Methods and systems for differential clustering
US20060019272A1 (en) 2004-05-03 2006-01-26 The Regents Of The University Of Colorado Diagnosis of disease and monitoring of therapy using gene expression analysis of peripheral blood cells
EP2290069A3 (de) 2004-05-28 2011-08-10 Asuragen, Inc. Verfahren und Zusammensetzungen mit MicroRNA
AU2005260113A1 (en) 2004-06-04 2006-01-12 Aviaradx, Inc. The importance of the gene HoxB13 for cancer
JP4533015B2 (ja) 2004-06-15 2010-08-25 キヤノン株式会社 化合物及びそれを用いた有機エレクトロルミネッセンス素子
US20060019615A1 (en) 2004-07-24 2006-01-26 Ditmer Larry R Wireless keying for a continuous wave (CW) amateur radio transmitter
GB0417740D0 (en) 2004-08-10 2004-09-08 Uc3 Methods and kit for the prognosis of breast cancer
JP2008509672A (ja) 2004-08-11 2008-04-03 ザ クリーブランド クリニック ファウンデーション 甲状腺癌の検出方法
CN101103357B (zh) 2004-08-13 2012-10-03 哈佛学院院长等 超高处理量光学-纳米孔dna读出平台
PL1810026T3 (pl) 2004-10-06 2018-08-31 Mayo Foundation For Medical Education And Research B7-H1 i PD-1 w leczeniu raka nerkowokomórkowego
US7629325B2 (en) 2004-10-11 2009-12-08 Technion Research & Development Foundation Ltd. Human Sef isoforms and methods of using same for cancer diagnosis and gene therapy
US7485468B2 (en) 2004-10-15 2009-02-03 Galapagos Bv Molecular targets and compounds, and methods to identify the same, useful in the treatment of joint degenerative and inflammatory diseases
EP1812601A2 (de) 2004-10-22 2007-08-01 Redpath Integrated Pathology, Inc. Dynamische expansion genomischer deletion und formulierung aus molekularmarkerpanels für integrierte molekulare pathologiediagnose und charakterisierung von gewebe, zellflüssigkeit und reinen flüssigproben
CA2587817A1 (en) 2004-11-11 2006-05-18 Garvan Institute Of Medical Research Method of diagnosing cancer and reagents therefor
JP2008521412A (ja) 2004-11-30 2008-06-26 ベリデックス・エルエルシー 肺癌予後判定手段
CA2589885A1 (en) 2004-12-03 2006-06-08 Schering Corporation Biomarkers for pre-selection of patients for anti-igf1r therapy
JP5629894B2 (ja) 2004-12-07 2014-11-26 国立大学法人大阪大学 甲状腺乳頭癌を診断するための新規のマーカー
US7862995B2 (en) 2004-12-10 2011-01-04 Targeted Molecular Diagnostics Methods and materials for predicting responsiveness to treatment with dual tyrosine kinase inhibitor
EP1851332A4 (de) 2005-01-21 2010-05-26 Gene Express Inc Verfahren und zusammensetzungen zur beurteilung von nukleinsäuren
US20120122718A1 (en) 2005-03-01 2012-05-17 Reisman David N BRM Expression and Related Diagnostics
WO2006105252A2 (en) 2005-03-28 2006-10-05 The Regents Of The University Of Colorado Diagnosis of chronic pulmonary obstructive disease and monitoring of therapy using gene expression analysis of peripheral blood cells
WO2006110593A2 (en) 2005-04-07 2006-10-19 Macrogenics, Inc. Biological targets for the diagnosis, treatment and prevention of cancer
US20200248274A1 (en) 2005-04-14 2020-08-06 Trustees Of Boston University Diagnostic for lung disorders using class prediction
CA2605158A1 (en) 2005-04-14 2006-10-26 The Trustees Of Boston University Diagnostic for lung disorders using class prediction
US20070020657A1 (en) 2005-05-20 2007-01-25 Grebe Stefan K Methods for detecting circulating tumor cells
US20070037186A1 (en) 2005-05-20 2007-02-15 Yuqiu Jiang Thyroid fine needle aspiration molecular assay
DK1899484T3 (da) 2005-06-03 2015-11-23 Biotheranostics Inc Identifikation af tumorer og væv
WO2006135886A2 (en) 2005-06-13 2006-12-21 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
WO2006138275A2 (en) 2005-06-13 2006-12-28 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
IL177006A0 (en) 2005-08-02 2006-12-10 Veridex Llc Predicting bone relapse of breast cancer
US7931902B2 (en) 2005-08-15 2011-04-26 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
JP2009507229A (ja) 2005-09-02 2009-02-19 ザ・ユニバーシティ・オブ・トレド 生物学的状態の診断及び/又は治療に有用なバイオマーカーを同定するための方法及び組成物
CN101313306B (zh) 2005-09-22 2011-11-16 中国合成橡胶股份有限公司 用于鉴别鼻咽癌中的预后型亚类的基因表达谱
US20070172844A1 (en) 2005-09-28 2007-07-26 University Of South Florida Individualized cancer treatments
US7962291B2 (en) 2005-09-30 2011-06-14 Affymetrix, Inc. Methods and computer software for detecting splice variants
US20080254470A1 (en) 2005-10-03 2008-10-16 Epigenomics Ag Methods and Nucleic Acids For the Analysis of Gene Expression Associated With the Prognosis of Cell Proliferative Disorders
US7598052B2 (en) 2005-10-11 2009-10-06 The Regents Of The University Of Michigan Expression profile of thyroid cancer
US20070220621A1 (en) 2005-10-31 2007-09-20 Clarke Michael F Genetic characterization and prognostic significance of cancer stem cells in cancer
US9347945B2 (en) 2005-12-22 2016-05-24 Abbott Molecular Inc. Methods and marker combinations for screening for predisposition to lung cancer
FR2896881B1 (fr) 2006-01-31 2008-04-18 Biomerieux Sa Procede de dosage du prongf pour le diagnostic in vitro du cancer du sein et utilisation du prongf en therapie
US7670775B2 (en) 2006-02-15 2010-03-02 The Ohio State University Research Foundation Method for differentiating malignant from benign thyroid tissue
EP1999472A2 (de) 2006-03-09 2008-12-10 The Trustees Of Boston University Diagnose- und prognoseverfahren für lungenerkrankungen mittels genexpressionsprofilen aus nasenepithelzellen
JP4867018B2 (ja) 2006-03-22 2012-02-01 富士フイルム株式会社 癌の検出方法および抑制方法
WO2007126882A2 (en) 2006-03-27 2007-11-08 Jivan Biologics, Inc. Analysis of splice variant expression data
US20080028302A1 (en) 2006-07-31 2008-01-31 Steffen Meschkat Method and apparatus for incrementally updating a web page
KR20080020083A (ko) 2006-08-30 2008-03-05 유영도 암 진단 마커로서의 Romo1의 용도
KR101443214B1 (ko) 2007-01-09 2014-09-24 삼성전자주식회사 폐암 환자 또는 폐암 치료를 받은 폐암 환자의 폐암 재발 위험을 진단하기 위한 조성물, 키트 및 마이크로어레이
SI2472264T1 (sl) 2007-02-27 2016-06-30 SentoClone International AB Karolinska Institute Science Park Multipleksna detekcija tumorskih celic z uporabo plošč vezavnih sredstev na zunajcelične markerje
WO2008109773A2 (en) 2007-03-06 2008-09-12 Cornell Research Foundation, Inc. Chronic obstructive pulmonary disease susceptibility and related compositions and methods
US9096906B2 (en) 2007-03-27 2015-08-04 Rosetta Genomics Ltd. Gene expression signature for classification of tissue of origin of tumor samples
US8802599B2 (en) 2007-03-27 2014-08-12 Rosetta Genomics, Ltd. Gene expression signature for classification of tissue of origin of tumor samples
WO2010073248A2 (en) 2008-12-24 2010-07-01 Rosetta Genomics Ltd. Gene expression signature for classification of tissue of origin of tumor samples
US20100273172A1 (en) 2007-03-27 2010-10-28 Rosetta Genomics Ltd. Micrornas expression signature for determination of tumors origin
JP2010522554A (ja) 2007-03-27 2010-07-08 ロゼッタ ゲノミックス エルティーディー. 癌の分類のための遺伝子発現サイン
EP1975245A1 (de) 2007-03-28 2008-10-01 Greenpeace e.V. Gen-Profiling zur Auswahl von politischen Kandidaten, kommerzielle Nutzung von Politikern
EP1975252A1 (de) 2007-03-29 2008-10-01 INSERM (Institut National de la Santé et de la Recherche Medicale) Verfahren zur Vorhersage oder Diagnose einer Schilddrüsenkrankheit
US8455208B2 (en) 2007-04-14 2013-06-04 The Regents Of The University Of Colorado Biomarkers for follicular thyroid carcinoma and methods of use
JP2010528261A (ja) 2007-05-08 2010-08-19 ピコベラ・リミテッド・ライアビリティ・カンパニー 前立腺癌および肺癌の診断および治療方法
US7901888B2 (en) 2007-05-09 2011-03-08 The Regents Of The University Of California Multigene diagnostic assay for malignant thyroid neoplasm
SG182183A1 (en) 2007-06-08 2012-07-30 Biogen Idec Inc Biomarkers for predicting anti-tnf responsiveness or non-responsiveness
ES2627059T3 (es) 2007-08-03 2017-07-26 The Ohio State University Research Foundation Regiones ultraconservadas que codifican ARNnc
WO2009029266A2 (en) 2007-08-27 2009-03-05 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health Of Human Services, National Institutes Of Health Diagnostic tool for diagnosing benign versus malignant thyroid lesions
AT505726A2 (de) 2007-08-30 2009-03-15 Arc Austrian Res Centers Gmbh Set von tumor-markern
RU2511408C2 (ru) 2007-09-17 2014-04-10 Конинклейке Филипс Электроникс Н.В. Способ анализа нарушений, связанных с раком яичников
AU2008302076B2 (en) 2007-09-19 2015-06-11 The Trustees Of Boston University Identification of novel pathways for drug development for lung disease
WO2009037337A1 (en) 2007-09-20 2009-03-26 Novartis Ag Robust and tissue independent gender-specific transcript markers for molecular gender determination
US20100285468A1 (en) 2007-09-24 2010-11-11 Allelogic Biosciences Corporation Detection and/or quantification of nucleic acids
EP2245188A2 (de) 2007-11-28 2010-11-03 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Biomarker für den ausbruch neurodegenerativer krankheiten
US8476420B2 (en) 2007-12-05 2013-07-02 The Wistar Institute Of Anatomy And Biology Method for diagnosing lung cancers using gene expression profiles in peripheral blood mononuclear cells
JP2011505840A (ja) 2007-12-14 2011-03-03 ベリデックス・エルエルシー ヒト乳癌の転移能を予測するコピー数変化
US20090191535A1 (en) 2007-12-22 2009-07-30 Mark Carle Connelly Method of assessing metastatic carcinomas from circulating endothelial cells and disseminated tumor cells
JP2011508598A (ja) 2008-01-04 2011-03-17 サントル ナショナル ドゥ ラ ルシェルシュ シアンティフィク 乳癌のインビトロ分子診断
US20090192045A1 (en) 2008-01-22 2009-07-30 Yuqiu Jiang Molecular staging of stage ii and iii colon cancer and prognosis
GB2469244A (en) 2008-01-28 2010-10-06 Univ Singapore Lipid tumour profile
US20090246779A1 (en) 2008-02-15 2009-10-01 University Of Washington Increasing genomic instability during premalignant neoplastic progression revealed through high resolution array-cgh
WO2009111881A1 (en) 2008-03-13 2009-09-17 British Columbia Cancer Agency Branch Biomarkers for diagnosis of differentiated thyroid cancer
WO2009120800A2 (en) 2008-03-25 2009-10-01 University Of Southern California Prognostic panel for urinary bladder cancer
US20100055689A1 (en) 2008-03-28 2010-03-04 Avrum Spira Multifactorial methods for detecting lung disorders
AU2009234444A1 (en) 2008-04-11 2009-10-15 China Synthetic Rubber Corporation Methods, agents and kits for the detection of cancer
EP2297345A1 (de) 2008-05-14 2011-03-23 DNAR, Inc Biomarker zur identifizierung, überwachung und behandlung von krebserkrankungen im kopf- und halsbereich
AU2009253675A1 (en) 2008-05-28 2009-12-03 Genomedx Biosciences, Inc. Systems and methods for expression-based discrimination of distinct clinical disease states in prostate cancer
JP5717178B2 (ja) * 2008-06-05 2015-05-13 国立大学法人佐賀大学 特発性間質性肺炎の検出方法
CA2726531A1 (en) 2008-06-05 2009-12-10 University Health Network Compositions and methods for classifying lung cancer and prognosing lung cancer survival
WO2009153775A2 (en) 2008-06-17 2009-12-23 Rosetta Genomics Ltd. Methods for distinguishing between specific types of lung cancers
GB0811413D0 (en) 2008-06-20 2008-07-30 Kanton Basel Stadt Gene expression signatures for lung cancers
ES2338843B1 (es) 2008-07-02 2011-01-24 Centro De Investigaciones Energeticas, Medioambientales Y Tecnologicas Huella genomica de cancer de mama.
WO2010004589A2 (en) 2008-07-07 2010-01-14 Decode Genetics Ehf Genetic variants predictive of cancer risk in humans
US20110190156A1 (en) 2008-07-15 2011-08-04 Trustees Of Dartmouth College Molecular signatures for diagnosing scleroderma
CN102177252B (zh) 2008-08-12 2014-12-24 解码遗传学私营有限责任公司 用于甲状腺癌症的风险评估的遗传变型
EP2329037B1 (de) 2008-08-15 2015-01-28 Decode Genetics EHF Genetische varianten zur vorhersage des krebsrisikos
CA2736124A1 (en) 2008-09-03 2010-03-11 The Johns Hopkins University Pathways underlying pancreatic tumorigenesis and an hereditary pancreatic cancer gene
WO2010028274A1 (en) * 2008-09-05 2010-03-11 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Marker panels for idiopathic pulmonary fibrosis diagnosis and evaluation
WO2010030365A2 (en) 2008-09-12 2010-03-18 Cornell Research Foundation, Inc. Wmc Thyroid tumors identified
CN102232117A (zh) 2008-10-14 2011-11-02 卡里斯Mpi公司 描绘肿瘤类型生物标志模式和特征集的基因靶和基因表达的蛋白靶
WO2010054233A1 (en) 2008-11-08 2010-05-14 The Wistar Institute Of Anatomy And Biology Biomarkers in peripheral blood mononuclear cells for diagnosing or detecting lung cancers
CA2742489A1 (en) 2008-11-14 2010-05-20 Intelligent Oncotherapeutics, Inc. Methods for identification of tumor phenotype and treatment
WO2010056351A2 (en) 2008-11-14 2010-05-20 Stc.Unm Gene expression classifiers for relapse free survival and minimal residual disease improve risk classification and out come prediction in pedeatric b-precursor acute lymphoblastic leukemia
US9495515B1 (en) 2009-12-09 2016-11-15 Veracyte, Inc. Algorithms for disease diagnostics
CA2743473C (en) 2008-11-17 2022-06-14 Veracyte, Inc. Methods and compositions of molecular profiling for disease diagnostics
US10236078B2 (en) 2008-11-17 2019-03-19 Veracyte, Inc. Methods for processing or analyzing a sample of thyroid tissue
AU2009316376B2 (en) 2008-11-21 2015-08-20 Celularity Inc. Treatment of diseases, disorders or conditions of the lung using placental cells
CA2777638A1 (en) 2008-11-26 2010-06-03 Decode Genetics Ehf. Genetic variants useful for risk assessment of thyroid cancer
US9090943B2 (en) 2008-11-30 2015-07-28 Rosetta Genomics Ltd. Methods for detecting an increased susceptibility to cancer
CA2749113A1 (en) 2009-01-09 2010-07-15 The Regents Of The University Of Michigan Recurrent gene fusions in cancer
US20100204058A1 (en) 2009-01-28 2010-08-12 Howard Yuan-Hao Chang Profiling for Determination of Response to Treatment for Inflammatory Disease
WO2010091049A2 (en) 2009-02-03 2010-08-12 Children's Medical Center Corporation Diagnosis and treatment of cancer
US9074258B2 (en) 2009-03-04 2015-07-07 Genomedx Biosciences Inc. Compositions and methods for classifying thyroid nodule disease
US8697275B2 (en) 2009-03-04 2014-04-15 Samsung Sdi Co., Ltd. Rechargeable battery having an extendable case region
WO2010123625A1 (en) 2009-04-24 2010-10-28 University Of Southern California Cd133 polymorphisms predict clinical outcome in patients with cancer
CA2753916C (en) 2009-04-29 2020-08-25 Genomedx Biosciences Inc. Systems and methods for expression-based classification of thyroid tissue
SG175832A1 (en) 2009-05-01 2011-12-29 Genomic Health Inc Gene expression profile algorithm and test for likelihood of recurrence of colorectal cancer and response to chemotherapy
EP2427575B1 (de) 2009-05-07 2018-01-24 Veracyte, Inc. Verfahren zur diagnose von schilddrüsenleiden
US20120264626A1 (en) 2009-05-08 2012-10-18 The Ohio State University Research Foundation MicroRNA Expression Profiling and Targeting in Chronic Obstructive Pulmonary Disease (COPD) Lung Tissue and Methods of Use Thereof
JP5995721B2 (ja) 2009-08-28 2016-09-21 アスラジェン, インコーポレイテッド 肺疾患のmiRNAバイオマーカー
WO2011023413A1 (en) 2009-08-31 2011-03-03 Alcedo Biotech Gmbh Microrna-based methods and compositions for reprogramming cells
US9110065B2 (en) 2009-09-21 2015-08-18 Paul Walfish Methods and compositions for the diagnosis and treatment of thyroid cancer
EP2483813A1 (de) 2009-10-01 2012-08-08 Chipdx LLC System und verfahren zur klassifikation von patienten
US20120329666A1 (en) 2009-10-05 2012-12-27 Duke University Peripheral Blood Biomarkers for Idiopathic Interstitial Pneumonia and Methods of Use
WO2011047358A1 (en) 2009-10-15 2011-04-21 Crescendo Bioscience Biomarkers and methods for measuring and monitoring inflammatory disease activity
US8975019B2 (en) 2009-10-19 2015-03-10 University Of Massachusetts Deducing exon connectivity by RNA-templated DNA ligation/sequencing
WO2011054893A2 (en) 2009-11-05 2011-05-12 Novartis Ag Biomarkers predictive of progression of fibrosis
US10446272B2 (en) 2009-12-09 2019-10-15 Veracyte, Inc. Methods and compositions for classification of samples
EP2519651A2 (de) 2009-12-30 2012-11-07 Rigshospitalet Klassifizierung von follikulärer thyroidneoplasie auf grundlage von mrna-expression
GB201000688D0 (en) 2010-01-15 2010-03-03 Diagenic Asa Product and method
HUE033044T2 (hu) 2010-01-26 2017-11-28 Nat Jewish Health Eljárás tüdõ rendellenességek kockázatának elõrejelzésére, diagnózisára, kórjóslatára
EP2366800A1 (de) 2010-03-01 2011-09-21 Centrum Onkologii-Instytut im M. Sklodowskiej-Curie Oddzial w Gliwicach Kit, Methode und Verwendung eines Genexpressionsprofils zur Diagnose von papillären Schilddrüsenkrebs
WO2011116380A2 (en) 2010-03-19 2011-09-22 H. Lee Moffitt Cancer Center And Research Institute, Inc. Hybrid model for the classification of carcinoma subtypes
WO2011127561A1 (en) * 2010-04-12 2011-10-20 University Health Network Methods and compositions for diagnosing pulmonary fibrosis subtypes and assessing the risk of primary graft dysfunction after lung transplantation
US9157123B2 (en) 2010-04-20 2015-10-13 The Johns Hopkins University Genetic amplification of IQGAP1 in cancer
US20110269142A1 (en) 2010-04-30 2011-11-03 President And Fellows Of Harvard College Clinical Method for Individualized Epithelial Cancer Screening Involving ERCC5 and IGF2R Genetic Testing and Gene-Environment Interactions
US20110312520A1 (en) 2010-05-11 2011-12-22 Veracyte, Inc. Methods and compositions for diagnosing conditions
US20190100809A1 (en) 2010-05-11 2019-04-04 Veracyte, Inc. Algorithms for disease diagnostics
CN102985819B (zh) 2010-07-09 2015-04-15 私募蛋白质体公司 肺癌生物标记及其用途
WO2012055565A1 (en) 2010-10-29 2012-05-03 Institut National De La Sante Et De La Recherche Medicale (Inserm) New marker of breast tumors from the luminal-b sybtype
JP6155194B2 (ja) 2010-11-17 2017-06-28 インターペース ダイアグノスティックス リミテッド ライアビリティ カンパニー 良性甲状腺新生物と悪性甲状腺新生物を区別するためのバイオマーカーとしてのmiRNA
US20130273543A1 (en) 2010-12-21 2013-10-17 Decode Genetics Ehf. Genetic variants useful for risk assessment of thyroid cancer
KR20190002733A (ko) * 2010-12-30 2019-01-08 파운데이션 메디신 인코포레이티드 종양 샘플의 다유전자 분석의 최적화
US20130303826A1 (en) 2011-01-11 2013-11-14 University Health Network Prognostic signature for oral squamous cell carcinoma
WO2012123972A1 (en) 2011-03-17 2012-09-20 Decode Genetics Ehf Genetic variants useful for risk assessment of thyroid cancer
US9585930B2 (en) 2011-03-20 2017-03-07 Trustees Of Boston University Therapeutic agent for emphysema and COPD
US8945829B2 (en) 2011-03-22 2015-02-03 Cornell University Distinguishing benign and malignant indeterminate thyroid lesions
EP2505664B1 (de) 2011-03-30 2014-12-03 Universität Leipzig Verfahren und Mittel zur Unterscheidung von gut- und bösartigen Tumorproben, insbesondere bei der FNAB
CA3120217A1 (en) * 2011-04-29 2012-11-01 Cancer Prevention And Cure, Ltd. Methods of identification and diagnosis of lung diseases using classification systems and kits thereof
US20120288860A1 (en) 2011-05-12 2012-11-15 George Mason University Differential gene expression for detecting and/or differentiating lung disease
WO2013006495A2 (en) 2011-07-01 2013-01-10 Dana-Farber Cancer Institute, Inc. Methods of predicting prognosis in cancer
KR20130017525A (ko) 2011-08-11 2013-02-20 주식회사 젠닥스 대장암, 유방암, 신장암 또는 갑상선암의 조기 진단을 위한 바이오 마커 및 이의 용도
US20150080243A1 (en) 2011-09-01 2015-03-19 Allegro Diagnostics Corp. Methods and compositions for detecting cancer based on mirna expression profiles
WO2013049152A2 (en) 2011-09-26 2013-04-04 Allegro Diagnostics Corp. Methods for evaluating lung cancer status
US20140243240A1 (en) 2011-10-26 2014-08-28 Georgetown University microRNA EXPRESSION PROFILING OF THYROID CANCER
WO2013063544A1 (en) 2011-10-27 2013-05-02 Asuragen, Inc. Mirnas as diagnostic biomarkers to distinguish benign from malignant thyroid tumors
US10036069B2 (en) 2011-11-18 2018-07-31 University of Pittsburgh—of the Commonwealth System of Higher Education Biomarkers for assessing idiopathic pulmonary fibrosis
US20140371096A1 (en) 2011-12-09 2014-12-18 The Johns Hopkins University Compositions and methods for characterizing thyroid neoplasia
EP2788771A1 (de) 2011-12-09 2014-10-15 Pronota NV Ltbp2 als biomarker für lungenläsion
WO2013086429A2 (en) 2011-12-09 2013-06-13 Veracyte, Inc. Methods and compositions for classification of samples
WO2013086522A1 (en) 2011-12-10 2013-06-13 Veracyte, Inc. Methods and compositions for sample identification
WO2013088457A1 (en) 2011-12-13 2013-06-20 Decode Genetics Ehf Genetic variants useful for risk assessment of thyroid cancer
CA2799163A1 (en) 2011-12-18 2013-06-18 20/20 Genesystems, Inc. Methods and algorithms for aiding in the detection of cancer
US20130184999A1 (en) 2012-01-05 2013-07-18 Yan Ding Systems and methods for cancer-specific drug targets and biomarkers discovery
EP2823306A4 (de) 2012-03-09 2015-11-11 Caris Life Sciences Switzerland Holdings Gmbh Biomarkerzusammensetzungen und verfahren
US20150038376A1 (en) 2012-03-15 2015-02-05 Qiagen Sciences Llc Thyroid cancer biomarker
KR102197524B1 (ko) * 2012-03-27 2020-12-31 제넨테크, 인크. 특발성 폐 섬유증의 예후, 진단 및 치료 방법
JP2013212052A (ja) 2012-03-30 2013-10-17 Yale Univ Krasバリアントおよび腫瘍生物学
AU2013243947A1 (en) * 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
EP2841603A4 (de) 2012-04-26 2016-05-25 Allegro Diagnostics Corp Verfahren zur beurteilung eines lungenkrebszustands
WO2013169858A1 (en) 2012-05-08 2013-11-14 The Broad Institute, Inc. Diagnostic and treatment methods in patients having or at risk of developing resistance to cancer therapy
WO2013177060A2 (en) 2012-05-20 2013-11-28 Trustees Of Boston University Methods and systems for monitoring, diagnosing, and treating chronic obstructive polmonary disease
RU2641975C2 (ru) 2012-06-08 2018-01-23 Конинклейке Филипс Н.В. Способ и система для наблюдения за функцией легких пациента
WO2013190092A1 (en) 2012-06-21 2013-12-27 Philip Morris Products S.A. Gene signatures for copd diagnosis
JP6581502B2 (ja) 2012-08-20 2019-09-25 ザ ユナイテッド ステイツ オブ アメリカ, アズ リプレゼンテッド バイ ザ セクレタリー, デパートメント オブ ヘルス アンド ヒューマン サービシーズ 初期段階の肺がんにおける予後指標としてのタンパク質コーディング遺伝子及び非コーディング遺伝子の発現
US9994907B2 (en) 2012-09-20 2018-06-12 Genomedx Biosciences, Inc. Thyroid cancer diagnostics
EP2906712A1 (de) 2012-10-10 2015-08-19 Stichting Het Nederlands Kanker Instituut- Antoni van Leeuwenhoek Ziekenhuis Verfahren und mittel zur vorhersage der resistenz gegen eine krebsbehandlung
US20140121126A1 (en) 2012-10-25 2014-05-01 Memorial Sloan-Kettering Cancer Center Methods of detecting axl and gas6 in cancer patients
US20140143188A1 (en) 2012-11-16 2014-05-22 Genformatic, Llc Method of machine learning, employing bayesian latent class inference: combining multiple genomic feature detection algorithms to produce an integrated genomic feature set with specificity, sensitivity and accuracy
WO2014085434A1 (en) 2012-11-27 2014-06-05 Pontificia Universidad Catolica De Chile Compositions and methods for diagnosing thyroid tumors
CA2893745A1 (en) 2012-12-04 2014-06-12 Caris Mpi, Inc. Molecular profiling for cancer
AU2013358890A1 (en) 2012-12-14 2015-06-04 Mindera Corporation Methods and devices for detection and acquisition of biomarkers
US20140220006A1 (en) 2013-02-01 2014-08-07 Meso Scale Technologies, Llc Lung cancer biomarkers
CA2902871A1 (en) 2013-03-12 2014-10-09 Cepheid Methods of detecting cancer
WO2014186036A1 (en) 2013-03-14 2014-11-20 Allegro Diagnostics Corp. Methods for evaluating copd status
BR112015023699A2 (pt) * 2013-03-15 2017-07-18 Intermune Inc método de determinação de um nível de expressão de uma proteína marcadora de doença pulmonar fibrótica, método de determinação se um indivíduo tem ou está em risco de desenvolvimento de uma doença pulmonar fibrótica, método de determinação se um paciente de doença pulmonar fibrótica está em risco de progressão da doença pulmonar fibrótica, método de determinação de uma atividade de doença pulmonar fibrótica em um paciente, e método de tratamento de uma doença pulmonar fibrótica em um indivíduo em necessidade do mesmo
WO2014160645A2 (en) 2013-03-15 2014-10-02 Biotheranostics, Inc. Neuroendocrine tumors
KR20150122731A (ko) 2013-03-15 2015-11-02 펀다시오 인스티튜트 드 르세르카 바이오메디카 (아이알비 바르셀로나) 암 전이의 예후 및 치료 방법
US20140378425A1 (en) 2013-03-15 2014-12-25 Veracyte, Inc. Biomarkers for diagnosis of lung diseases and methods of use thereof
BR112015022490A2 (pt) 2013-03-15 2017-07-18 Veracyte Inc métodos e composições para classificação de amostras
EP2986736B1 (de) 2013-04-17 2019-09-18 Life Technologies Corporation Genfusionen und genvarianten im zusammenhang mit krebs
AU2014262450A1 (en) 2013-05-10 2015-12-03 Memorial Sloan Kettering Cancer Center Lipid scavenging in Ras cancers
KR102310799B1 (ko) 2013-11-07 2021-10-12 미디얼 리서치 리미티드 폐암 위험 평가 방법 및 시스템
PL406033A1 (pl) 2013-11-14 2015-05-25 Warszawski Uniwersytet Medyczny Sposób diagnozowania raka brodawkowatego tarczycy, zastosowanie markera mikroRNA do diagnozowania nowotworu tarczycy, oceny stopnia zaawansowania choroby oraz oceny podatności pacjenta i/lub choroby na zaproponowane leczenie oraz zawierający takie markery zestaw diagnostyczny
US9708667B2 (en) 2014-05-13 2017-07-18 Rosetta Genomics, Ltd. MiRNA expression signature in the classification of thyroid tumors
AU2015289758B2 (en) 2014-07-14 2021-09-09 Veracyte, Inc. Methods for evaluating lung cancer status
EP3770274A1 (de) 2014-11-05 2021-01-27 Veracyte, Inc. Systeme und verfahren zur diagnose von idiopathischer pulmonaler fibrose auf transbronchialen biopsien mit maschinellem lernen und hochdimensionalen transkriptionsdaten
WO2016094330A2 (en) 2014-12-08 2016-06-16 20/20 Genesystems, Inc Methods and machine learning systems for predicting the liklihood or risk of having cancer
AU2016226253A1 (en) 2015-03-04 2017-09-21 Veracyte, Inc. Methods for assessing the risk of disease occurrence or recurrence using expression level and sequence variant information
US20170127976A1 (en) 2015-06-11 2017-05-11 Michael Phillips Method and apparatus for identification of biomarkers in breath and methods of usng same for prediction of lung cancer
GB2557818A (en) 2015-09-25 2018-06-27 Veracyte Inc Methods and compositions that utilize transciptome sequencing data in machine learning-based classification
EP3455381A4 (de) 2016-05-12 2020-03-04 Trustees of Boston University Nasenepithelgenexpressionssignatur und klassifikator zur vorhersage von lungenkrebs
WO2018009915A1 (en) 2016-07-08 2018-01-11 Trustees Of Boston University Gene expression-based biomarker for the detection and monitoring of bronchial premalignant lesions
AU2017324949B2 (en) 2016-09-07 2024-05-23 Veracyte, Inc. Methods and systems for detecting usual interstitial pneumonia
WO2018223066A1 (en) 2017-06-02 2018-12-06 Veracyte, Inc. Methods and systems for identifying or monitoring lung disease
US11217329B1 (en) 2017-06-23 2022-01-04 Veracyte, Inc. Methods and systems for determining biological sample integrity
GB2581584A (en) 2017-07-27 2020-08-26 Veracyte Inc Genomic sequencing classifier

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10526655B2 (en) 2013-03-14 2020-01-07 Veracyte, Inc. Methods for evaluating COPD status
US11976329B2 (en) * 2013-03-15 2024-05-07 Veracyte, Inc. Methods and systems for detecting usual interstitial pneumonia
US11639527B2 (en) 2014-11-05 2023-05-02 Veracyte, Inc. Methods for nucleic acid sequencing
US11753628B2 (en) 2017-01-23 2023-09-12 Regeneron Pharmaceuticals, Inc. HSD17B13 variants and uses thereof
US11845963B2 (en) 2017-01-23 2023-12-19 Regeneron Pharmaceuticals, Inc. HSD17B13 variants and uses thereof
US11479802B2 (en) 2017-04-11 2022-10-25 Regeneron Pharmaceuticals, Inc. Assays for screening activity of modulators of members of the hydroxy steroid (17-beta) dehydrogenase (HSD17B) family
US11702700B2 (en) 2017-10-11 2023-07-18 Regeneron Pharmaceuticals, Inc. Inhibition of HSD17B13 in the treatment of liver disease in patients expressing the PNPLA3 I148M variation
CN110819700A (zh) * 2018-08-10 2020-02-21 杭州米天基因科技有限公司 一种构建肺部小结节计算机辅助检测模型的方法
WO2022261437A3 (en) * 2021-06-11 2023-01-05 Acceleron Pharma Inc. Actrii proteins and uses thereof
US11670417B2 (en) * 2021-10-05 2023-06-06 Neumora Therapeutics, Inc. Machine learning systems for processing multi-modal patient data

Also Published As

Publication number Publication date
CN114606309A (zh) 2022-06-10
CN107206043A (zh) 2017-09-26
WO2016073768A1 (en) 2016-05-12
JP2018504138A (ja) 2018-02-15
US20240110242A1 (en) 2024-04-04
EP3215170A1 (de) 2017-09-13
US11639527B2 (en) 2023-05-02
US20210079471A1 (en) 2021-03-18
EP3770274A1 (de) 2021-01-27
EP3215170A4 (de) 2018-04-25
US20210324464A1 (en) 2021-10-21
JP2021164484A (ja) 2021-10-14
JP7356788B2 (ja) 2023-10-05

Similar Documents

Publication Publication Date Title
US11639527B2 (en) Methods for nucleic acid sequencing
EP2971128B1 (de) Biomarker zur diagnose von lungenerkrankungen und verfahren zur verwendung davon
US20210355524A1 (en) Methods for Nucleic Acid Sequencing
EP2776830B1 (de) Verfahren zur vorhersage der brustkrebsprognose
EP3179393B1 (de) Genexpressionsprofilalgorithmus und test zur bestimmung der prognose von prostatakrebs
WO2019018440A1 (en) HUMAN COLON CELL ATLAS IN GOOD HEALTH AND WITH HEMORRHAGIC RECTO-COLITIS
US20210115519A1 (en) Methods and kits for diagnosis and triage of patients with colorectal liver metastases
EP3044333A1 (de) Verfahren und systeme zur analyse einer organtransplantation
US20130317083A1 (en) Non-coding transcripts for determination of cellular states
US11814687B2 (en) Methods for characterizing bladder cancer
US20190367964A1 (en) Dissociation of human tumor to single cell suspension followed by biological analysis
US11976329B2 (en) Methods and systems for detecting usual interstitial pneumonia
US20230265522A1 (en) Multi-gene expression assay for prostate carcinoma

Legal Events

Date Code Title Description
AS Assignment

Owner name: VERACYTE, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KENNEDY, GIULIA C.;HUANG, JING;CHOI, YOONHA;AND OTHERS;SIGNING DATES FROM 20150512 TO 20150526;REEL/FRAME:043496/0735

AS Assignment

Owner name: VERACYTE, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:MADRYN HEALTH PARTNERS, LP;REEL/FRAME:044043/0880

Effective date: 20171106

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: VERACYTE, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KIM, SU YEON;DIGGANS, JAMES;PANKRATZ, DAN;AND OTHERS;SIGNING DATES FROM 20150512 TO 20150526;REEL/FRAME:053595/0627