WO2011085163A2 - Protein markers for lung cancer detection and methods of using thereof - Google Patents

Protein markers for lung cancer detection and methods of using thereof Download PDF

Info

Publication number
WO2011085163A2
WO2011085163A2 PCT/US2011/020463 US2011020463W WO2011085163A2 WO 2011085163 A2 WO2011085163 A2 WO 2011085163A2 US 2011020463 W US2011020463 W US 2011020463W WO 2011085163 A2 WO2011085163 A2 WO 2011085163A2
Authority
WO
WIPO (PCT)
Prior art keywords
lung cancer
amounts
subject
mir
equal
Prior art date
Application number
PCT/US2011/020463
Other languages
French (fr)
Other versions
WO2011085163A3 (en
Inventor
Steven M. Dubinett
Brian K. Gardner
David Elashoff
Kostyantyn Krysan
Original Assignee
The Regents Of The University Of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Regents Of The University Of California filed Critical The Regents Of The University Of California
Priority to US13/520,660 priority Critical patent/US20120315641A1/en
Priority to EP11732181.0A priority patent/EP2521920A4/en
Publication of WO2011085163A2 publication Critical patent/WO2011085163A2/en
Publication of WO2011085163A3 publication Critical patent/WO2011085163A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57488Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds identifable in body fluids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression 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
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/60Complex ways of combining multiple protein biomarkers for diagnosis

Definitions

  • the present invention generally relates to protein markers and methods for the detection of lung cancer.
  • Lung cancer is the leading cause of death from cancer in the United States.
  • US 20090068685 does not disclose anything about any differential expression patterns between lung cancer subjects vs. subjects at high risk for lung cancer (who may or may not have indeterminate pulmonary nodules).
  • the biomarker panels disclosed in US 20090068685 cannot be used to accurately determine whether a subject at high risk for lung cancer actually has lung cancer. This is because different factors, such as smoking, cause one to have different biomarker expression profiles.
  • the differential expression profile of one set of factors e.g. asthma
  • the present invention provides methods of detecting, diagnosing, or
  • categorizing a subject as having a lung cancer which comprises determining the amounts of at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, in a blood, serum or plasma sample from the subject, and determining whether the amounts are indicative of the lung cancer.
  • logistic regression analysis is used to calculate a predicted probability of the lung cancer.
  • the lung cancer is non-small cell lung cancer.
  • the amounts of VEGF, GCSF, MIG and RANTES are determined and logistic regression analysis is used to calculate a predicted probability of the lung cancer.
  • the lung cancer is stage I non-small cell lung cancer.
  • the amounts of IL-2, IL-3 and MDC are determined and logistic regression analysis is used to calculate a predicted probability of the lung cancer.
  • the subject is categorized as being at high risk for lung cancer.
  • the subject smokes or has smoked at least 20 packs of cigarettes, preferably at least 30 packs of cigarettes per year and is at least 35 years of age, preferably at least 45 years of age.
  • the amounts are indicative of the lung cancer where the predicted probability is greater than or equal to 0.6, preferably greater than or equal to 0.7, more preferably greater than or equal to 0.8, most preferably greater than or equal to 0.9.
  • the amounts are not indicative of the lung cancer where the predicted probability is less than or equal to 0.4, preferably less than or equal to 0.3, more preferably less than or equal to 0.2, most preferably less than or equal to 0.1.
  • the methods further comprise determining the amounts of one or more of the following protein biomarkers: CXCL1 (GROa), CXCL3 (GROy), CXCL5 (ENA-78), CCL1 (1309), CXCL11 (I-TAC), CXCL12 (SDF-1), CCL3 (MIP-la), CCL4 (MIP- ⁇ ), CCL11 (eotaxin), CCL15 ( ⁇ ), CCL19 ( ⁇ 3 ⁇ ), IL-4, IL-6, IL-7, IL-10, IL-12B (p40), IL-12 (p70), IL-13, IL-15, IL-17, GM-CSF, INF- ⁇ , IL-l , IL- ⁇ , ILIRa, and TNFp, and determining whether the amounts are indicative of the lung cancer.
  • the methods further comprise determining the amounts of one or more of the following protein biomarkers: CXCL3 (GROy), CCL3 (MIP-la), CCL15 ( ⁇ ), IL-6, IL-la, and IL- 1 ⁇ , and determining whether the amounts are indicative of the lung cancer.
  • the methods further comprise determining the amounts of one or more miRNAs selected from the group consisting of miR-21, miR-25, miR-34a, miR-200c and miR-146b, and determining whether the amounts are indicative of the lung cancer.
  • the present invention provides methods of monitoring or treating a subject who is at high risk of having a lung cancer, who has the lung cancer or who has had the lung cancer, which comprises determining the amounts of at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL- 2, IL-3 and MDC, in a blood, serum or plasma sample from the subject, and treating the subject in accordance with the amounts.
  • the present invention provides devices which comprise at least three capture reagents immobilized on one or more substrates, which each capture reagent specifically binds one protein biomarker selected from the group consisting of: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC.
  • kits which comprise reagents for assaying the amounts of at least three of the protein biomarkers as disclosed herein, e.g. at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, packaged together.
  • Figure 1 is a ROC curve for a predictive profile of stages I-IV NSCLC vs. control (non-NSCLC) using 33 biomarkers. This model provides a sensitivity of
  • Figure 2 is a ROC curve for a predictive profile model of stages I-IV NSCLC vs. control (non-NSCLC) using 4 biomarkers, i.e. VEGF, GCSF, MIG and RANTES.
  • 4 biomarkers i.e. VEGF, GCSF, MIG and RANTES.
  • This model provides a sensitivity of 88%, a specificity of 79% and an AUC of 0.89.
  • Figure 3 is a ROC curve for a predictive profile model of stage I NSCLC vs. control (non-NSCLC) using 3 biomarkers, i.e. IL-2, IL-3 and MDC. This model provides a sensitivity of 97%, a specificity of 77%, and an AUC of 0.93.
  • the present invention provides a plurality of protein biomarkers which may be used in diagnostic methods and devices for detecting and/or diagnosing whether a subject has non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • the expression levels of some or all of the biomarkers in a peripheral blood sample of a subject may be used to detect and/or diagnose whether the subject has NSCLC.
  • the present invention also provides methods and devices for detecting and/or diagnosing whether a subject has NSCLC. As disclosed herein, the methods and devices of the present invention may be used to detect and/or diagnose whether a subject has stage I NSCLC.
  • the red top serum collection tubes were allowed to sit at room temperature for 30 minutes to allow the blood to clot.
  • the purple top tubes were centrifuged at 2,000 g for 10 minutes and the supernatant was collected. After incubation for clotting, the red top tubes were centrifuged at 2,000g for 10 minutes and the supernatant was collected. To insure sample integrity all samples were processed and the serum and plasma were aliquoted into 1.0, 0.5 and 0.1 milliliter aliquots, frozen and stored at -80°C within 2 hours of collection.
  • 40 candidate protein biomarkers that could be associated with lung cancer progression or whose levels may be altered as a result of tumorigenesis were selected.
  • the 40 candidate protein biomarkers are set forth in Table 1 as follows:
  • these protein biomarker candidates are not specific cancer markers and whose levels can be altered in conditions and disorders other than lung cancer, use of one or more of these 40 candidate biomarkers in a biomarker panel might not reliably allow the detection or diagnosis of lung cancer in a subject with sufficient specificity and sensitivity. Thus, in order to determine whether one or more of these candidate biomarkers have any utility in detecting or diagnosing lung cancer, the following experiments were conducted.
  • a bead-based multiplexed immunoassay was used. Specifically, a LUMINEX immunoassay system was used to determine the concentration of each of the 40 biomarkers in serum samples obtained from lung cancer patients and individuals at elevated risk for lung cancer based on their smoking history and age.
  • BSA/PBS was added to the 96-well filter plate and removed by vacuum filtration. Then the bead set for the assay was added, typically 3,000 beads per analyte per well. The buffer the beads were suspended in was removed by vacuum filtration, and the beads were washed twice with 100 ⁇ BSA/PBS before sample addition. Sample and standards (50 ⁇ per well) were then added to the wells of the filter plate and incubated for 2 hr on a shaker at room temperature. A detection antibody cocktail solution was made by mixing together biotinylated antibodies for each of the target analytes in the assay.
  • the beads were washed 3 times with 100 ⁇ BSA/PBS and then 25 ⁇ of detection antibody cocktail was added for 2 hours. The beads were then washed 3 times with 100 ⁇ BSA/PBS and incubated with 50 ⁇ of streptavidin-R-phycoerythrin reporter (4 ⁇ g/ml in BSA/PBS) for 30 minutes. The plate was then washed with 100 ⁇ BSA/PBS three times and the beads were resuspended in 125 ⁇ of BSA/PBS for reading in the LUMINEX analyzer. Biomarker concentration values were then determined by an 8 point standard calibration curve using methods known in the art.
  • sample groups control and cancer
  • sample groups were randomized across the assay plates.
  • all samples were run in triplicate, and these replicates were also randomized across the assay plates.
  • sample groups were not processed separately, but samples and controls were instead processed together, so they were all treated in the same manner. This prevents processing errors from affecting specific groups of samples.
  • reference standards on each assay plate may be included so results can be normalized from plate to plate and for assays run on different days. Antibodies and assay reagents known in the art were used. Because of potential lot-to-lot variability of protein standards and antibodies, each lot of reagents used in the immunoassays may be standardized.
  • the 33 biomarkers are as follows: CXCL1 (GROa), CXCL3 (GROy), CXCL5 (ENA-78), CCLl (1309), CXCL9 (MIG), CXCLl l (I-TAC), CXCL12 (SDF- 1), CCL3 (MIP-la), CCL4 (MIP- ⁇ ), CCL5 (RANTES), CCL11 (eotaxin), CCLl 5 ( ⁇ ), CCLl 9 ( ⁇ 3 ⁇ ), CCL22 (MDC), IL-2, IL-3, IL-4, IL-6, IL-7, IL-10, IL- 12B (p40), IL-12 (p70), IL-13, IL-15, IL-17, GCSF, GM-CSF, INF- ⁇ , IL-l , IL- ⁇ , ILIR
  • the 21 biomarkers are as follows: CXCLl (GROa), CCL2 (MCP-1), CXCL9 (MIG), CCL3 (MIP-la), CCL4 (MIP- ⁇ ), CCL5 (RANTES), CCL15 ( ⁇ ), CCL22 (MDC), IL-2, IL-7, IL-10, IL-12B (p40), IL-12 p70, IL-13, IL-15, IL-17, GCSF, INF- ⁇ , IL- ⁇ , ILIRa, TNFp, and VEGF.
  • the first type is a logistic regression model using small subsets of the markers.
  • the second type combines the whole set (33) of significant markers (this was done for the all stages scenario).
  • Logistic regression models the log odd (or logit). The odds defined as the ratio of Pz/(1-P z ) where P z is the probability of cancer given the set of biomarkers.
  • a is the intercept term in the model
  • the ⁇ terms are the regression coefficient for the ith biomarker and the Xi is the value for the ith biomaker.
  • the unknown parameters a and the ⁇ are estimated by maximum likelihood using a method common to all generalized linear models as known in the art.
  • the maximum likelihood estimates were computed numerically by using iteratively reweighted least squares.
  • PROC LOGISTIC in the statistical software package SAS SAS Institute Inc., Cary, NC was to compute the estimates for the a and the ⁇ that are given in the tables below.
  • the same technique is employed to compute the estimate of the intercept (a) as for the biomarker coefficients ( ⁇ ).
  • b. The predicted probability of cancer from the model would then be:
  • the ROC curve was constructed for these two models by examining a number of cut-points of the predicted probabilities.
  • the sensitivity and specificity indicated below is based on finding the cut-point of the predicted probability that maximizes the sum of the sensitivity plus specificity (e.g. maximizing Youden's J statistic).
  • a panel consisting of only 4 biomarkers, i.e. VEGF, GCSF, MIG and RANTES, was used to create a predictive profile model of stages I-IV NSCLC vs. control (non-NSCLC). These biomarkers were combined together to compute predicted probability of cancer status based on logistic regression. For this case the releveant coefficients are provided in the table below.
  • Figure 2 is a ROC curve for the logistic regression model of stages I-IV NSCLC vs. control (non-NSCLC) using 4 biomarkers, i.e. VEGF, GCSF, MIG and RANTES. This model provides a sensitivity of 88%, a specificity of 79% and an AUC of 0.89.
  • Figure 3 is a ROC curve for a predictive profile model of stage I NSCLC vs. control (non-NSCLC) using 3 biomarkers, i.e. IL-2, IL-3 and MDC. This model provides a sensitivity of 97%, a specificity of 77%, and an AUC of 0.93.
  • each biomarker was categorized into high or low categories. This categorization was based on a biomarker specific cut-point which was the median value for that marker across the whole subject pool (NSCLC and controls). A summary score was then created by adding up the number of markers that were greater than their cut-point. This summary score was then used to create an ROC curve and the sensitivity and specificity for the summary score was assessed by identifying the value of the summary score which resulted in the maximum of the sum of the sensitivity and specificity.
  • each biomarker concentration was categorized as high or low based on a threshold computed for the given biomarker. This threshold was established based on the median of each biomarker across the combined subject set of NSCLC and high- risk controls.
  • an overall marker score which is the number of biomarkers higher than the median value for each specific marker, was computed for each sample. This median of each marker was the median value for the marker across the entire cohort (including the overall marker score input into a logistic regression model for computing an individual subject's cancer risk probability). Then the sensitivity, specificity and area under the ROC curve (AUC) of given panels of selected biomarkers were calculated using the cut-point that maximized Youden's J statistic (i.e.
  • the probability of lung cancer may be calculated using the biomarker concentration values obtained from a sample. For example, amounts of VEGF, GCSF, MIG and RANTES in a blood, plasma, or serum sample from a subject at high risk for lung cancer are determined and the biomarker concentration values are calculated. Then the regression coefficients and the intercept value for these 4 biomarkers are used to calculate the predicted probability of lung cancer. For example, the regression coefficients and the intercept value provided above are used along with the biomarker concentration values to obtain the predicted probability, Pz, above.
  • a Pz value near 0 or 0 indicates that the subject does not likely have lung cancer.
  • a Pz value near 1 or 1 indicates that the subject likely has lung cancer.
  • a Pz value of 0.9 indicates that the subject has a 90% likelihood of having lung cancer.
  • the predictive model is for determining the probability of stage I NSCLC, e.g. using the model employing IL-2, IL-3 and MDC
  • the amounts of IL-2, IL-3 and MDC in a blood, plasma, or serum sample from a subject at high risk for lung cancer are determined and the biomarker concentration values are calculated. Then the regression coefficients and the intercept value for the given biomarkers are used to calculate the predicted probability of stage I NSCLC.
  • a Pz value near 0 or 0 indicates that the subject does not likely have stage I NSCLC.
  • a Pz value near 1 or 1 indicates that the subject likely has stage I NSCLC. For example, a Pz value of 0.2 indicates that the subject has a 20% likelihood of having stage I NSCLC.
  • the methods of the present invention may be used to determine whether a high-risk subject should be subjected to further diagnostic procedures to detect lung cancer. For example, where the biomarker expression profile obtained from a subject is the same or substantially similar to a biomarker expression profile that is indicative of lung cancer, one may determine that the subject should undergo further diagnostic testing such as an imaging study, fiberoptic bronchoscopy, cytologic examination of materials obtained via endobronchial brushings,
  • bronchoalveolar lavage and endo- and transbronchial biopsies or a combination thereof.
  • the methods of the present invention may also be used to monitor lung cancer treatments and/or cancer progression/remission.
  • a biomarker expression profile that is the same or substantially similar to a biomarker expression profile that is indicative of a high risk subject that does not have lung cancer i.e. the biomarker expression profile changes from being the same or substantially similar to a biomarker expression profile that is indicative of lung cancer
  • the subject can then be treated based on the amounts of the biomarkers. For example, if the biomarker expression profile is indicative of lung cancer, the subject can them be subjected to one or more cancer treatments known in the art.
  • the methods of the present invention may be used to diagnose lung cancer or monitor a subject for lung cancer who exhibits an indeterminate pulmonary nodule.
  • a subject exhibits an indeterminate pulmonary nodule, but has a biomarker expression profile that is the same or substantially similar to a biomarker expression profile that is indicative of lung cancer
  • be subject may be categorized as having lung cancer, closely monitored for developing lung cancer, and or subjected to further diagnostic tests for lung cancer.
  • RNAs microRNAs
  • the expression levels of various microRNAs (miRNAs) in serum and/or plasma samples from lung cancer subjects and high-risk control subjects were measured. Specifically, the expression levels of a let-7f, miR-16, miR-17, miR-21, miR-24, miR-25, miR-34a, miR-106a, miR-125a-3p, miR-126*, miR-128, miR-146b-5p, miR-155, miR-199a, miR-200c, miR-221 and miR-222 were assayed in a subset of the serum samples that were used in the protein biomarker assays described above.
  • the accession numbers of each of the miRNAs are set forth in Table 3 as follows:
  • the methods and devices of the present invention employing some or all of the protein biomarkers as disclosed herein may be multiplexed with microRNA (miRNA) assays.
  • miRNA microRNA
  • the concentrations of a given set of protein biomarkers and the concentrations of a given set of miRNAs may be measured in a test serum and/or plasma sample of a subject and then the subject is diagnosed as having lung cancer based on the concentrations of the protein biomarkers and the miRNAs.
  • one or more miRNAs selected from the group consisting of miR-21, miR-25, miR-34a, miR-200c and miR-146b are assayed.
  • about 4-8 protein biomarkers and one or more of the miRNAs as described herein may be used to detect or diagnose the presence or absence of lung cancer in a subject.
  • the concentrations of CXCL3, CCL3, CCL15, IL-6, GMCSF, ILl , ILip, VEGF, miR-21, miR-25, miR- 34a, and miR-200c in a serum sample of a subject may be used to detect or diagnose the presence or absence of lung cancer, such as stage 1 NSCLC, in the subject.
  • the miRNA expression levels may be assayed using methods known in the art.
  • the following protocol can be used.
  • RNA is be isolated from 200 ⁇ of human serum using MIRNEASY kit (Qiagen, Valencia, CA) according to the modified manufacturer's protocol for the liquid samples. 200 ⁇ of serum is thawed on ice and mixed thoroughly by vortexing with 5 volumes of QIAZOL LYSIS REAGENT from the MIRNEASY miRNA isolation kit and is subsequently incubated at room temperature for 5 minutes. At this point, synthetic C.
  • elegans miRNAs cel-miR-39, cel-miR-54 and cel-miR-238 (synthesized by IDT, Coralville, IA) is added to the samples as a mixture of 25 fmol of each miRNA in a 5 ⁇ total volume using methods known in the art to serve as
  • RNA expression is determined by quantitative RT-PCR using Qiagen's MiSCRiPT platform. Briefly, 10 ⁇ of total RNA eluted from the MIRNAEASY column is polyadenylated in vitro and reversely transcribed utilizing MiSCRiPT REVERSE TRANSRIPTION KJT.
  • qPCR is performed using QUANTITECT SYBR GREEN mix and primers as recommended by the manufacturer. PCR reactions and data analysis is performed using ICYCLER and lQ5 software package (Bio-Rad, Hercules, CA) respectively. Data is normalized to the spike-in synthetic miRNA controls. All sample groups in the PCR experiments are run in triplicate and randomized to prevent experimental bias.
  • the methods and devices of the present invention employing some or all of the protein biomarkers, with or without one or more miRNAs, as disclosed herein may also be multiplexed with other diagnostic methods known in the art for detecting or diagnosing NSCLC and/or other cancers, such as imaging studies, fiberoptic bronchoscopies, cytologic examinations, bronchoalveolar lavage and endo- and transbronchial biopsies, transthoracic biopsies, exploratory thoracotomies, and the like.
  • the methods and devices of the present invention may be performed using whole blood samples.
  • the experiments described herein were performed using a specific high risk control group, i.e. former smokers at risk for lung cancer (> 30 pack years, age > 45, smoking cessation of at least 1 year)
  • the methods and devices described herein may be applied to other high risk subjects, e.g. current smokers, younger subjects, subjects who smoke or smoked less than 30, e.g. 20-29, packs per year, ceased smoking less than one year prior to being tested, or a combination thereof.
  • Devices according to the present invention comprise one or more substrates having capture reagents immobilized thereon, e.g. antibodies which specifically bind a given set of protein biomarkers and/or miRNAs and/or nucleic acid molecules which hybridize to a given set of miRNAs. After the substrate is contacted with a sample, the amount of each protein biomarker and/or miRNA captured by the capture reagent may be determined using methods known in the art.
  • Kits according to the present invention comprise reagents for assaying the amounts of at least three of the protein biomarkers as disclosed herein, e.g. at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, packaged together.
  • the kits may further comprise tools and devices for collecting and storing samples obtained from subjects.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Hospice & Palliative Care (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biotechnology (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • General Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Disclosed herein are methods, devices and kits for detecting, diagnosing, or categorizing a subject as having lung cancer. As disclosed herein, at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, are used to determine whether a subject at high-risk for lung cancer likely has lung cancer, such as stage I non-small cell lung cancer.

Description

PROTEIN MARKERS FOR LUNG CANCER DETECTION AND METHODS OF USING THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[01] This application claims the benefit of U.S. Application Serial No. 61/293,550, filed 8 January 2010, which is herein incorporated by reference in its entirety.
ACKNOWLEDGEMENT OF GOVERNMENT SUPPORT
[02] This invention was made with Government support under Grant Nos. CA
090338 and DA 016339, awarded by the National Institutes of Health. The
Government has certain rights in this invention.
[03] This work was also supported by the U.S. Department of Veterans Affairs, and the Federal Government has certain rights of this invention.
BACKGROUND OF THE INVENTION
1. FIELD OF THE INVENTION.
[04] The present invention generally relates to protein markers and methods for the detection of lung cancer.
2. DESCRIPTION OF THE RELATED ART.
[05] Lung cancer is the leading cause of death from cancer in the United States.
Currently, the overall five-year survival rate is only 14%, and this figure has not changed significantly over the last three decades. At time of clinical presentation, only about 25% of subjects have surgically resectable lung cancer. See Birring, et al. (2005) Thorax. 60(4):268-269. Moreover, subjects having pathologic stage IA lung cancers who undergo surgical resection only have a five-year survival rate of 67%. It is estimated that it can take up to 8 years for a lung carcinoma to reach clinical detection providing an opportunity for early detection.
[06] US 20090068685 discloses various biomarkers which are differentially
expressed among lung cancer subjects vs. asthma subjects and lung cancer subjects vs. normal subjects. Unfortunately, US 20090068685 does not disclose anything about any differential expression patterns between lung cancer subjects vs. subjects at high risk for lung cancer (who may or may not have indeterminate pulmonary nodules). As such, the biomarker panels disclosed in US 20090068685 cannot be used to accurately determine whether a subject at high risk for lung cancer actually has lung cancer. This is because different factors, such as smoking, cause one to have different biomarker expression profiles. The differential expression profile of one set of factors (e.g. asthma) can not be correlated to or suggest a differential expression profile of a different set of factors (exposure to cigarette smoke). In addition, the differential expression patterns of US 20090068685 cannot account for any similarities of biomarker expression patterns between high risk subjects and lung cancer subjects. Specifically, smoking causes chronic inflammation, deregulated cells, aberrant repair, increased product of cytokines and growth factors which are associated with the development of lung cancer. See Walser et al.(2008) Proc Am Thorac Soc 5(8):811-5; Auerbach et al. (1961) N Engl J Med 265:253-67; and Wistuba, II, (2007) Curr Mol Med 7(1):3-14. As such, it is unknown whether such biochemical and physiological effects will result in biomarker expression profiles which are indistinguishable between high risk subjects and subjects who have lung cancer.
[07] Thus, a need exists for diagnostics and methods for the early detection of lung cancer in high risk subjects, including the detection of subclinical lung cancer.
SUMMARY OF THE INVENTION
[08] The present invention provides methods of detecting, diagnosing, or
categorizing a subject as having a lung cancer which comprises determining the amounts of at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, in a blood, serum or plasma sample from the subject, and determining whether the amounts are indicative of the lung cancer. In some embodiments, logistic regression analysis is used to calculate a predicted probability of the lung cancer. In some embodiments, the lung cancer is non-small cell lung cancer. In some embodiments, the amounts of VEGF, GCSF, MIG and RANTES are determined and logistic regression analysis is used to calculate a predicted probability of the lung cancer. In some embodiments, the lung cancer is stage I non-small cell lung cancer. In some embodiments, the amounts of IL-2, IL-3 and MDC are determined and logistic regression analysis is used to calculate a predicted probability of the lung cancer. In some embodiments, the subject is categorized as being at high risk for lung cancer. In some embodiments, the subject smokes or has smoked at least 20 packs of cigarettes, preferably at least 30 packs of cigarettes per year and is at least 35 years of age, preferably at least 45 years of age. In some embodiments, the amounts are indicative of the lung cancer where the predicted probability is greater than or equal to 0.6, preferably greater than or equal to 0.7, more preferably greater than or equal to 0.8, most preferably greater than or equal to 0.9. In some
embodiments, the amounts are not indicative of the lung cancer where the predicted probability is less than or equal to 0.4, preferably less than or equal to 0.3, more preferably less than or equal to 0.2, most preferably less than or equal to 0.1.
[09] In some embodiments, the methods further comprise determining the amounts of one or more of the following protein biomarkers: CXCL1 (GROa), CXCL3 (GROy), CXCL5 (ENA-78), CCL1 (1309), CXCL11 (I-TAC), CXCL12 (SDF-1), CCL3 (MIP-la), CCL4 (MIP-Ιβ), CCL11 (eotaxin), CCL15 (ΜΙΡΙδ), CCL19 (ΜΙΡ3β), IL-4, IL-6, IL-7, IL-10, IL-12B (p40), IL-12 (p70), IL-13, IL-15, IL-17, GM-CSF, INF-γ, IL-l , IL-Ιβ, ILIRa, and TNFp, and determining whether the amounts are indicative of the lung cancer. In some embodiments, the methods further comprise determining the amounts of one or more of the following protein biomarkers: CXCL3 (GROy), CCL3 (MIP-la), CCL15 (ΜΙΡΙδ), IL-6, IL-la, and IL- 1β, and determining whether the amounts are indicative of the lung cancer. In some embodiments, the methods further comprise determining the amounts of one or more miRNAs selected from the group consisting of miR-21, miR-25, miR-34a, miR-200c and miR-146b, and determining whether the amounts are indicative of the lung cancer.
[10] In some embodiments, the present invention provides methods of monitoring or treating a subject who is at high risk of having a lung cancer, who has the lung cancer or who has had the lung cancer, which comprises determining the amounts of at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL- 2, IL-3 and MDC, in a blood, serum or plasma sample from the subject, and treating the subject in accordance with the amounts.
[11] In some embodiments, the present invention provides devices which comprise at least three capture reagents immobilized on one or more substrates, which each capture reagent specifically binds one protein biomarker selected from the group consisting of: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC.
[12] In some embodiments, the present invention provides kits which comprise reagents for assaying the amounts of at least three of the protein biomarkers as disclosed herein, e.g. at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, packaged together. [13] Both the foregoing general description and the following detailed description are exemplary and explanatory only and are intended to provide further explanation of the invention as claimed. The accompanying drawings are included to provide a further understanding of the invention and are incorporated in and constitute part of this specification, illustrate several embodiments of the invention, and together with the description serve to explain the principles of the invention.
DESCRIPTION OF THE DRAWINGS
[14] This invention is further understood by reference to the drawings wherein:
[15] Figure 1 is a ROC curve for a predictive profile of stages I-IV NSCLC vs. control (non-NSCLC) using 33 biomarkers. This model provides a sensitivity of
87%, a specificity of 78% and an AUC of 0.92.
[16] Figure 2 is a ROC curve for a predictive profile model of stages I-IV NSCLC vs. control (non-NSCLC) using 4 biomarkers, i.e. VEGF, GCSF, MIG and RANTES.
This model provides a sensitivity of 88%, a specificity of 79% and an AUC of 0.89.
[17] Figure 3 is a ROC curve for a predictive profile model of stage I NSCLC vs. control (non-NSCLC) using 3 biomarkers, i.e. IL-2, IL-3 and MDC. This model provides a sensitivity of 97%, a specificity of 77%, and an AUC of 0.93.
DETAILED DESCRIPTION OF THE INVENTION
[18] The present invention provides a plurality of protein biomarkers which may be used in diagnostic methods and devices for detecting and/or diagnosing whether a subject has non-small cell lung cancer (NSCLC). In particular, the expression levels of some or all of the biomarkers in a peripheral blood sample of a subject may be used to detect and/or diagnose whether the subject has NSCLC. Thus, the present invention also provides methods and devices for detecting and/or diagnosing whether a subject has NSCLC. As disclosed herein, the methods and devices of the present invention may be used to detect and/or diagnose whether a subject has stage I NSCLC.
[19] Blood samples were collected from 89 human subjects who were clinically diagnosed as having lung cancer (lung cancer subjects) and 56 human subjects at high-risk for obtaining lung cancer (high-risk control subjects). Of the 89 lung cancer subjects, 31 subjects had stage I NSCLC. The high-risk control subjects were former smokers (at least a year of cessation) ages 45 years or older who smoked > 30 packs of cigarettes per year prior to cessation. All control subjects underwent extensive screening to rule out pre-existing lung cancer, which was comprised of
comprehensive clinical laboratory studies (complete blood count, chemistry panel, and coagulation studies), spirometry, helical CT scans and LIFE (fluorescence) bronchoscopy with BAL and bronchial biopsies.
[20] All specimens utilized herein were collected from subjects who provided
informed consent utilizing forms approved by the UCLA IRB. All specimens were complemented with collection of general health and medical information, including clinical and pathologic stages, medication history and comorbidity. The control specimens were comprised of former smokers at risk for lung cancer (> 30 pack years, age > 45, smoking cessation of at least 1 year). All control subjects underwent extensive screening to rule out preexisting lung cancer which was included
comprehensive clinical laboratory studies (complete blood count, chemistry panel, and coagulation studies), spirometry, helical CT scans and LIFE (fluorescence) bronchoscopy with BAL and bronchial biopsies. All lung cancer and control blood samples were collected and processed utilizing a standardized collection and storage protocol that was based on the blood sample collection protocol utilized by the NIH/NHLBI sponsored Lung Health Study trial (LHS). This protocol is designed to standardize collection methods to minimize sample degradation and sample variability due to non-standardized sample processing. All blood utilized herein was collected into BD Vacutainer® blood collection tubes (BD Diagnostics, Franklin Lakes, NJ). The order of collection was red top first for serum collection followed by purple top for plasma. The red top serum collection tubes were allowed to sit at room temperature for 30 minutes to allow the blood to clot. The purple top tubes were centrifuged at 2,000 g for 10 minutes and the supernatant was collected. After incubation for clotting, the red top tubes were centrifuged at 2,000g for 10 minutes and the supernatant was collected. To insure sample integrity all samples were processed and the serum and plasma were aliquoted into 1.0, 0.5 and 0.1 milliliter aliquots, frozen and stored at -80°C within 2 hours of collection.
[21] 40 candidate protein biomarkers that could be associated with lung cancer progression or whose levels may be altered as a result of tumorigenesis were selected. The 40 candidate protein biomarkers are set forth in Table 1 as follows:
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
re erence n e r en re y.
[22] Since these protein biomarker candidates are not specific cancer markers and whose levels can be altered in conditions and disorders other than lung cancer, use of one or more of these 40 candidate biomarkers in a biomarker panel might not reliably allow the detection or diagnosis of lung cancer in a subject with sufficient specificity and sensitivity. Thus, in order to determine whether one or more of these candidate biomarkers have any utility in detecting or diagnosing lung cancer, the following experiments were conducted.
[23] To determine the concentration of these potential biomarkers in blood
samples, a bead-based multiplexed immunoassay was used. Specifically, a LUMINEX immunoassay system was used to determine the concentration of each of the 40 biomarkers in serum samples obtained from lung cancer patients and individuals at elevated risk for lung cancer based on their smoking history and age.
[24] Briefly, 100 μΐ of 1% bovine serum albumin/phosphate buffered saline
(BSA/PBS) was added to the 96-well filter plate and removed by vacuum filtration. Then the bead set for the assay was added, typically 3,000 beads per analyte per well. The buffer the beads were suspended in was removed by vacuum filtration, and the beads were washed twice with 100 μΐ BSA/PBS before sample addition. Sample and standards (50 μΐ per well) were then added to the wells of the filter plate and incubated for 2 hr on a shaker at room temperature. A detection antibody cocktail solution was made by mixing together biotinylated antibodies for each of the target analytes in the assay. Following the first incubation the beads were washed 3 times with 100 μΐ BSA/PBS and then 25 μΐ of detection antibody cocktail was added for 2 hours. The beads were then washed 3 times with 100 μΐ BSA/PBS and incubated with 50 μΐ of streptavidin-R-phycoerythrin reporter (4 μg/ml in BSA/PBS) for 30 minutes. The plate was then washed with 100 μΐ BSA/PBS three times and the beads were resuspended in 125 μΐ of BSA/PBS for reading in the LUMINEX analyzer. Biomarker concentration values were then determined by an 8 point standard calibration curve using methods known in the art. In order to prevent experimental artifacts from corrupting the data, all sample groups (control and cancer) were randomized across the assay plates. In addition, all samples were run in triplicate, and these replicates were also randomized across the assay plates. Thus, sample groups were not processed separately, but samples and controls were instead processed together, so they were all treated in the same manner. This prevents processing errors from affecting specific groups of samples. In order to minimize the effects of assay variability, reference standards on each assay plate may be included so results can be normalized from plate to plate and for assays run on different days. Antibodies and assay reagents known in the art were used. Because of potential lot-to-lot variability of protein standards and antibodies, each lot of reagents used in the immunoassays may be standardized.
Of the 40 biomarkers, 33 were determined to be statistically different between NSCLC for all stages and high-risk control samples (P<0.05) using the Wilcoxon rank sum test. The 33 biomarkers are as follows: CXCL1 (GROa), CXCL3 (GROy), CXCL5 (ENA-78), CCLl (1309), CXCL9 (MIG), CXCLl l (I-TAC), CXCL12 (SDF- 1), CCL3 (MIP-la), CCL4 (MIP-Ιβ), CCL5 (RANTES), CCL11 (eotaxin), CCLl 5 (ΜΙΡΙδ), CCLl 9 (ΜΙΡ3β), CCL22 (MDC), IL-2, IL-3, IL-4, IL-6, IL-7, IL-10, IL- 12B (p40), IL-12 (p70), IL-13, IL-15, IL-17, GCSF, GM-CSF, INF-γ, IL-l , IL-Ιβ, ILIRa, TNFP, and VEGF. [26] Of the 40 biomarkers, 21 were determined to be statistically different between stage 1 NSCLC samples and high-risk control samples (p<0.05) using the Wilcoxon rank sum test. The 21 biomarkers are as follows: CXCLl (GROa), CCL2 (MCP-1), CXCL9 (MIG), CCL3 (MIP-la), CCL4 (MIP-Ιβ), CCL5 (RANTES), CCL15 (ΜΙΡΙδ), CCL22 (MDC), IL-2, IL-7, IL-10, IL-12B (p40), IL-12 p70, IL-13, IL-15, IL-17, GCSF, INF-γ, IL-Ιβ, ILIRa, TNFp, and VEGF.
[27] Then two types of diagnostic models were constructed. The first type is a logistic regression model using small subsets of the markers. The second type combines the whole set (33) of significant markers (this was done for the all stages scenario).
[28] For the first type, subsets of the markers were chosen for the two scenarios (all stages or stage I) using stepwise logistic regression. This resulted in the 4 marker model for all stages and the 3 marker model for stage I. In these logistic regression models the markers were entered into the model as continuous variables (that is there was no marker specific cut-points or categorizations). The logistic regression outputs a predicted probability of cancer for each subject based on a weighted combination of the markers in the model.
[29] Specific details of logistic regression models: Logistic regression models the log odd (or logit). The odds defined as the ratio of Pz/(1-Pz) where Pz is the probability of cancer given the set of biomarkers. In a model with P number of predictors, the regression equation is: \n(odds) = a + βγΧγ + β2Χ2 + ... + βΡΧΡ + ε a. Where a is the intercept term in the model, the βί terms are the regression coefficient for the ith biomarker and the Xi is the value for the ith biomaker. The unknown parameters a and the βί (regression coefficients in the logistic regression model) are estimated by maximum likelihood using a method common to all generalized linear models as known in the art. The maximum likelihood estimates were computed numerically by using iteratively reweighted least squares. In this case, PROC LOGISTIC in the statistical software package SAS (SAS Institute Inc., Cary, NC) was to compute the estimates for the a and the βί that are given in the tables below. The same technique is employed to compute the estimate of the intercept (a) as for the biomarker coefficients (βί). b. The predicted probability of cancer from the model would then be:
Figure imgf000011_0001
c. Therefore, once the estimated regression coefficients are obtained, one can compute the sum of the products of the coefficients with their corresponding biomarker concentration values based on the formulation above to compute predicted probabilities.
[30] The ROC curve was constructed for these two models by examining a number of cut-points of the predicted probabilities. The sensitivity and specificity indicated below is based on finding the cut-point of the predicted probability that maximizes the sum of the sensitivity plus specificity (e.g. maximizing Youden's J statistic).
[31] In particular, a panel consisting of only 4 biomarkers, i.e. VEGF, GCSF, MIG and RANTES, was used to create a predictive profile model of stages I-IV NSCLC vs. control (non-NSCLC). These biomarkers were combined together to compute predicted probability of cancer status based on logistic regression. For this case the releveant coefficients are provided in the table below. Figure 2 is a ROC curve for the logistic regression model of stages I-IV NSCLC vs. control (non-NSCLC) using 4 biomarkers, i.e. VEGF, GCSF, MIG and RANTES. This model provides a sensitivity of 88%, a specificity of 79% and an AUC of 0.89.
Figure imgf000011_0002
The concentrations of IL-2, IL-3 and MDC in serum samples of stage I NSCLC subjects and high-risk control subjects were used to construct a logistic regression model of stage I NSCLC vs. control (non-NSCLC). Figure 3 is a ROC curve for a predictive profile model of stage I NSCLC vs. control (non-NSCLC) using 3 biomarkers, i.e. IL-2, IL-3 and MDC. This model provides a sensitivity of 97%, a specificity of 77%, and an AUC of 0.93.
Figure imgf000011_0003
Figure imgf000012_0001
[33] For the second type, which is a simple voting model, each biomarker was categorized into high or low categories. This categorization was based on a biomarker specific cut-point which was the median value for that marker across the whole subject pool (NSCLC and controls). A summary score was then created by adding up the number of markers that were greater than their cut-point. This summary score was then used to create an ROC curve and the sensitivity and specificity for the summary score was assessed by identifying the value of the summary score which resulted in the maximum of the sum of the sensitivity and specificity.
[34] In particular, in order to provide a predictive model for the presence of
NSCLC, each biomarker concentration was categorized as high or low based on a threshold computed for the given biomarker. This threshold was established based on the median of each biomarker across the combined subject set of NSCLC and high- risk controls. Next, an overall marker score, which is the number of biomarkers higher than the median value for each specific marker, was computed for each sample. This median of each marker was the median value for the marker across the entire cohort (including the overall marker score input into a logistic regression model for computing an individual subject's cancer risk probability). Then the sensitivity, specificity and area under the ROC curve (AUC) of given panels of selected biomarkers were calculated using the cut-point that maximized Youden's J statistic (i.e. the sum of the sensitivity + specificity) for the biomarker scores over all of the 33 significant biomarkers from the NSCLC all stages vs control. Based on the cut-off for the overall marker score a sensitivity of 87% and a specificity of 78% were obtained for all stages of lung cancer detection. Additionally, the AUC for this risk predictor is 0.92. The area under the ROC curve provides a single index that summarizes the diagnostic ability of the marker under consideration. The area under the curve is computed by performing numerical integration of the ROC curve. The computations were performed using the SAS statistical software package (SAS Institute Inc., Cary, NC). Figure 1 shows a ROC curve for this predictive model for NSCLC vs. control (non-NSCLC).
I I Coefficients I
Figure imgf000013_0001
[35] Thus, for a given set of biomarkers, once their regression coefficients and the intercept term are obtained, the probability of lung cancer may be calculated using the biomarker concentration values obtained from a sample. For example, amounts of VEGF, GCSF, MIG and RANTES in a blood, plasma, or serum sample from a subject at high risk for lung cancer are determined and the biomarker concentration values are calculated. Then the regression coefficients and the intercept value for these 4 biomarkers are used to calculate the predicted probability of lung cancer. For example, the regression coefficients and the intercept value provided above are used along with the biomarker concentration values to obtain the predicted probability, Pz, above. A Pz value near 0 or 0 indicates that the subject does not likely have lung cancer. A Pz value near 1 or 1 indicates that the subject likely has lung cancer. For example, a Pz value of 0.9 indicates that the subject has a 90% likelihood of having lung cancer.
[36] Similarly, where the predictive model is for determining the probability of stage I NSCLC, e.g. using the model employing IL-2, IL-3 and MDC, the amounts of IL-2, IL-3 and MDC in a blood, plasma, or serum sample from a subject at high risk for lung cancer are determined and the biomarker concentration values are calculated. Then the regression coefficients and the intercept value for the given biomarkers are used to calculate the predicted probability of stage I NSCLC. A Pz value near 0 or 0 indicates that the subject does not likely have stage I NSCLC. A Pz value near 1 or 1 indicates that the subject likely has stage I NSCLC. For example, a Pz value of 0.2 indicates that the subject has a 20% likelihood of having stage I NSCLC.
[37] Analysis of clinical specimens from stage I NSCLC subjects and high-risk control subjects revealed increased expression of pro-angiogenic and proinflammatory cytokines in the NSCLC subjects compared to high-risk control subjects and diminished expression of anti-angiogenic and anti -inflammatory cytokines in the NSCLC subjects. Based on these results, one or more additional protein biomarkers associated with anti-angiogenic and anti-inflammatory biochemical pathways, such as those set forth in Table 2 may be included in methods and devices according to the instant invention.
Figure imgf000013_0002
Figure imgf000014_0001
reference in their entiret y-
[38] Therefore, the methods of the present invention may be used to determine whether a high-risk subject should be subjected to further diagnostic procedures to detect lung cancer. For example, where the biomarker expression profile obtained from a subject is the same or substantially similar to a biomarker expression profile that is indicative of lung cancer, one may determine that the subject should undergo further diagnostic testing such as an imaging study, fiberoptic bronchoscopy, cytologic examination of materials obtained via endobronchial brushings,
bronchoalveolar lavage and endo- and transbronchial biopsies, or a combination thereof.
[39] The methods of the present invention may also be used to monitor lung cancer treatments and/or cancer progression/remission. For example, a biomarker expression profile that is the same or substantially similar to a biomarker expression profile that is indicative of a high risk subject that does not have lung cancer (i.e. the biomarker expression profile changes from being the same or substantially similar to a biomarker expression profile that is indicative of lung cancer) could be used to indicate that the given treatment was successful and/or remission. The subject can then be treated based on the amounts of the biomarkers. For example, if the biomarker expression profile is indicative of lung cancer, the subject can them be subjected to one or more cancer treatments known in the art. [40] The methods of the present invention may be used to diagnose lung cancer or monitor a subject for lung cancer who exhibits an indeterminate pulmonary nodule. For example, where a subject exhibits an indeterminate pulmonary nodule, but has a biomarker expression profile that is the same or substantially similar to a biomarker expression profile that is indicative of lung cancer, be subject may be categorized as having lung cancer, closely monitored for developing lung cancer, and or subjected to further diagnostic tests for lung cancer.
[41] In addition to assaying protein biomarkers, the expression levels of various microRNAs (miRNAs) in serum and/or plasma samples from lung cancer subjects and high-risk control subjects were measured. Specifically, the expression levels of a let-7f, miR-16, miR-17, miR-21, miR-24, miR-25, miR-34a, miR-106a, miR-125a-3p, miR-126*, miR-128, miR-146b-5p, miR-155, miR-199a, miR-200c, miR-221 and miR-222 were assayed in a subset of the serum samples that were used in the protein biomarker assays described above. The accession numbers of each of the miRNAs are set forth in Table 3 as follows:
Figure imgf000015_0001
Figure imgf000016_0001
[42] It was found that miR-21, miR-25, miR-34a and miR-200c were significantly differentially expressed between stage 1 NSCLC subjects and high-risk controls (p<0.05) and miR-146b gave a p value of <0.08. Thus, the methods and devices of the present invention employing some or all of the protein biomarkers as disclosed herein may be multiplexed with microRNA (miRNA) assays. For example, the concentrations of a given set of protein biomarkers and the concentrations of a given set of miRNAs may be measured in a test serum and/or plasma sample of a subject and then the subject is diagnosed as having lung cancer based on the concentrations of the protein biomarkers and the miRNAs. In some embodiments, one or more miRNAs selected from the group consisting of miR-21, miR-25, miR-34a, miR-200c and miR-146b are assayed. In some embodiments, about 4-8 protein biomarkers and one or more of the miRNAs as described herein may be used to detect or diagnose the presence or absence of lung cancer in a subject. For example, the concentrations of CXCL3, CCL3, CCL15, IL-6, GMCSF, ILl , ILip, VEGF, miR-21, miR-25, miR- 34a, and miR-200c in a serum sample of a subject may be used to detect or diagnose the presence or absence of lung cancer, such as stage 1 NSCLC, in the subject.
[43] In embodiments which include miRNA assays, the miRNA expression levels may be assayed using methods known in the art. For example, the following protocol can be used. RNA is be isolated from 200 μΐ of human serum using MIRNEASY kit (Qiagen, Valencia, CA) according to the modified manufacturer's protocol for the liquid samples. 200 μΐ of serum is thawed on ice and mixed thoroughly by vortexing with 5 volumes of QIAZOL LYSIS REAGENT from the MIRNEASY miRNA isolation kit and is subsequently incubated at room temperature for 5 minutes. At this point, synthetic C. elegans miRNAs cel-miR-39, cel-miR-54 and cel-miR-238 (synthesized by IDT, Coralville, IA) is added to the samples as a mixture of 25 fmol of each miRNA in a 5 μΐ total volume using methods known in the art to serve as
normalization controls. One volume (200 μΐ) of chloroform is then added to each sample. The resulting suspensions are vortexed for 15 seconds and spun for 15 minutes at 12000 g at 4°C. The aqueous phase is collected, mixed with 1.5 volume of 100% ethanol and passed through a column provided with the kit. The column is washed and RNA is eluted with 40 μΐ of elution buffer according to the manufacturer's protocol. miRNA expression is determined by quantitative RT-PCR using Qiagen's MiSCRiPT platform. Briefly, 10 μΐ of total RNA eluted from the MIRNAEASY column is polyadenylated in vitro and reversely transcribed utilizing MiSCRiPT REVERSE TRANSRIPTION KJT. qPCR is performed using QUANTITECT SYBR GREEN mix and primers as recommended by the manufacturer. PCR reactions and data analysis is performed using ICYCLER and lQ5 software package (Bio-Rad, Hercules, CA) respectively. Data is normalized to the spike-in synthetic miRNA controls. All sample groups in the PCR experiments are run in triplicate and randomized to prevent experimental bias.
[44] The methods and devices of the present invention employing some or all of the protein biomarkers, with or without one or more miRNAs, as disclosed herein may also be multiplexed with other diagnostic methods known in the art for detecting or diagnosing NSCLC and/or other cancers, such as imaging studies, fiberoptic bronchoscopies, cytologic examinations, bronchoalveolar lavage and endo- and transbronchial biopsies, transthoracic biopsies, exploratory thoracotomies, and the like.
[45] Although the experiments described herein were performed on plasma and serum samples, the methods and devices of the present invention may be performed using whole blood samples. In addition, although the experiments described herein were performed using a specific high risk control group, i.e. former smokers at risk for lung cancer (> 30 pack years, age > 45, smoking cessation of at least 1 year), the methods and devices described herein may be applied to other high risk subjects, e.g. current smokers, younger subjects, subjects who smoke or smoked less than 30, e.g. 20-29, packs per year, ceased smoking less than one year prior to being tested, or a combination thereof.
[46] Devices according to the present invention comprise one or more substrates having capture reagents immobilized thereon, e.g. antibodies which specifically bind a given set of protein biomarkers and/or miRNAs and/or nucleic acid molecules which hybridize to a given set of miRNAs. After the substrate is contacted with a sample, the amount of each protein biomarker and/or miRNA captured by the capture reagent may be determined using methods known in the art.
[47] Kits according to the present invention comprise reagents for assaying the amounts of at least three of the protein biomarkers as disclosed herein, e.g. at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, packaged together. The kits may further comprise tools and devices for collecting and storing samples obtained from subjects.
[48] To the extent necessary to understand or complete the disclosure of the present invention, all publications, patents, and patent applications mentioned herein are expressly incorporated by reference therein to the same extent as though each were individually so incorporated.
[49] Having thus described exemplary embodiments of the present invention, it should be noted by those skilled in the art that the within disclosures are exemplary only and that various other alternatives, adaptations, and modifications may be made within the scope of the present invention. Accordingly, the present invention is not limited to the specific embodiments as illustrated herein, but is only limited by the following claims.

Claims

What is claimed is:
1. A method of detecting, diagnosing, or categorizing a subject as having a lung cancer which comprises
determining the amounts of at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, in a blood, serum or plasma sample from the subject, and
determining whether the amounts are indicative of the lung cancer.
2. The method of claim 1, which further comprises using logistic regression analysis to calculate a predicted probability of the lung cancer.
3. The method of claim 1, wherein the lung cancer is non-small cell lung cancer.
4. The method of claim 3, wherein the amounts of VEGF, GCSF, MIG and RANTES are determined and logistic regression analysis is used to calculate a predicted probability of the lung cancer.
5. The method of claim 1, wherein the lung cancer is stage I non-small cell lung cancer.
6. The method of claim 5, wherein the amounts of IL-2, IL-3 and MDC are determined and logistic regression analysis is used to calculate a predicted probability of the lung cancer.
7. The method according to any one of claims 1-6, wherein the subject is categorized as being at high risk for lung cancer.
8. The method according to any one of claims 1-7, wherein the subject smokes or has smoked at least 20 packs of cigarettes, preferably at least 30 packs of cigarettes per year and is at least 35 years of age, preferably at least 45 years of age.
9. The method according to any one of claims 1-8, wherein the amounts are indicative of the lung cancer where the predicted probability is greater than or equal to 0.6, preferably greater than or equal to 0.7, more preferably greater than or equal to 0.8, most preferably greater than or equal to 0.9.
10. The method according to any one of claims 1-8, wherein the amounts are not indicative of the lung cancer where the predicted probability is less than or equal to 0.4, preferably less than or equal to 0.3, more preferably less than or equal to 0.2, most preferably less than or equal to 0.1.
11. The method according to any one of claims 1-10, which further comprises
determining the amounts of one or more of the following protein biomarkers: CXCL1 (GROa), CXCL3 (GROy), CXCL5 (ENA-78), CCL1 (1309), CXCL11 (I-TAC), CXCL12 (SDF-1), CCL3 (MIP-la), CCL4 (MIP-Ιβ), CCL11 (eotaxin), CCL15 (ΜΙΡΙδ), CCL19 (ΜΙΡ3β), IL-4, IL-6, IL-7, IL-10, IL-12B (p40), IL-12 (p70), IL-13, IL-15, IL-17, GM-CSF, INF-γ, IL-l , IL-Ιβ, ILIRa, TNFp, Lipocalin, LIF, sE-cadherin, CXCL7 (CTAP III), SCF, TGF-β, PDGF-BB, TRAIL, MMP-9, and MIF and
determining whether the amounts are indicative of the lung cancer.
12. The method according to any one of claims 1-10, which further comprises
determining the amounts of one or more of the following protein biomarkers: CXCL3 (GROy), CCL3 (MIP-l ), CCL15 (ΜΙΡΙδ), IL-6, IL-l , and IL-Ιβ, and
determining whether the amounts are indicative of the lung cancer.
13. The method according to any one of the preceding claims, which further comprises determining the amounts of one or more miRNAs selected from the group consisting of miR-21, miR-25, miR-34a, miR-200c and miR-146b, and
determining whether the amounts are indicative of the lung cancer.
14. A method of monitoring or treating a subject who is at high risk of having a lung cancer, who has the lung cancer or who has had the lung cancer, which comprises
determining the amounts of at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, in a blood, serum or plasma sample from the subject, and
treating the subject in accordance with the amounts.
15. A device which comprises at least three capture reagents immobilized on one or more substrates, which each capture reagent specifically binds one protein biomarker selected from the group consisting of: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC.
16. A kit which comprises reagents for assaying the amounts of at least three of the protein biomarkers as disclosed herein, e.g. at least three of the following protein biomarkers: VEGF, CGSF, MIG, RANTES, IL-2, IL-3 and MDC, packaged together.
17. The invention as disclosed herein.
PCT/US2011/020463 2010-01-08 2011-01-07 Protein markers for lung cancer detection and methods of using thereof WO2011085163A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/520,660 US20120315641A1 (en) 2010-01-08 2011-01-07 Protein Markers for Lung Cancer Detection and Methods of Using Thereof
EP11732181.0A EP2521920A4 (en) 2010-01-08 2011-01-07 Protein markers for lung cancer detection and methods of using thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US29355010P 2010-01-08 2010-01-08
US61/293,550 2010-01-08

Publications (2)

Publication Number Publication Date
WO2011085163A2 true WO2011085163A2 (en) 2011-07-14
WO2011085163A3 WO2011085163A3 (en) 2011-12-29

Family

ID=44306158

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/020463 WO2011085163A2 (en) 2010-01-08 2011-01-07 Protein markers for lung cancer detection and methods of using thereof

Country Status (3)

Country Link
US (1) US20120315641A1 (en)
EP (1) EP2521920A4 (en)
WO (1) WO2011085163A2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2607494A1 (en) * 2011-12-23 2013-06-26 Philip Morris Products S.A. Biomarkers for lung cancer risk assessment
US20130230877A1 (en) * 2011-12-21 2013-09-05 Integrated Diagnostics, Inc. Compositions, Methods and Kits for Diagnosis of Lung Cancer
WO2013096845A3 (en) * 2011-12-21 2013-09-19 Integrated Diagnostics, Inc. Methods for diagnosis of lung cancer
WO2013177422A1 (en) * 2012-05-24 2013-11-28 Allegheny Singer Research Institute System for ordering and arranging a data set
WO2014100717A3 (en) * 2012-12-21 2014-10-30 Integrated Diagnostics, Inc. Compositions, methods and kits for diagnosis of lung cancer
US9297805B2 (en) 2013-07-26 2016-03-29 Integrated Diagnostics, Inc. Compositions, methods and kits for diagnosis of lung cancer
CN105572380A (en) * 2014-11-06 2016-05-11 上海市东方医院 Application of IL-12 to diagnosis and treatment of non-small cell lung cancer (NSCLC) distant metastasis
WO2017027898A1 (en) * 2015-08-18 2017-02-23 University Of South Australia Novel cancer treatment involving modulation of il-3 activity
US10684290B2 (en) 2014-02-18 2020-06-16 Dignity Health LKB1 related diagnostics and treatments of cancer
EP3690444A4 (en) * 2017-09-26 2021-05-26 The University Of Tokyo Method for confirming prdm14 expression
US11913957B2 (en) 2011-12-21 2024-02-27 Biodesix, Inc. Compositions, methods and kits for diagnosis of lung cancer

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105209631A (en) 2013-03-14 2015-12-30 奥特拉西斯公司 A method for improving disease diagnosis using measured analytes
JP6952056B2 (en) 2016-01-22 2021-10-20 オートレイシーズ, インク.Otraces, Inc. Systems and methods to improve disease diagnosis
CN110114680A (en) * 2016-05-05 2019-08-09 佰欧迪塞克斯公司 For the composition of diagnosing, method and kit
JP6975697B2 (en) * 2018-09-11 2021-12-01 株式会社日立製作所 Cancer test processing device, cancer test system and cancer test processing method
WO2021011491A1 (en) * 2019-07-13 2021-01-21 Otraces Inc. Improving diagnosis for various diseases using tumor microenvironment active proteins

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4772559A (en) * 1985-10-10 1988-09-20 Monell Chemical Senses Center Method of detecting the presence of bronchogenic carcinoma by analysis of expired lung air
NZ337750A (en) * 1997-02-12 2001-08-31 Univ Michigan Protein markers for lung cancer and use thereof
WO2006080597A1 (en) * 2005-01-31 2006-08-03 Digital Genomics Inc. Markers for the diagnosis of lung cancer
TW200716764A (en) * 2005-05-02 2007-05-01 Genenews Inc Bladder cancer biomarkers and uses thereof
WO2007089911A2 (en) * 2006-01-30 2007-08-09 The Scripps Research Institute Methods for detection of circulating tumor cells and methods of diagnosis of cancer in a mammalian subject
US8207325B2 (en) * 2006-04-03 2012-06-26 Univ. of Copenhagen MicroRNA biomarkers for human breast and lung cancer
US8637080B2 (en) * 2007-06-28 2014-01-28 Osmotica Kereskedelmi és Szolgáltató, KFT Rupturing controlled release device comprising a subcoat
US7888051B2 (en) * 2007-09-11 2011-02-15 Cancer Prevention And Cure, Ltd. Method of identifying biomarkers in human serum indicative of pathologies of human lung tissues
EP3115469B1 (en) * 2007-11-19 2020-04-29 Celera Corporation Lung cancer markers and uses thereof
WO2009075799A2 (en) * 2007-12-05 2009-06-18 The Wistar Institute Of Anatomy And Biology Method for diagnosing lung cancers using gene expression profiles in peripheral blood mononuclear cells

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2521920A4 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9588127B2 (en) 2011-12-21 2017-03-07 Integrated Diagnostics, Inc. Compositions, methods and kits for diagnosis of lung cancer
US20130230877A1 (en) * 2011-12-21 2013-09-05 Integrated Diagnostics, Inc. Compositions, Methods and Kits for Diagnosis of Lung Cancer
WO2013096845A3 (en) * 2011-12-21 2013-09-19 Integrated Diagnostics, Inc. Methods for diagnosis of lung cancer
US9201044B2 (en) 2011-12-21 2015-12-01 Integrated Diagnostics, Inc. Compositions, methods and kits for diagnosis of lung cancer
US11913957B2 (en) 2011-12-21 2024-02-27 Biodesix, Inc. Compositions, methods and kits for diagnosis of lung cancer
US9304137B2 (en) 2011-12-21 2016-04-05 Integrated Diagnostics, Inc. Compositions, methods and kits for diagnosis of lung cancer
US10338074B2 (en) 2011-12-21 2019-07-02 Biodesix, Inc. Compositions, methods and kits for diagnosis of lung cancer
AU2012358246B2 (en) * 2011-12-21 2018-01-25 Integrated Diagnostics, Inc. Methods for diagnosis of lung cancer
EP2607494A1 (en) * 2011-12-23 2013-06-26 Philip Morris Products S.A. Biomarkers for lung cancer risk assessment
WO2013177422A1 (en) * 2012-05-24 2013-11-28 Allegheny Singer Research Institute System for ordering and arranging a data set
WO2014100717A3 (en) * 2012-12-21 2014-10-30 Integrated Diagnostics, Inc. Compositions, methods and kits for diagnosis of lung cancer
US11193935B2 (en) 2013-07-26 2021-12-07 Biodesix, Inc. Compositions, methods and kits for diagnosis of lung cancer
US9297805B2 (en) 2013-07-26 2016-03-29 Integrated Diagnostics, Inc. Compositions, methods and kits for diagnosis of lung cancer
US10684290B2 (en) 2014-02-18 2020-06-16 Dignity Health LKB1 related diagnostics and treatments of cancer
CN105572380A (en) * 2014-11-06 2016-05-11 上海市东方医院 Application of IL-12 to diagnosis and treatment of non-small cell lung cancer (NSCLC) distant metastasis
WO2017027898A1 (en) * 2015-08-18 2017-02-23 University Of South Australia Novel cancer treatment involving modulation of il-3 activity
EP3690444A4 (en) * 2017-09-26 2021-05-26 The University Of Tokyo Method for confirming prdm14 expression

Also Published As

Publication number Publication date
EP2521920A4 (en) 2013-06-05
WO2011085163A3 (en) 2011-12-29
US20120315641A1 (en) 2012-12-13
EP2521920A2 (en) 2012-11-14

Similar Documents

Publication Publication Date Title
WO2011085163A2 (en) Protein markers for lung cancer detection and methods of using thereof
US20200370127A1 (en) Biomarkers in Peripheral Blood Mononuclear Cells for Diagnosing or Detecting Lung Cancers
US11733249B2 (en) Methods and algorithms for aiding in the detection of cancer
EP2362942B1 (en) Biomarkers
CN107209184B (en) Marker combinations for diagnosing multiple infections and methods of use thereof
EP2812693B1 (en) A multi-biomarker-based outcome risk stratification model for pediatric septic shock
US11104953B2 (en) Septic shock endotyping strategy and mortality risk for clinical application
US10246748B2 (en) Biomarker combinations for colorectal tumors
WO2014074682A1 (en) Chronic obstructive pulmonary disease (copd) biomarkers and uses thereof
US20230251275A1 (en) Npersevere: biomarkers estimating baseline mortality risk for neonatal sepsis and necrotizing enterocolitis
JP7392224B2 (en) miRNA diagnostic biomarker for drug-induced interstitial pneumonia with diffuse alveolar injury
CN113358881B (en) Biomarker for NMOSD prediction or recurrence monitoring and application thereof
US20230167504A1 (en) Method for aiding detection of pancreatic cancer
US10436799B2 (en) Methods for diagnosing and treating subjects for pulmonary exacerbation
JP2022022709A (en) miRNA DIAGNOSIS BIOMARKER FOR SEVERE DRUG ERUPTION
TW202409297A (en) Molecular biomarkers and methods of analysis for acute diagnosis of kawasaki disease
WO2023079150A1 (en) Biomarker panel for diagnosing pulmonary dysfunction
CN118028453A (en) Application of CXCL1 and TNFRSF12A in AKI diagnosis
CN113999902A (en) Gene combination and application thereof in preparation of product for evaluating drug sensitivity of subject to golimumab
CN113897428A (en) Application of biomarker in judging sensitivity of ulcerative colitis patient to golimumab
CN113957139A (en) Application of human-derived gene in preparation of product for predicting sensitivity of patient to golimumab

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11732181

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2011732181

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011732181

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13520660

Country of ref document: US