1
|
Kammer MN, Deppen SA, Antic S, Jamshedur Rahman S, Eisenberg R, Maldonado F, Aldrich MC, Sandler KL, Landman B, Massion PP, Grogan EL. The impact of the lung EDRN-CVC on Phase 1, 2, & 3 biomarker validation studies. Cancer Biomark 2022; 33:449-465. [DOI: 10.3233/cbm-210382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The Early Detection Research Network’s (EDRN) purpose is to discover, develop and validate biomarkers and imaging methods to detect early-stage cancers or at-risk individuals. The EDRN is composed of sites that fall into four categories: Biomarker Developmental Laboratories (BDL), Biomarker Reference Laboratories (BRL), Clinical Validation Centers (CVC) and Data Management and Coordinating Centers. Each component has a crucial role to play within the mission of the EDRN. The primary role of the CVCs is to support biomarker developers through validation trials on promising biomarkers discovered by both EDRN and non-EDRN investigators. The second round of funding for the EDRN Lung CVC at Vanderbilt University Medical Center (VUMC) was funded in October 2016 and we intended to accomplish the three missions of the CVCs: To conduct innovative research on the validation of candidate biomarkers for early cancer detection and risk assessment of lung cancer in an observational study; to compare biomarker performance; and to serve as a resource center for collaborative research within the Network and partner with established EDRN BDLs and BRLs, new laboratories and industry partners. This report outlines the impact of the VUMC EDRN Lung CVC and describes the role in promoting and validating biological and imaging biomarkers.
Collapse
Affiliation(s)
- Michael N. Kammer
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen A. Deppen
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA
| | - Sanja Antic
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - S.M. Jamshedur Rahman
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rosana Eisenberg
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fabien Maldonado
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melinda C. Aldrich
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kim L. Sandler
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bennett Landman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Pierre P. Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric L. Grogan
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA
| |
Collapse
|
2
|
A model based on the quantification of complement C4c, CYFRA 21-1 and CRP exhibits high specificity for the early diagnosis of lung cancer. Transl Res 2021; 233:77-91. [PMID: 33618009 PMCID: PMC8931205 DOI: 10.1016/j.trsl.2021.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/26/2021] [Accepted: 02/13/2021] [Indexed: 12/15/2022]
Abstract
Lung cancer screening detects early-stage cancers, but also a large number of benign nodules. Molecular markers can help in the lung cancer screening process by refining inclusion criteria or guiding the management of indeterminate pulmonary nodules. In this study, we developed a diagnostic model based on the quantification in plasma of complement-derived fragment C4c, cytokeratin fragment 21-1 (CYFRA 21-1) and C-reactive protein (CRP). The model was first validated in two independent cohorts, and showed a good diagnostic performance across a range of lung tumor types, emphasizing its high specificity and positive predictive value. We next tested its utility in two clinically relevant contexts: assessment of lung cancer risk and nodule malignancy. The scores derived from the model were associated with a significantly higher risk of having lung cancer in asymptomatic individuals enrolled in a computed tomography (CT)-screening program (OR = 1.89; 95% CI = 1.20-2.97). Our model also served to discriminate between benign and malignant pulmonary nodules (AUC: 0.86; 95% CI = 0.80-0.92) with very good specificity (92%). Moreover, the model performed better in combination with clinical factors, and may be used to reclassify patients with intermediate-risk indeterminate pulmonary nodules into patients who require a more aggressive work-up. In conclusion, we propose a new diagnostic biomarker panel that may dictate which incidental or screening-detected pulmonary nodules require a more active work-up.
Collapse
|
3
|
Baseline and early changes in circulating Serum Amyloid A (SAA) predict survival outcomes in advanced non-small cell lung cancer patients treated with Anti-PD-1/PD-L1 monotherapy. Lung Cancer 2021; 158:1-8. [PMID: 34087538 DOI: 10.1016/j.lungcan.2021.05.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Systemic inflammation plays an important role in carcinogenesis and is associated with overall survival in patients with different cancer types, including those treated with immune checkpoint blockade (ICB). Serum Amyloid A (SAA) is an acute-phase protein and a marker of persistent inflammation. We hypothesized that circulating SAA may predict outcomes in advanced non-small cell lung (aNSCLC) patients treated with PD-1/PD-L1 ICB. MATERIALS AND METHODS This retrospective study included 91 aNSCLC patients who received anti-PD-(L)1 monotherapy in Sun Yat-sen University Cancer Center (Guangzhou, China) between August 2016 and June 2018. We examined the impact of circulating SAA at baseline and 8 (±2) weeks later on overall survival (OS). X-tile program was used to determine the cut-off values which optimized the significance of the split between Kaplan-Meier survival curves. Kaplan-Meier methodology and Cox regression analyses were conducted for survival analyses. RESULTS The optimal cut-off value of baseline SAA for OS stratification was 137.6 mg/L. In univariate analysis, both high level of baseline SAA (hazard ratio [HR], 2.76; 95% confidence interval [CI], 1.47-5.18; P = 0.002) and lack of early SAA descent (HR, 1.51; 95% CI, 1.11-2.06; P = 0.009) were significantly associated with inferior OS. In multivariate analysis, gender, smoking status, performance status, liver metastasis, neutrophil-to-lymphocyte ratio, baseline SAA and early changes in SAA independently predicted OS (all with P < 0.05). A combined baseline SAA ≥ 137.6 mg/L and without early SAA descent identified a small cohort with remarkably worse OS (median, 3.2 months). CONCLUSIONS Both high baseline and lack of early decline in circulating SAA are significantly associated with inferior outcomes in aNSCLC patients treated with PD-1/PD-L1 ICB. Combined these two SAA indexes provided improved risk stratification. The prognostic value of this simple, readily-available, and cost-effective biomarker warrants larger, prospective validation before definitive recommendation can be made.
Collapse
|
4
|
Abstract
The 2010's saw demonstration of the power of lung cancer screening to reduce mortality. However, with implementation of lung cancer screening comes the challenge of diagnosing millions of lung nodules every year. When compared to other cancers with widespread screening strategies (breast, colorectal, cervical, prostate, and skin), obtaining a lung nodule tissue biopsy to confirm a positive screening test remains associated with higher morbidity and cost. Therefore, non-invasive diagnostic biomarkers may have a unique opportunity in lung cancer to greatly improve the management of patients at risk. This review covers recent advances in the field of liquid biomarkers and computed tomographic imaging features, with special attention to new methods for combination of biomarkers as well as the use of artificial intelligence for the discrimination of benign from malignant nodules.
Collapse
Affiliation(s)
- Michael N Kammer
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.,Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pierre P Massion
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN, USA.,Medical Service, Tennessee Valley Healthcare Systems, Nashville Campus, Nashville, TN, USA
| |
Collapse
|
5
|
Noreldeen HAA, Liu X, Xu G. Metabolomics of lung cancer: Analytical platforms and their applications. J Sep Sci 2019; 43:120-133. [DOI: 10.1002/jssc.201900736] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/05/2019] [Accepted: 11/15/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Hamada A. A. Noreldeen
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences Dalian P. R. China
- University of Chinese Academy of Sciences Beijing P. R. China
- Marine Chemistry LabMarine Environment DivisionNational Institute of Oceanography and Fisheries Hurghada Egypt
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences Dalian P. R. China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences Dalian P. R. China
| |
Collapse
|
6
|
Chen S, Kang J, Xing Y, Zhao Y, Milton DK. Estimating large covariance matrix with network topology for high-dimensional biomedical data. Comput Stat Data Anal 2018. [DOI: 10.1016/j.csda.2018.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
7
|
Zhang C, Leng W, Sun C, Lu T, Chen Z, Men X, Wang Y, Wang G, Zhen B, Qin J. Urine Proteome Profiling Predicts Lung Cancer from Control Cases and Other Tumors. EBioMedicine 2018; 30:120-128. [PMID: 29576497 PMCID: PMC5952250 DOI: 10.1016/j.ebiom.2018.03.009] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/01/2018] [Accepted: 03/09/2018] [Indexed: 12/31/2022] Open
Abstract
Development of noninvasive, reliable biomarkers for lung cancer diagnosis has many clinical benefits knowing that most of lung cancer patients are diagnosed at the late stage. For this purpose, we conducted proteomic analyses of 231 human urine samples in healthy individuals (n = 33), benign pulmonary diseases (n = 40), lung cancer (n = 33), bladder cancer (n = 17), cervical cancer (n = 25), colorectal cancer (n = 22), esophageal cancer (n = 14), and gastric cancer (n = 47) patients collected from multiple medical centers. By random forest modeling, we nominated a list of urine proteins that could separate lung cancers from other cases. With a feature selection algorithm, we selected a panel of five urinary biomarkers (FTL: Ferritin light chain; MAPK1IP1L: Mitogen-Activated Protein Kinase 1 Interacting Protein 1 Like; FGB: Fibrinogen Beta Chain; RAB33B: RAB33B, Member RAS Oncogene Family; RAB15: RAB15, Member RAS Oncogene Family) and established a combinatorial model that can correctly classify the majority of lung cancer cases both in the training set (n = 46) and the test sets (n = 14–47 per set) with an AUC ranging from 0.8747 to 0.9853. A combination of five urinary biomarkers not only discriminates lung cancer patients from control groups but also differentiates lung cancer from other common tumors. The biomarker panel and the predictive model, when validated by more samples in a multi-center setting, may be used as an auxiliary diagnostic tool along with imaging technology for lung cancer detection. A case-control study of biomarker discovery for lung cancer diagnosis was conducted. Human urine profiles in control cases and cancers were characterized. A list of candidate biomarkers was nominated and evaluated. A panel of urinary biomarkers was established and tumor-specificity was evaluated.
Cancer diagnosis with a noninvasive method at the early stage of the disease is highly desirable. Here, we analyzed hundreds of human urine samples from healthy individuals, patients with benign pulmonary diseases, and 6 types of cancers by proteomics and developed a panel of five urinary proteins that can separate the lung cancer from benign pulmonary diseases as well as the other 5 cancers (bladder, cervical, colorectal, esophageal and gastric) with a good sensitivity and disease specificity. Further validation experiments with expanded sample numbers are required to investigate whether this method can be applied in a clinical setting for the diagnosis of lung cancer.
Collapse
Affiliation(s)
- Chunchao Zhang
- Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Wenchuan Leng
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing 102206, China; Joint Center for Translational Medicine, Tianjin, Baodi Hospital, Tianjin 301800, China
| | - Changqing Sun
- Joint Center for Translational Medicine, Tianjin, Baodi Hospital, Tianjin 301800, China
| | - Tianyuan Lu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing 102206, China
| | - Zhengang Chen
- Joint Center for Translational Medicine, Tianjin, Baodi Hospital, Tianjin 301800, China
| | - Xuebo Men
- Joint Center for Translational Medicine, Tianjin, Baodi Hospital, Tianjin 301800, China
| | - Yi Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing 102206, China; Joint Center for Translational Medicine, Tianjin, Baodi Hospital, Tianjin 301800, China; Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Guangshun Wang
- Joint Center for Translational Medicine, Tianjin, Baodi Hospital, Tianjin 301800, China.
| | - Bei Zhen
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing 102206, China; Joint Center for Translational Medicine, Tianjin, Baodi Hospital, Tianjin 301800, China.
| | - Jun Qin
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing 102206, China; Joint Center for Translational Medicine, Tianjin, Baodi Hospital, Tianjin 301800, China; Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Lu Z, Chen Y, Jing X, Hu C. Diagnostic accuracy of MALDI-TOF mass spectrometry for non-small cell lung cancer: a meta-analysis. Biomarkers 2018; 23:245-252. [PMID: 29264950 DOI: 10.1080/1354750x.2017.1420822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Zhaolian Lu
- School of Graduate, Second Military Medical University, Shanghai, China
- Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, China
| | - Yingjian Chen
- Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, China
| | - Xinyan Jing
- School of Graduate, Weifang Medical University, Weifang, China
| | - Chengjin Hu
- Department of Laboratory Medicine, General Hospital of Jinan Military Command Region, Jinan, China
| |
Collapse
|
9
|
|
10
|
Serum lipid profile discriminates patients with early lung cancer from healthy controls. Lung Cancer 2017; 112:69-74. [DOI: 10.1016/j.lungcan.2017.07.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 07/11/2017] [Accepted: 07/31/2017] [Indexed: 01/09/2023]
|
11
|
Stanford TE, Bagley CJ, Solomon PJ. Informed baseline subtraction of proteomic mass spectrometry data aided by a novel sliding window algorithm. Proteome Sci 2016; 14:19. [PMID: 27980460 PMCID: PMC5142289 DOI: 10.1186/s12953-016-0107-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 11/01/2016] [Indexed: 11/10/2022] Open
Abstract
Background Proteomic matrix-assisted laser desorption/ionisation (MALDI) linear time-of-flight (TOF) mass spectrometry (MS) may be used to produce protein profiles from biological samples with the aim of discovering biomarkers for disease. However, the raw protein profiles suffer from several sources of bias or systematic variation which need to be removed via pre-processing before meaningful downstream analysis of the data can be undertaken. Baseline subtraction, an early pre-processing step that removes the non-peptide signal from the spectra, is complicated by the following: (i) each spectrum has, on average, wider peaks for peptides with higher mass-to-charge ratios (m/z), and (ii) the time-consuming and error-prone trial-and-error process for optimising the baseline subtraction input arguments. With reference to the aforementioned complications, we present an automated pipeline that includes (i) a novel ‘continuous’ line segment algorithm that efficiently operates over data with a transformed m/z-axis to remove the relationship between peptide mass and peak width, and (ii) an input-free algorithm to estimate peak widths on the transformed m/z scale. Results The automated baseline subtraction method was deployed on six publicly available proteomic MS datasets using six different m/z-axis transformations. Optimality of the automated baseline subtraction pipeline was assessed quantitatively using the mean absolute scaled error (MASE) when compared to a gold-standard baseline subtracted signal. Several of the transformations investigated were able to reduce, if not entirely remove, the peak width and peak location relationship resulting in near-optimal baseline subtraction using the automated pipeline. The proposed novel ‘continuous’ line segment algorithm is shown to far outperform naive sliding window algorithms with regard to the computational time required. The improvement in computational time was at least four-fold on real MALDI TOF-MS data and at least an order of magnitude on many simulated datasets. Conclusions The advantages of the proposed pipeline include informed and data specific input arguments for baseline subtraction methods, the avoidance of time-intensive and subjective piecewise baseline subtraction, and the ability to automate baseline subtraction completely. Moreover, individual steps can be adopted as stand-alone routines. Electronic supplementary material The online version of this article (doi:10.1186/s12953-016-0107-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tyman E Stanford
- School of Mathematical Sciences, The University of Adelaide, North Terrace, Adelaide, 5005 Australia
| | - Christopher J Bagley
- School of Mathematical Sciences, The University of Adelaide, North Terrace, Adelaide, 5005 Australia
| | - Patty J Solomon
- School of Mathematical Sciences, The University of Adelaide, North Terrace, Adelaide, 5005 Australia
| |
Collapse
|
12
|
Broodman I, Lindemans J, van Sten J, Bischoff R, Luider T. Serum Protein Markers for the Early Detection of Lung Cancer: A Focus on Autoantibodies. J Proteome Res 2016; 16:3-13. [DOI: 10.1021/acs.jproteome.6b00559] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
| | | | | | - Rainer Bischoff
- Analytical
Biochemistry, Department of Pharmacy, University of Groningen, Antonius
Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | |
Collapse
|
13
|
Hayes SA, Haefliger S, Harris B, Pavlakis N, Clarke SJ, Molloy MP, Howell VM. Exhaled breath condensate for lung cancer protein analysis: a review of methods and biomarkers. J Breath Res 2016; 10:034001. [PMID: 27380020 DOI: 10.1088/1752-7155/10/3/034001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lung cancer is a leading cause of cancer-related deaths worldwide, and is considered one of the most aggressive human cancers, with a 5 year overall survival of 10-15%. Early diagnosis of lung cancer is ideal; however, it is still uncertain as to what technique will prove successful in the systematic screening of high-risk populations, with the strongest evidence currently supporting low dose computed tomography (LDCT). Analysis of exhaled breath condensate (EBC) has recently been proposed as an alternative low risk and non-invasive screening method to investigate early-stage neoplastic processes in the airways. However, there still remains a relative paucity of lung cancer research involving EBC, particularly in the measurement of lung proteins that are centrally linked to pathogenesis. Considering the ease and safety associated with EBC collection, and advances in the area of mass spectrometry based profiling, this technology has potential for use in screening for the early diagnosis of lung cancer. This review will examine proteomics as a method of detecting markers of neoplasia in patient EBC with a particular emphasis on LC, as well as discussing methodological challenges involving in proteomic analysis of EBC specimens.
Collapse
Affiliation(s)
- Sarah A Hayes
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, Northern Sydney Local Health District, St. Leonards, New South Wales, Australia. Sydney Medical School Northern, University of Sydney, New South Wales, Australia
| | | | | | | | | | | | | |
Collapse
|
14
|
Widlak P, Pietrowska M, Polanska J, Marczyk M, Ros-Mazurczyk M, Dziadziuszko R, Jassem J, Rzyman W. Serum mass profile signature as a biomarker of early lung cancer. Lung Cancer 2016; 99:46-52. [PMID: 27565913 DOI: 10.1016/j.lungcan.2016.06.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/12/2016] [Accepted: 06/11/2016] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Circulating molecular biomarkers of lung cancer may allow the pre-selection of candidates for computed tomography screening or increase its efficacy. We aimed to identify features of serum mass profile distinguishing individuals with early lung cancer from healthy participants of the lung cancer screening program. METHODS Blood samples were collected during a low-dose computed tomography (LD-CT) screening program performed by one institution (Medical University of Gdansk, Poland). MALDI-ToF mass spectrometry was used to characterize the low-molecular-weight (1000-14,000Da) serum fraction. The analysis comprised 95 patients with early stage lung cancer (including 30 screen-detected cases) and a matched group of 285 healthy controls. The cases were split into two independent cohorts (discovery and validation), analyzed separately 6 months apart. RESULTS Several molecular components of serum (putatively components of endogenous peptidome) discriminating patients with early lung cancer from controls were identified in a discovery cohort. This allowed building an effective cancer classifier as a model tuned to maximize negative predictive value, with an area under the curve (AUC) of 0.88, a negative predictive value of 100%, and a positive predictive value of 48%. However, the classifier performed worse in a validation cohort including independent sample sets (AUC 0.73, NPV 88% and PPV 30%). CONCLUSIONS We developed a serum mass profile-based signature identifying patients with early lung cancer. Although this marker has insufficient value as a stand-alone preselecting tool for LD-CT screening, its potential clinical usefulness in evaluation of indeterminate pulmonary nodules deserves further investigation.
Collapse
Affiliation(s)
- Piotr Widlak
- Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, ul. Wybrzeże Armii Krajowej 15, 44-100 Gliwice, Poland.
| | - Monika Pietrowska
- Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, ul. Wybrzeże Armii Krajowej 15, 44-100 Gliwice, Poland.
| | - Joanna Polanska
- Silesian University of Technology, ul. Akademicka 16, 44-100 Gliwice, Poland.
| | - Michal Marczyk
- Silesian University of Technology, ul. Akademicka 16, 44-100 Gliwice, Poland.
| | - Malgorzata Ros-Mazurczyk
- Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, ul. Wybrzeże Armii Krajowej 15, 44-100 Gliwice, Poland.
| | | | - Jacek Jassem
- Medical University of Gdańsk, ul. Dębinki 7, 80-211 Gdańsk, Poland.
| | - Witold Rzyman
- Medical University of Gdańsk, ul. Dębinki 7, 80-211 Gdańsk, Poland.
| |
Collapse
|
15
|
Yu H, Han Z, Wang Y, Xin H. The clonal evolution and therapeutic approaches of lung cancer. Cell Biochem Biophys 2015; 70:63-71. [PMID: 24639115 DOI: 10.1007/s12013-014-9910-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
According to the World Cancer Research Foundation, the newly diagnosed annual lung cancer cases all over the world are alarmingly high at 12.5 %. It also shows the highest mortality rate among all the cancer types. Nearly 225,000 new lung cancer patients are reported annually in the USA. The lung cancer cells also have very fast growth rates. As a result of this rapid proliferation rate, the lung cancer cells are sensitive to the available therapeutics like the radiation, surgical, or chemo therapy. Notwithstanding all the advances in the field of tumor biology, the mortality rate with lung cancer has remained significantly high. Precise and early diagnosis of the disease can be an important step in the proper and successful setting up of the treatment modalities. There are no comprehensive reviews available that discusses all the basic and updated aspects of lung cancer. This review focuses on the basic aspects of lung cancer like the etiology, risk factors, and clonal evolution. Exposure to smoking comes up as a single major environmental cause of the disease. The classification of lung cancer has also been discussed in detail based on immunohistochemistry. The existing therapeutic approaches as well as the upcoming modern day interventions have been discussed with their pros and cons. Recent techniques like molecular profiling can prove to be highly beneficial if properly standardized. With such advancements in therapy in conjunction with the updated diagnostics, there is a real hope in the treatment of lung cancer.
Collapse
Affiliation(s)
- Haixiang Yu
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033, Jilin, China
| | | | | | | |
Collapse
|
16
|
Birse CE, Lagier RJ, FitzHugh W, Pass HI, Rom WN, Edell ES, Bungum AO, Maldonado F, Jett JR, Mesri M, Sult E, Joseloff E, Li A, Heidbrink J, Dhariwal G, Danis C, Tomic JL, Bruce RJ, Moore PA, He T, Lewis ME, Ruben SM. Blood-based lung cancer biomarkers identified through proteomic discovery in cancer tissues, cell lines and conditioned medium. Clin Proteomics 2015; 12:18. [PMID: 26279647 PMCID: PMC4537594 DOI: 10.1186/s12014-015-9090-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 07/07/2015] [Indexed: 12/18/2022] Open
Abstract
Background Support for early detection of lung cancer has emerged from the National Lung Screening Trial (NLST), in which low-dose computed tomography (LDCT) screening reduced lung cancer mortality by 20 % relative to chest x-ray. The US Preventive Services Task Force (USPSTF) recently recommended annual screening for the high-risk population, concluding that the benefits (life years gained) outweighed harms (false positive findings, abortive biopsy/surgery, radiation exposure). In making their recommendation, the USPSTF noted that the moderate net benefit of screening was dependent on the resolution of most false-positive results without invasive procedures. Circulating biomarkers may serve as a valuable adjunctive tool to imaging. Results We developed a broad-based proteomics discovery program, integrating liquid chromatography/mass spectrometry (LC/MS) analyses of freshly resected lung tumor specimens (n = 13), lung cancer cell lines (n = 17), and conditioned media collected from tumor cell lines (n = 7). To enrich for biomarkers likely to be found at elevated levels in the peripheral circulation of lung cancer patients, proteins were prioritized based on predicted subcellular localization (secreted, cell-membrane associated) and differential expression in disease samples. 179 candidate biomarkers were identified. Several markers selected for further validation showed elevated levels in serum collected from subjects with stage I NSCLC (n = 94), relative to healthy smoker controls (n = 189). An 8-marker model was developed (TFPI, MDK, OPN, MMP2, TIMP1, CEA, CYFRA 21–1, SCC) which accurately distinguished subjects with lung cancer (n = 50) from high risk smokers (n = 50) in an independent validation study (AUC = 0.775). Conclusions Integrating biomarker discovery from multiple sample types (fresh tissue, cell lines and conditioned medium) has resulted in a diverse repertoire of candidate biomarkers. This unique collection of biomarkers may have clinical utility in lung cancer detection and diagnoses. Electronic supplementary material The online version of this article (doi:10.1186/s12014-015-9090-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charles E Birse
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Robert J Lagier
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - William FitzHugh
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Langone Medical Center, 530 First Avenue, New York, NY USA
| | - William N Rom
- Division of Pulmonary, Critical Care, and Sleep Medicine, NYU School of Medicine, New York, NY USA
| | - Eric S Edell
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN USA
| | - Aaron O Bungum
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN USA
| | - Fabien Maldonado
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN USA
| | - James R Jett
- Division of Oncology, National Jewish Health, Denver, CO USA
| | - Mehdi Mesri
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Erin Sult
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Elizabeth Joseloff
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Aiqun Li
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Jenny Heidbrink
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Gulshan Dhariwal
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Chad Danis
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Jennifer L Tomic
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Robert J Bruce
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Paul A Moore
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Tao He
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Marcia E Lewis
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| | - Steve M Ruben
- Celera employees during the course of these studies, Celera, 1311 Harbor Bay Parkway, Alameda, CA 94502 USA
| |
Collapse
|
17
|
Doseeva V, Colpitts T, Gao G, Woodcock J, Knezevic V. Performance of a multiplexed dual analyte immunoassay for the early detection of non-small cell lung cancer. J Transl Med 2015; 13:55. [PMID: 25880432 PMCID: PMC4335536 DOI: 10.1186/s12967-015-0419-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/25/2015] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES "PAULA's" test (Protein Assays Utilizing Lung cancer Analytes) is a novel multiplex immunoassay blood test that incorporates both tumor antigens and autoantibodies to determine the risk that lung cancer (LC) is present in individuals from a high-risk population. The test's performance characteristics were evaluated in a study using 380 retrospective clinical serum samples. METHODS PAULA's test is performed on the Luminex xMAP technology platform, and detects a panel of 3 tumor antigens (CEA, CA-125, and CYFRA 21-1) and 1 autoantibody marker (NY-ESO-1). A training set (n = 230) consisting of 115 confirmed diagnoses of non-small cell lung carcinoma (NSCLC) cases and 115 age- and smoking history-matched controls was used to develop the LC predictive model. Data from an independent matched validation set (n = 150) was then used to evaluate the model developed, and determine the ability of the test to distinguish NSCLC cases from controls. RESULTS The 4-biomarker panel was able to discriminate NSCLC cases from controls with 74% sensitivity, 80% specificity, and 0.81 AUC in the training set and with 77% sensitivity, 80% specificity, and 0.85 AUC in the independent validation set. The use of NY-ESO-1 autoantibodies substantially increased the overall sensitivity of NSCLC detection as compared to the 3 tumor markers alone. Overall, the multiplexed 4-biomarker panel assay demonstrated comparable performance to a previously employed 8-biomarker non-multiplexed assay. CONCLUSIONS These studies confirm the value of using a mixed panel of tumor antigens and autoantibodies in the early detection of NSCLC in high-risk individuals. The results demonstrate that the performance of PAULA's test makes it suitable for use as an aid to determine which high-risk patients need to be directed to appropriate noninvasive diagnostic follow-up testing, especially low-dose CT (LDCT).
Collapse
Affiliation(s)
- Victoria Doseeva
- 20/20 GeneSystems, 9430 Key West Avenue, Rockville, MD, 20850, USA.
| | - Tracey Colpitts
- Abbott Molecular Inc, 1300 E Touhy Avenue, Des Plaines, IL, 60018, USA.
| | - Grace Gao
- 20/20 GeneSystems, 9430 Key West Avenue, Rockville, MD, 20850, USA.
| | - Juliana Woodcock
- 20/20 GeneSystems, 9430 Key West Avenue, Rockville, MD, 20850, USA.
| | | |
Collapse
|
18
|
Lung Cancer Screening Beyond Low-Dose Computed Tomography: The Role of Novel Biomarkers. Lung 2014; 192:639-48. [DOI: 10.1007/s00408-014-9636-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/28/2014] [Indexed: 02/07/2023]
|
19
|
Li XJ, Hayward C, Fong PY, Dominguez M, Hunsucker SW, Lee LW, McLean M, Law S, Butler H, Schirm M, Gingras O, Lamontagne J, Allard R, Chelsky D, Price ND, Lam S, Massion PP, Pass H, Rom WN, Vachani A, Fang KC, Hood L, Kearney P. A blood-based proteomic classifier for the molecular characterization of pulmonary nodules. Sci Transl Med 2014; 5:207ra142. [PMID: 24132637 DOI: 10.1126/scitranslmed.3007013] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Each year, millions of pulmonary nodules are discovered by computed tomography and subsequently biopsied. Because most of these nodules are benign, many patients undergo unnecessary and costly invasive procedures. We present a 13-protein blood-based classifier that differentiates malignant and benign nodules with high confidence, thereby providing a diagnostic tool to avoid invasive biopsy on benign nodules. Using a systems biology strategy, we identified 371 protein candidates and developed a multiple reaction monitoring (MRM) assay for each. The MRM assays were applied in a three-site discovery study (n = 143) on plasma samples from patients with benign and stage IA lung cancer matched for nodule size, age, gender, and clinical site, producing a 13-protein classifier. The classifier was validated on an independent set of plasma samples (n = 104), exhibiting a negative predictive value (NPV) of 90%. Validation performance on samples from a nondiscovery clinical site showed an NPV of 94%, indicating the general effectiveness of the classifier. A pathway analysis demonstrated that the classifier proteins are likely modulated by a few transcription regulators (NF2L2, AHR, MYC, and FOS) that are associated with lung cancer, lung inflammation, and oxidative stress networks. The classifier score was independent of patient nodule size, smoking history, and age, which are risk factors used for clinical management of pulmonary nodules. Thus, this molecular test provides a potential complementary tool to help physicians in lung cancer diagnosis.
Collapse
Affiliation(s)
- Xiao-jun Li
- Integrated Diagnostics, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Sayyouh M, Vummidi DR, Kazerooni EA. Evaluation and management of pulmonary nodules: state-of-the-art and future perspectives. ACTA ACUST UNITED AC 2014; 7:629-44. [PMID: 24175679 DOI: 10.1517/17530059.2013.858117] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The imaging evaluation of pulmonary nodules, often incidentally detected on imaging examinations performed for other clinical reasons, is a frequently encountered clinical circumstance. With advances in imaging modalities, both the detection and characterization of pulmonary nodules continue to evolve and improve. AREAS COVERED This article will review the imaging modalities used to detect and diagnose benign and malignant pulmonary nodules, with a focus on computed tomography (CT), which continues to be the mainstay for evaluation. The authors discuss recent advances in the lung nodule management, and an algorithm for the management of indeterminate pulmonary nodules. EXPERT OPINION There are set of criteria that define a benign nodule, the most important of which are the lack of temporal change for 2 years or more, and certain benign imaging criteria, including specific patterns of calcification or the presence of fat. Although some indeterminate pulmonary nodules are immediately actionable, generally those approaching 1 cm or larger in diameter, at which size the diagnostic accuracy of tools such as positron emission tomography (PET)/CT, single photon emission CT (SPECT) and biopsy techniques are sufficient to warrant their use. The majority of indeterminate pulmonary nodules are under 1 cm, for which serial CT examinations through at least 2 years for solid nodules and 3 years for ground-glass nodules, are used to demonstrate either benign biologic behavior or otherwise. The management of incidental pulmonary nodules involves a multidisciplinary approach in which radiology plays a pivotal role. Newer imaging and postprocessing techniques have made this a more accurate technique eliminating ambiguity and unnecessary follow-up.
Collapse
Affiliation(s)
- Mohamed Sayyouh
- University of Michigan Health System, Division of Cardiothoracic Radiology, Department of Radiology , Ann Arbor, MI , USA
| | | | | |
Collapse
|
21
|
Hirales Casillas CE, Flores Fernández JM, Camberos EP, Herrera López EJ, Pacheco GL, Velázquez MM. Current status of circulating protein biomarkers to aid the early detection of lung cancer. Future Oncol 2014; 10:1501-13. [DOI: 10.2217/fon.14.21] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
ABSTRACT: Considerable efforts have been undertaken to produce an effective screening method to reduce lung cancer mortality. Imaging tools such as low-dose computed tomography has shown an increase in the detection of early disease and a reduction in the rate of death. This screening modality has, however, several limitations, such as overdiagnosis and a high rate of false positives. Therefore, new screening methods, such as the use of circulating protein biomarkers, have emerged as an option that could complement imaging studies. In this review, current imaging techniques applied to lung cancer screening protocols are presented, as well as up-to-date status of circulating protein biomarker panels that may improve lung cancer diagnosis. Additionally, diverse statistical and artificial intelligence tools applied to the design and optimization of these panels are discussed along with the presentation of two commercially available blood tests recently developed to help detect lung cancer early.
Collapse
Affiliation(s)
- Carlos Enrique Hirales Casillas
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco Avenida Normalistas 800, Colonia Colinas de la Normal, 44270, Guadalajara, Jalisco, México
| | - José Miguel Flores Fernández
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco Avenida Normalistas 800, Colonia Colinas de la Normal, 44270, Guadalajara, Jalisco, México
| | - Eduardo Padilla Camberos
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco Avenida Normalistas 800, Colonia Colinas de la Normal, 44270, Guadalajara, Jalisco, México
| | - Enrique J Herrera López
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco Avenida Normalistas 800, Colonia Colinas de la Normal, 44270, Guadalajara, Jalisco, México
| | - Gisela Leal Pacheco
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco Avenida Normalistas 800, Colonia Colinas de la Normal, 44270, Guadalajara, Jalisco, México
| | - Moisés Martínez Velázquez
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco Avenida Normalistas 800, Colonia Colinas de la Normal, 44270, Guadalajara, Jalisco, México
| |
Collapse
|
22
|
Kathuria H, Gesthalter Y, Spira A, Brody JS, Steiling K. Updates and controversies in the rapidly evolving field of lung cancer screening, early detection, and chemoprevention. Cancers (Basel) 2014; 6:1157-79. [PMID: 24840047 PMCID: PMC4074822 DOI: 10.3390/cancers6021157] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/25/2014] [Accepted: 05/08/2014] [Indexed: 12/21/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death in the United States. Cigarette smoking is a well-recognized risk factor for lung cancer, and a sustained elevation of lung cancer risk persists even after smoking cessation. Despite identifiable risk factors, there has been minimal improvement in mortality for patients with lung cancer primarily stemming from diagnosis at a late stage when there are few effective therapeutic options. Early detection of lung cancer and effective screening of high-risk individuals may help improve lung cancer mortality. While low dose computerized tomography (LDCT) screening of high risk smokers has been shown to reduce lung cancer mortality, the high rates of false positives and potential for over-diagnosis have raised questions on how to best implement lung cancer screening. The rapidly evolving field of lung cancer screening and early-detection biomarkers may ultimately improve the ability to diagnose lung cancer in its early stages, identify smokers at highest-risk for this disease, and target chemoprevention strategies. This review aims to provide an overview of the opportunities and challenges related to lung cancer screening, the field of biomarker development for early lung cancer detection, and the future of lung cancer chemoprevention.
Collapse
Affiliation(s)
- Hasmeena Kathuria
- The Pulmonary Center, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| | - Yaron Gesthalter
- The Pulmonary Center, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| | - Avrum Spira
- Division of Computational Biomedicine, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| | - Jerome S Brody
- The Pulmonary Center, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| | - Katrina Steiling
- Division of Computational Biomedicine, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| |
Collapse
|
23
|
Viglio S, Stolk J, Iadarola P, Giuliano S, Luisetti M, Salvini R, Fumagalli M, Bardoni A. Respiratory Proteomics Today: Are Technological Advances for the Identification of Biomarker Signatures Catching up with Their Promise? A Critical Review of the Literature in the Decade 2004-2013. Proteomes 2014; 2:18-52. [PMID: 28250368 PMCID: PMC5302730 DOI: 10.3390/proteomes2010018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/08/2014] [Accepted: 01/10/2014] [Indexed: 01/14/2023] Open
Abstract
To improve the knowledge on a variety of severe disorders, research has moved from the analysis of individual proteins to the investigation of all proteins expressed by a tissue/organism. This global proteomic approach could prove very useful: (i) for investigating the biochemical pathways involved in disease; (ii) for generating hypotheses; or (iii) as a tool for the identification of proteins differentially expressed in response to the disease state. Proteomics has not been used yet in the field of respiratory research as extensively as in other fields, only a few reproducible and clinically applicable molecular markers, which can assist in diagnosis, having been currently identified. The continuous advances in both instrumentation and methodology, which enable sensitive and quantitative proteomic analyses in much smaller amounts of biological material than before, will hopefully promote the identification of new candidate biomarkers in this area. The aim of this report is to critically review the application over the decade 2004-2013 of very sophisticated technologies to the study of respiratory disorders. The observed changes in protein expression profiles from tissues/fluids of patients affected by pulmonary disorders opens the route for the identification of novel pathological mediators of these disorders.
Collapse
Affiliation(s)
- Simona Viglio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3/B, Pavia 27100, Italy.
| | - Jan Stolk
- Department of Pulmonology, Leiden University Medical Center, Leiden 2333, The Netherlands.
| | - Paolo Iadarola
- Department of Biology and Biotechnologies, Biochemistry Unit, University of Pavia, Via Taramelli 3/B, Pavia 27100, Italy.
| | - Serena Giuliano
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3/B, Pavia 27100, Italy.
- Faculty of Science "Parc Valrose", University of Nice "Sophia Antipolis", FRE 3472 CNRS, LP2M Nice, France.
| | - Maurizio Luisetti
- Department of Molecular Medicine, Division of Pneumology, University of Pavia & IRCCS Policlinico San Matteo, Via Taramelli 5, Pavia 27100, Italy.
| | - Roberta Salvini
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3/B, Pavia 27100, Italy.
| | - Marco Fumagalli
- Department of Biology and Biotechnologies, Biochemistry Unit, University of Pavia, Via Taramelli 3/B, Pavia 27100, Italy.
| | - Anna Bardoni
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3/B, Pavia 27100, Italy.
| |
Collapse
|
24
|
Mascaux C, Peled N, Garg K, Kato Y, Wynes MW, Hirsch FR. Early detection and screening of lung cancer. Expert Rev Mol Diagn 2014; 10:799-815. [PMID: 20843203 DOI: 10.1586/erm.10.60] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Celine Mascaux
- University of Colorado Denver, Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Haick H, Broza YY, Mochalski P, Ruzsanyi V, Amann A. Assessment, origin, and implementation of breath volatile cancer markers. Chem Soc Rev 2013; 43:1423-49. [PMID: 24305596 DOI: 10.1039/c3cs60329f] [Citation(s) in RCA: 371] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A new non-invasive and potentially inexpensive frontier in the diagnosis of cancer relies on the detection of volatile organic compounds (VOCs) in exhaled breath samples. Breath can be sampled and analyzed in real-time, leading to fascinating and cost-effective clinical diagnostic procedures. Nevertheless, breath analysis is a very young field of research and faces challenges, mainly because the biochemical mechanisms behind the cancer-related VOCs are largely unknown. In this review, we present a list of 115 validated cancer-related VOCs published in the literature during the past decade, and classify them with respect to their "fat-to-blood" and "blood-to-air" partition coefficients. These partition coefficients provide an estimation of the relative concentrations of VOCs in alveolar breath, in blood and in the fat compartments of the human body. Additionally, we try to clarify controversial issues concerning possible experimental malpractice in the field, and propose ways to translate the basic science results as well as the mechanistic understanding to tools (sensors) that could serve as point-of-care diagnostics of cancer. We end this review with a conclusion and a future perspective.
Collapse
Affiliation(s)
- Hossam Haick
- The Department of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| | | | | | | | | |
Collapse
|
26
|
Alberg AJ, Brock MV, Ford JG, Samet JM, Spivack SD. Epidemiology of lung cancer: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 2013; 143:e1S-e29S. [PMID: 23649439 DOI: 10.1378/chest.12-2345] [Citation(s) in RCA: 481] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Ever since a lung cancer epidemic emerged in the mid-1900 s, the epidemiology of lung cancer has been intensively investigated to characterize its causes and patterns of occurrence. This report summarizes the key findings of this research. METHODS A detailed literature search provided the basis for a narrative review, identifying and summarizing key reports on population patterns and factors that affect lung cancer risk. RESULTS Established environmental risk factors for lung cancer include smoking cigarettes and other tobacco products and exposure to secondhand tobacco smoke, occupational lung carcinogens, radiation, and indoor and outdoor air pollution. Cigarette smoking is the predominant cause of lung cancer and the leading worldwide cause of cancer death. Smoking prevalence in developing nations has increased, starting new lung cancer epidemics in these nations. A positive family history and acquired lung disease are examples of host factors that are clinically useful risk indicators. Risk prediction models based on lung cancer risk factors have been developed, but further refinement is needed to provide clinically useful risk stratification. Promising biomarkers of lung cancer risk and early detection have been identified, but none are ready for broad clinical application. CONCLUSIONS Almost all lung cancer deaths are caused by cigarette smoking, underscoring the need for ongoing efforts at tobacco control throughout the world. Further research is needed into the reasons underlying lung cancer disparities, the causes of lung cancer in never smokers, the potential role of HIV in lung carcinogenesis, and the development of biomarkers.
Collapse
Affiliation(s)
- Anthony J Alberg
- Hollings Cancer Center and the Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC.
| | - Malcolm V Brock
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Jean G Ford
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jonathan M Samet
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Simon D Spivack
- Division of Pulmonary Medicine, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
27
|
Hensing TA, Salgia R. Molecular biomarkers for future screening of lung cancer. J Surg Oncol 2013; 108:327-33. [PMID: 23893423 DOI: 10.1002/jso.23382] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 06/28/2013] [Indexed: 12/28/2022]
Abstract
The Landmark National Lung Screening Trial established the potential for low dose CT screening (LDCT) to reduce lung cancer-specific mortality in high-risk patients as defined by smoking history and age. However, the prevalence of lung cancer in asymptomatic smokers selected based on the NLST criteria is low. Recent advances have facilitated biomarker discovery for early diagnosis of lung cancer through the analysis of surrogate tissues, including airway epithelium, sputum, exhaled breath, and blood. Although a number of candidate diagnostic biomarkers have been described, none have been validated for use in the clinical setting. The NLST ACRIN biomarker repository is a valuable resource of annotated biological specimens that were collected during the NLST trial, which has the potential to facilitate validation of candidate biomarkers for early diagnosis identified in discovery trials. It will be important to perform retrospective and prospective analysis of biomarkers to screen for lung cancer. The review below summarizes some of our understanding of biomarkers in screening.
Collapse
Affiliation(s)
- Thomas A Hensing
- NorthShore University HealthSystem, Clinical Associate Professor of Medicine, University of Chicago Pritzker, Chicago, Illinois
| | | |
Collapse
|
28
|
Pastor MD, Nogal A, Molina-Pinelo S, Carnero A, Paz-Ares L. Proteomic biomarkers in lung cancer. Clin Transl Oncol 2013; 15:671-82. [DOI: 10.1007/s12094-013-1034-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/25/2013] [Indexed: 12/12/2022]
|
29
|
Simsek C, Sonmez O, Yurdakul AS, Ozmen F, Zengin N, Keyf AI, Kubilay D, GUlbahar O, Karatayli SC, Bozdayi M, Ozturk C. Importance of Serum SELDI-TOF-MS Analysis in the Diagnosis of Early Lung Cancer. Asian Pac J Cancer Prev 2013; 14:2037-42. [DOI: 10.7314/apjcp.2013.14.3.2037] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
30
|
Indovina P, Marcelli E, Pentimalli F, Tanganelli P, Tarro G, Giordano A. Mass spectrometry-based proteomics: the road to lung cancer biomarker discovery. MASS SPECTROMETRY REVIEWS 2013; 32:129-142. [PMID: 22829143 DOI: 10.1002/mas.21355] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 04/18/2012] [Accepted: 04/18/2012] [Indexed: 06/01/2023]
Abstract
Lung cancer is the leading cause of cancer death in men and women in Western nations, and is among the deadliest cancers with a 5-year survival rate of 15%. The high mortality caused by lung cancer is attributable to a late-stage diagnosis and the lack of effective treatments. So, it is crucial to identify new biomarkers that could function not only to detect lung cancer at an early stage but also to shed light on the molecular mechanisms that underlie cancer development and serve as the basis for the development of novel therapeutic strategies. Considering that DNA-based biomarkers for lung cancer showed inadequate sensitivity, specificity, and reproducibility, proteomics could represent a better tool for the identification of useful biomarkers and therapeutic targets for this cancer type. Among the proteomics technologies, the most powerful tool is mass spectrometry. In this review, we describe studies that use mass spectrometry-based proteomics technologies to analyze tumor proteins and peptides, which might represent new diagnostic, prognostic, and predictive markers for lung cancer. We focus in particular on those findings that hold promise to impact significantly on the clinical management of this disease.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Biomarkers/blood
- Biomarkers/metabolism
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/chemistry
- Biomarkers, Tumor/metabolism
- Chromatography, High Pressure Liquid
- Glycosylation/drug effects
- Humans
- Lung Neoplasms/blood
- Lung Neoplasms/diagnosis
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Pleural Effusion, Malignant/blood
- Pleural Effusion, Malignant/drug therapy
- Pleural Effusion, Malignant/metabolism
- Prognosis
- Protein Processing, Post-Translational/drug effects
- Proteomics/methods
- Saliva/chemistry
- Saliva/drug effects
- Spectrometry, Mass, Electrospray Ionization
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Tandem Mass Spectrometry
Collapse
Affiliation(s)
- Paola Indovina
- Department of Human Pathology and Oncology, University of Siena, Siena, Italy
| | | | | | | | | | | |
Collapse
|
31
|
Pass HI, Beer DG, Joseph S, Massion P. Biomarkers and molecular testing for early detection, diagnosis, and therapeutic prediction of lung cancer. Thorac Surg Clin 2013; 23:211-24. [PMID: 23566973 DOI: 10.1016/j.thorsurg.2013.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The search for biomarkers in the management of lung cancer involves the use of multiple platforms to examine changes in gene, protein, and microRNA expression. Multiple studies have been published in an attempt to describe early detection, diagnostic, prognostic, and predictive biomarkers using chiefly tissues and blood elements. Studies are characterized by a lack of commonality of specific biomarkers, and a lack of validated, clinically useful markers. The future of biomarker discovery as a means of tailoring therapy for patients with lung cancer will involve next-generation sequencing along with collaborative efforts to integrate and validate candidate markers.
Collapse
Affiliation(s)
- Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Langone Medical Center, 530 First Avenue, 9V, New York, NY 10016, USA.
| | | | | | | |
Collapse
|
32
|
Serum biomarkers identification by mass spectrometry in high-mortality tumors. INTERNATIONAL JOURNAL OF PROTEOMICS 2013; 2013:125858. [PMID: 23401773 PMCID: PMC3562576 DOI: 10.1155/2013/125858] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 11/16/2012] [Accepted: 12/11/2012] [Indexed: 02/08/2023]
Abstract
Cancer affects millions of people worldwide. Tumor mortality is substantially due to diagnosis at stages that are too late for therapies to be effective. Advances in screening methods have improved the early diagnosis, prognosis, and survival for some cancers. Several validated biomarkers are currently used to diagnose and monitor the progression of cancer, but none of them shows adequate specificity, sensitivity, and predictive value for population screening. So, there is an urgent need to isolate novel sensitive, specific biomarkers to detect the disease early and improve prognosis, especially in high-mortality tumors. Proteomic techniques are powerful tools to help in diagnosis and monitoring of treatment and progression of the disease. During the last decade, mass spectrometry has assumed a key role in most of the proteomic analyses that are focused on identifying cancer biomarkers in human serum, making it possible to identify and characterize at the molecular level many proteins or peptides differentially expressed. In this paper we summarize the results of mass spectrometry serum profiling and biomarker identification in high mortality tumors, such as ovarian, liver, lung, and pancreatic cancer.
Collapse
|
33
|
Tabb DL. Quality assessment for clinical proteomics. Clin Biochem 2012; 46:411-20. [PMID: 23246537 DOI: 10.1016/j.clinbiochem.2012.12.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 12/01/2012] [Accepted: 12/03/2012] [Indexed: 12/21/2022]
Abstract
Proteomics has emerged from the labs of technologists to enter widespread application in clinical contexts. This transition, however, has been hindered by overstated early claims of accuracy, concerns about reproducibility, and the challenges of handling batch effects properly. New efforts have produced sets of performance metrics and measurements of variability that establish sound expectations for experiments in clinical proteomics. As researchers begin incorporating these metrics in a quality by design paradigm, the variability of individual steps in experimental pipelines will be reduced, regularizing overall outcomes. This review discusses the evolution of quality assessment in 2D gel electrophoresis, mass spectrometry-based proteomic profiling, tandem mass spectrometry-based protein inventories, and proteomic quantitation. Taken together, the advances in each of these technologies are establishing databases that will be increasingly useful for decision-making in clinical experimentation.
Collapse
Affiliation(s)
- David L Tabb
- Department of Biomedical Informatics, Vanderbilt University, USA.
| |
Collapse
|
34
|
Abstract
BACKGROUND High-throughput laboratory technologies coupled with sophisticated bioinformatics algorithms have tremendous potential for discovering novel biomarkers, or profiles of biomarkers, that could serve as predictors of disease risk, response to treatment or prognosis. We discuss methodological issues in wedding high-throughput approaches for biomarker discovery with the case-control study designs typically used in biomarker discovery studies, especially focusing on nested case-control designs. METHODS We review principles for nested case-control study design in relation to biomarker discovery studies and describe how the efficiency of biomarker discovery can be effected by study design choices. We develop a simulated prostate cancer cohort data set and a series of biomarker discovery case-control studies nested within the cohort to illustrate how study design choices can influence biomarker discovery process. RESULT Common elements of nested case-control design, incidence density sampling and matching of controls to cases are not typically factored correctly into biomarker discovery analyses, inducing bias in the discovery process. We illustrate how incidence density sampling and matching of controls to cases reduce the apparent specificity of truly valid biomarkers 'discovered' in a nested case-control study. We also propose and demonstrate a new case-control matching protocol, we call 'antimatching', that improves the efficiency of biomarker discovery studies. CONCLUSIONS For a valid, but as yet undiscovered, biomarker(s) disjunctions between correctly designed epidemiologic studies and the practice of biomarker discovery reduce the likelihood that true biomarker(s) will be discovered and increases the false-positive discovery rate.
Collapse
Affiliation(s)
- Andrew Rundle
- Department of Epidemiology, Mailman School of Public Health, and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA.
| | | | | |
Collapse
|
35
|
Hakim M, Broza YY, Barash O, Peled N, Phillips M, Amann A, Haick H. Volatile organic compounds of lung cancer and possible biochemical pathways. Chem Rev 2012; 112:5949-66. [PMID: 22991938 DOI: 10.1021/cr300174a] [Citation(s) in RCA: 523] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Meggie Hakim
- The Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
36
|
Luque de Castro M, Fernández-Peralbo M. Analytical methods based on exhaled breath for early detection of lung cancer. Trends Analyt Chem 2012. [DOI: 10.1016/j.trac.2012.03.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
37
|
MALDI-MS-Based Profiling of Serum Proteome: Detection of Changes Related to Progression of Cancer and Response to Anticancer Treatment. INTERNATIONAL JOURNAL OF PROTEOMICS 2012; 2012:926427. [PMID: 22900176 PMCID: PMC3413974 DOI: 10.1155/2012/926427] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/12/2012] [Accepted: 06/12/2012] [Indexed: 01/25/2023]
Abstract
Mass spectrometry-based analyses of the low-molecular-weight fraction of serum proteome allow identifying proteome profiles (signatures) that are potentially useful in detection and classification of cancer. Several published studies have shown that multipeptide signatures selected in numerical tests have potential values for diagnostics of different types of cancer. However due to apparent problems with standardization of methodological details, both experimental and computational, none of the proposed peptide signatures analyzed directly by MALDI/SELDI-ToF spectrometry has been approved for routine diagnostics. Noteworthy, several components of proposed cancer signatures, especially those characteristic for advanced cancer, were identified as fragments of blood proteins involved in the acute phase and inflammatory response. This indicated that among cancer biomarker candidates to be possibly identified by serum proteome profiling were rather those reflecting overall influence of a disease (and the therapy) upon the human organism, than products of cancer-specific genes. Current paper focuses on changes in serum proteome that are related to response of patient's organism to progressing malignancy and toxicity of anticancer treatment. In addition, several methodological issues that affect robustness and interlaboratory reproducibility of MS-based serum proteome profiling are discussed.
Collapse
|
38
|
Hassanein M, Callison JC, Callaway-Lane C, Aldrich MC, Grogan EL, Massion PP. The state of molecular biomarkers for the early detection of lung cancer. Cancer Prev Res (Phila) 2012; 5:992-1006. [PMID: 22689914 DOI: 10.1158/1940-6207.capr-11-0441] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Using biomarkers to select the most at-risk population, to detect the disease while measurable and yet not clinically apparent has been the goal of many investigations. Recent advances in molecular strategies and analytic platforms, including genomics, epigenomics, proteomics, and metabolomics, have identified increasing numbers of potential biomarkers in the blood, urine, exhaled breath condensate, bronchial specimens, saliva, and sputum, but none have yet moved to the clinical setting. Therefore, there is a recognized gap between the promise and the product delivery in the cancer biomarker field. In this review, we define clinical contexts where risk and diagnostic biomarkers may have use in the management of lung cancer, identify the most relevant candidate biomarkers of early detection, provide their state of development, and finally discuss critical aspects of study design in molecular biomarkers for early detection of lung cancer.
Collapse
Affiliation(s)
- Mohamed Hassanein
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt-Ingram Cancer Center, Nashville TN 37232, USA
| | | | | | | | | | | |
Collapse
|
39
|
Seven-signal proteomic signature for detection of operable pancreatic ductal adenocarcinoma and their discrimination from autoimmune pancreatitis. INTERNATIONAL JOURNAL OF PROTEOMICS 2012; 2012:510397. [PMID: 22675630 PMCID: PMC3361197 DOI: 10.1155/2012/510397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 03/09/2012] [Indexed: 12/24/2022]
Abstract
There is urgent need for biomarkers that provide early detection of pancreatic ductal adenocarcinoma (PDAC) as well as discrimination of autoimmune pancreatitis, as current clinical approaches are not suitably accurate for precise diagnosis. We used mass spectrometry to analyze protein profiles of more than 300 plasma specimens obtained from PDAC, noncancerous pancreatic diseases including autoimmune pancreatitis patients and healthy subjects. We obtained 1063 proteomic signals from 160 plasma samples in the training cohort. A proteomic signature consisting of 7 mass spectrometry signals was used for construction of a proteomic model for detection of PDAC patients. Using the test cohort, we confirmed that this proteomic model had discrimination power equal to that observed with the training cohort. The overall sensitivity and specificity for detection of cancer patients were 82.6% and 90.9%, respectively. Notably, 62.5% of the stage I and II cases were detected by our proteomic model. We also found that 100% of autoimmune pancreatitis patients were correctly assigned as noncancerous individuals. In the present paper, we developed a proteomic model that was shown able to detect early-stage PDAC patients. In addition, our model appeared capable of discriminating patients with autoimmune pancreatitis from those with PDAC.
Collapse
|
40
|
Bigbee WL, Gopalakrishnan V, Weissfeld JL, Wilson DO, Dacic S, Lokshin AE, Siegfried JM. A multiplexed serum biomarker immunoassay panel discriminates clinical lung cancer patients from high-risk individuals found to be cancer-free by CT screening. J Thorac Oncol 2012; 7:698-708. [PMID: 22425918 PMCID: PMC3308353 DOI: 10.1097/jto.0b013e31824ab6b0] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Clinical decision making in the setting of computed tomography (CT) screening could benefit from accessible biomarkers that help predict the level of lung cancer risk in high-risk individuals with indeterminate pulmonary nodules. METHODS To identify candidate serum biomarkers, we measured 70 cancer-related proteins by Luminex xMAP (Luminex Corporation) multiplexed immunoassays in a training set of sera from 56 patients with biopsy-proven primary non-small-cell lung cancer and 56 age-, sex-, and smoking-matched CT-screened controls. RESULTS We identified a panel of 10 serum biomarkers-prolactin, transthyretin, thrombospondin-1, E-selectin, C-C motif chemokine 5, macrophage migration inhibitory factor, plasminogen activator inhibitor, receptor tyrosine-protein kinase, erbb-2, cytokeratin fragment 21.1, and serum amyloid A-that distinguished lung cancer patients from controls with an estimated balanced accuracy (average of sensitivity and specificity) of 76.0 ± 3.8% from 20-fold internal cross-validation. We then iteratively evaluated this model in an independent test and verification case/control studies confirming the initial classification performance of the panel. The classification performance of the 10-biomarker panel was also analytically validated using enzyme-linked immunosorbent assays in a second independent case/control population, further validating the robustness of the panel. CONCLUSIONS The performance of this 10-biomarker panel-based model was 77.1% sensitivity/76.2% specificity in cross-validation in the expanded training set, 73.3% sensitivity/93.3% specificity (balanced accuracy 83.3%) in the blinded verification set with the best discriminative performance in stage I/II cases: 85% sensitivity (balanced accuracy 89.2%). Importantly, the rate of misclassification of CT-screened controls was not different in most control subgroups with or without airflow obstruction or emphysema or pulmonary nodules. These biomarkers have potential to aid in the early detection of lung cancer and more accurate interpretation of indeterminate pulmonary nodules detected by CT screening.
Collapse
Affiliation(s)
- William L Bigbee
- Mass Spectrometry Platform, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Pecot CV, Li M, Zhang XJ, Rajanbabu R, Calitri C, Bungum A, Jett JR, Putnam JB, Callaway-Lane C, Deppen S, Grogan EL, Carbone DP, Worrell JA, Moons KGM, Shyr Y, Massion PP. Added value of a serum proteomic signature in the diagnostic evaluation of lung nodules. Cancer Epidemiol Biomarkers Prev 2012; 21:786-92. [PMID: 22374995 DOI: 10.1158/1055-9965.epi-11-0932] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Current management of lung nodules is complicated by nontherapeutic resections and missed chances for cure. We hypothesized that a serum proteomic signature may add diagnostic information beyond that provided by combined clinical and radiographic data. METHODS Cohort A included 265 and cohort B 114 patients. Using multivariable logistic regression analysis we calculated the area under the receiver operating characteristic curve (AUC) and quantified the added value of a previously described serum proteomic signature beyond clinical and radiographic risk factors for predicting lung cancer using the integration discrimination improvement (IDI) index. RESULTS The average computed tomography (CT) measured nodule size in cohorts A and B was 37.83 versus 23.15 mm among patients with lung cancer and 15.82 versus 17.18 mm among those without, respectively. In cohort A, the AUC increased from 0.68 to 0.86 after adding chest CT imaging variables to the clinical results, but the proteomic signature did not provide meaningful added value. In contrast, in cohort B, the AUC improved from 0.46 with clinical data alone to 0.61 when combined with chest CT imaging data and to 0.69 after adding the proteomic signature (IDI of 20% P = 0.0003). In addition, in a subgroup of 100 nodules between 5 and 20 mm in diameter, the proteomic signature added value with an IDI of 15% (P ≤ 0.0001). CONCLUSIONS The results show that this serum proteomic biomarker signature may add value to the clinical and chest CT evaluation of indeterminate lung nodules. IMPACT This study suggests a possible role of a blood biomarker in the evaluation of indeterminate lung nodules.
Collapse
Affiliation(s)
- Chad V Pecot
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Musharraf SG, Hashmi N, Choudhary MI, Rizvi N, Usman A, Atta-ur-Rahman. Comparison of plasma from healthy nonsmokers, smokers, and lung cancer patients: pattern-based differentiation profiling of low molecular weight proteins and peptides by magnetic bead technology with MALDI-TOF MS. Biomarkers 2012; 17:223-30. [PMID: 22356277 DOI: 10.3109/1354750x.2012.657245] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Smoking is the major contributor of lung cancer (LC), which accounts for millions of death. OBJECTIVE This study focused on the correlation between the proteomic profiling of LC patients, and healthy nonsmokers and smokers. METHOD Pattern-based peptide profiling of 186 plasma samples was performed through reversed-phase chromatography-18 magnetic bead fractionation coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis and resulted data were evaluated statistically by ClinProTool. RESULTS Marker peaks at m/z 1760, 5773, 5851, 2940, and 7172 were found with an excellent statistical figure. CONCLUSION Selected marker peaks can be served as a differentiated tool of LC patients with high sensitivity and specificity.
Collapse
Affiliation(s)
- Syed G Musharraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, University of Karachi, Karachi, Pakistan.
| | | | | | | | | | | |
Collapse
|
43
|
Ganchev P, Malehorn D, Bigbee WL, Gopalakrishnan V. Transfer learning of classification rules for biomarker discovery and verification from molecular profiling studies. J Biomed Inform 2011; 44 Suppl 1:S17-S23. [PMID: 21571094 PMCID: PMC3706089 DOI: 10.1016/j.jbi.2011.04.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 04/25/2011] [Accepted: 04/26/2011] [Indexed: 12/14/2022]
Abstract
We present a novel framework for integrative biomarker discovery from related but separate data sets created in biomarker profiling studies. The framework takes prior knowledge in the form of interpretable, modular rules, and uses them during the learning of rules on a new data set. The framework consists of two methods of transfer of knowledge from source to target data: transfer of whole rules and transfer of rule structures. We evaluated the methods on three pairs of data sets: one genomic and two proteomic. We used standard measures of classification performance and three novel measures of amount of transfer. Preliminary evaluation shows that whole-rule transfer improves classification performance over using the target data alone, especially when there is more source data than target data. It also improves performance over using the union of the data sets.
Collapse
Affiliation(s)
- Philip Ganchev
- Intelligent Systems Program, University of Pittsburgh, Pittsburgh, PA, United States.
| | - David Malehorn
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - William L Bigbee
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Vanathi Gopalakrishnan
- Intelligent Systems Program, University of Pittsburgh, Pittsburgh, PA, United States; Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
44
|
Bateson H, Saleem S, Loadman PM, Sutton CW. Use of matrix-assisted laser desorption/ionisation mass spectrometry in cancer research. J Pharmacol Toxicol Methods 2011; 64:197-206. [DOI: 10.1016/j.vascn.2011.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/18/2011] [Accepted: 04/08/2011] [Indexed: 02/04/2023]
|
45
|
Bateson H, Saleem S, Loadman PM, Sutton CW. Use of matrix-assisted laser desorption/ionisation mass spectrometry in cancer research. J Pharmacol Toxicol Methods 2011; 64:197-206. [DOI: https:/doi.org/10.1016/j.vascn.2011.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
46
|
Abstract
Since the advent of the new proteomics era more than a decade ago, large-scale studies of protein profiling have been exploited to identify the distinctive molecular signatures in a wide array of biological systems spanning areas of basic biological research, various disease states, and biomarker discovery directed toward therapeutic applications. Recent advances in protein separation and identification techniques have significantly improved proteomics approaches, leading to enhancement of the depth and breadth of proteome coverage. Proteomic signatures specific for invasive lung cancer and preinvasive lesions have begun to emerge. In this review we provide a critical assessment of the state of recent advances in proteomic approaches and the biological lessons they have yielded, with specific emphasis on the discovery of biomarker signatures for the early detection of lung cancer.
Collapse
|
47
|
Zhu P, Bowden P, Zhang D, Marshall JG. Mass spectrometry of peptides and proteins from human blood. MASS SPECTROMETRY REVIEWS 2011; 30:685-732. [PMID: 24737629 DOI: 10.1002/mas.20291] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 12/09/2009] [Accepted: 01/19/2010] [Indexed: 06/03/2023]
Abstract
It is difficult to convey the accelerating rate and growing importance of mass spectrometry applications to human blood proteins and peptides. Mass spectrometry can rapidly detect and identify the ionizable peptides from the proteins in a simple mixture and reveal many of their post-translational modifications. However, blood is a complex mixture that may contain many proteins first expressed in cells and tissues. The complete analysis of blood proteins is a daunting task that will rely on a wide range of disciplines from physics, chemistry, biochemistry, genetics, electromagnetic instrumentation, mathematics and computation. Therefore the comprehensive discovery and analysis of blood proteins will rank among the great technical challenges and require the cumulative sum of many of mankind's scientific achievements together. A variety of methods have been used to fractionate, analyze and identify proteins from blood, each yielding a small piece of the whole and throwing the great size of the task into sharp relief. The approaches attempted to date clearly indicate that enumerating the proteins and peptides of blood can be accomplished. There is no doubt that the mass spectrometry of blood will be crucial to the discovery and analysis of proteins, enzyme activities, and post-translational processes that underlay the mechanisms of disease. At present both discovery and quantification of proteins from blood are commonly reaching sensitivities of ∼1 ng/mL.
Collapse
Affiliation(s)
- Peihong Zhu
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, Canada M5B 2K3
| | | | | | | |
Collapse
|
48
|
Ostroff RM, Bigbee WL, Franklin W, Gold L, Mehan M, Miller YE, Pass HI, Rom WN, Siegfried JM, Stewart A, Walker JJ, Weissfeld JL, Williams S, Zichi D, Brody EN. Unlocking biomarker discovery: large scale application of aptamer proteomic technology for early detection of lung cancer. PLoS One 2010; 5:e15003. [PMID: 21170350 PMCID: PMC2999620 DOI: 10.1371/journal.pone.0015003] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Accepted: 10/07/2010] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer deaths worldwide. New diagnostics are needed to detect early stage lung cancer because it may be cured with surgery. However, most cases are diagnosed too late for curative surgery. Here we present a comprehensive clinical biomarker study of lung cancer and the first large-scale clinical application of a new aptamer-based proteomic technology to discover blood protein biomarkers in disease. METHODOLOGY/PRINCIPAL FINDINGS We conducted a multi-center case-control study in archived serum samples from 1,326 subjects from four independent studies of non-small cell lung cancer (NSCLC) in long-term tobacco-exposed populations. Sera were collected and processed under uniform protocols. Case sera were collected from 291 patients within 8 weeks of the first biopsy-proven lung cancer and prior to tumor removal by surgery. Control sera were collected from 1,035 asymptomatic study participants with ≥ 10 pack-years of cigarette smoking. We measured 813 proteins in each sample with a new aptamer-based proteomic technology, identified 44 candidate biomarkers, and developed a 12-protein panel (cadherin-1, CD30 ligand, endostatin, HSP90α, LRIG3, MIP-4, pleiotrophin, PRKCI, RGM-C, SCF-sR, sL-selectin, and YES) that discriminates NSCLC from controls with 91% sensitivity and 84% specificity in cross-validated training and 89% sensitivity and 83% specificity in a separate verification set, with similar performance for early and late stage NSCLC. CONCLUSIONS/SIGNIFICANCE This study is a significant advance in clinical proteomics in an area of high unmet clinical need. Our analysis exceeds the breadth and dynamic range of proteome interrogated of previously published clinical studies of broad serum proteome profiling platforms including mass spectrometry, antibody arrays, and autoantibody arrays. The sensitivity and specificity of our 12-biomarker panel improves upon published protein and gene expression panels. Separate verification of classifier performance provides evidence against over-fitting and is encouraging for the next development phase, independent validation. This careful study provides a solid foundation to develop tests sorely needed to identify early stage lung cancer.
Collapse
Affiliation(s)
| | - William L. Bigbee
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Wilbur Franklin
- University of Colorado Cancer Center, University of Colorado at Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Larry Gold
- SomaLogic, Boulder, Colorado, United States of America
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Mike Mehan
- SomaLogic, Boulder, Colorado, United States of America
| | - York E. Miller
- University of Colorado Cancer Center, University of Colorado at Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Denver Veterans Affairs Medical Center, Denver, Colorado, United States of America
| | - Harvey I. Pass
- Langone Medical Center and Cancer Center, New York University School of Medicine, New York, New York, United States of America
| | - William N. Rom
- Division of Pulmonary, and Critical Care, and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Jill M. Siegfried
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Alex Stewart
- SomaLogic, Boulder, Colorado, United States of America
| | | | - Joel L. Weissfeld
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | | | - Dom Zichi
- SomaLogic, Boulder, Colorado, United States of America
| | | |
Collapse
|
49
|
de Costa D, Broodman I, Vanduijn MM, Stingl C, Dekker LJM, Burgers PC, Hoogsteden HC, Sillevis Smitt PAE, van Klaveren RJ, Luider TM. Sequencing and quantifying IgG fragments and antigen-binding regions by mass spectrometry. J Proteome Res 2010; 9:2937-45. [PMID: 20387908 DOI: 10.1021/pr901114w] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In cancer and autoimmune diseases, immunoglobulins with a specific molecular signature that could potentially be used as diagnostic or prognostic markers are released into body fluids. An immunomics approach based on this phenomenon relies on the ability to identify the specific amino acid sequences of the complementarity-determining regions (CDR) of these immunoglobulins, which in turn depends on the level of accuracy, resolution, and sensitivity that can be achieved by advanced mass spectrometry. Reproducible isolation and sequencing of antibody fragments (e.g., Fab) by high-resolution mass spectrometry (MS) from seven healthy donors revealed 43 217 MS signals: 225 could be associated with CDR1 peptides, 513 with CDR2 peptides, and 19 with CDR3 peptides. Seventeen percent of the 43 217 MS signals did not overlap between the seven donors. The Fab isolation method used is reproducible and fast, with a high yield. It provides only one Fab sample fraction for subsequent characterization by high-resolution MS. In 17% and 4% of these seven healthy donors, qualitative (presence/absence) and quantitative (intensity) differences in Fab fragments could be demonstrated, respectively. From these results, we conclude that the identification of a CDR signature as biomarker for autoimmune diseases and cancer without prior knowledge of the antigen is feasible.
Collapse
Affiliation(s)
- Dominique de Costa
- Department of Pulmonology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Friedman DB. An Introduction to Proteomics Technologies for the Genomics Scientist. Genomics 2010. [DOI: 10.1002/9780470711675.ch13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|