Preclinical Biomarkers for the Early Detection of Lung Cancer





Summary of Key Points





  • This chapter provides a review of some of the most promising recent studies of diagnostic biomarkers in lung cancer.



  • We discuss the challenges and the importance of biomarker validation. Current guidelines recommend a study design to include prospective collections of specimens and retrospective blinded evaluation.



  • A novel multiomics approach to biomarker discovery has greatly advanced the field of early lung cancer detection.



  • Noninvasive biomarkers in the blood, sputum, airway epithelium, or exhaled breath can be combined with imaging to detect early stage lung cancer and improve mortality.





Acknowledgment


This work was supported in part by NCI EDRN-sponsored Clinical Validation Center CA086137 (W.N.R.) and by NCI EDRN-sponsored Clinical Validation Center CA152662 (P.P.M.).


Lung cancer is the leading cause of cancer deaths in the United States and worldwide. This statistic is largely due to the persistent poor survival of patients diagnosed with lung cancer. In the United States as of 2009, the overall 5-year survival for nonsmall cell lung cancer (NSCLC) remained at only 16.6%. However, if the cancer is detected at an early stage, the 5-year survival exceeds 50%. For this reason, in the last decade, the quest for an effective means of early diagnosis has intensified. In 2011, the results of the randomized multicenter National Lung Screening Trial (NLST) were published, confirming that early diagnosis of lung cancer can improve survival. Screening for lung cancer in the high-risk group studied in the NLST now has the support of the US Preventive Services Task Force (grade B recommendation). However, low-dose computed tomography (CT) of the chest for lung cancer screening has significant drawbacks, including cost, radiation exposure, high false-positive rates, and a risk of overdiagnosis of indolent cancers. Thus the results of NLST have sparked even greater interest in developing more practical and more specific means of early detection of lung cancer, using noninvasive biomarkers of early disease.


Biomarkers for lung cancer have several potential clinical uses in addition to early detection ( Fig. 8.1 ). They may be used for risk stratification, optimal treatment selection, prognostication, and monitoring for recurrence. Markers of risk can help identify a population to be screened. At this preclinical stage, the marker identifies individuals without disease but with factors that may predispose them to lung cancer. Given the high false-positive rate with CT screening, a marker that could more clearly define the at-risk population could decrease the number of screening CT scans conducted and also improve the specificity of CT screening, thus decreasing patient anxiety and the need for repeated CT and invasive procedures induced by false-positive nodules.




Fig. 8.1


Lung cancer biomarkers have many potential clinical uses, depending on the marker and the clinical stage. Four clinical contexts for biomarker use include the following: (1) During the period before lung cancer is detectable, markers may be used for risk assessment and to identify populations that may benefit from lung cancer screening or chemopreventive measures. (2) When lung cancer is in the preneoplastic stage, it is generally not clinically detectable. Biomarkers that identify preneoplasia would lead clinicians to recommend close monitoring and chemoprevention if available. (3) Early stage disease can be detected by thoracic imaging, but this technique is nonspecific, and indeterminate nodules are frequent. Lung cancer biomarkers may be used either to identify individuals who should undergo computed tomography (CT) screening or to differentiate benign from malignant nodules. At this stage, biomarkers may also be used for prognostication and treatment selection if they can distinguish indolent from aggressive disease. (4) After lung cancer has been treated, biomarkers may be useful for monitoring for recurrence or to determine prognosis and select patients for adjuvant chemotherapy or tertiary chemoprevention.


Markers are currently used for treatment selection, prognostication, and monitoring for recurrence in patients with known disease. A variety of markers, reflecting the biology of lung cancer progression from premalignant lesions to invasive lung cancer, may prove to be more useful for each of these roles. In this chapter, we focus on current and potential biomarkers for the early detection of lung cancer. Markers of risk and prognosis are not reviewed.




Early Detection


For the foreseeable future, CT will undoubtedly remain an important part of any program for the early detection of lung cancer. CT can detect the small noncalcified nodules that may represent early lung cancers. However, as a stand-alone screening tool, this technique is problematic. First, it has poor specificity because of the high prevalence of nonspecific benign pulmonary nodules. Second, CT is costly, and the necessity for repeated CT to determine growth rates over time can expose patients to potentially harmful radiation. Lastly, we cannot predict which early lung cancers will progress and which will remain indolent for prolonged periods.


The ultimate goal of lung cancer early detection biomarker research is to develop a marker that identifies early stage lung cancer (or even preneoplasia) and prompts a change in clinical practice that saves lives. A more obtainable target may be a marker that can be used in conjunction with chest CT to help distinguish malignant from benign nodules found on CT images or identify aggressive or indolent phenotypes of early lung cancers found by imaging. Depending on the selected size cutoff, 15% to more than 50% of individuals in CT screening programs have nodules. NLST demonstrated that more than 96% of the nodules identified were thought to be benign based on stability on follow-up CT. Of nodules that are ultimately surgically resected, up to 30% are found to have benign pathology. In the NLST, 24% of patients who underwent an invasive diagnostic procedure were found to have nodules of benign etiology. To address the issue of large numbers of false-positive findings on CT, experts have suggested using a larger nodule size cutoff of 7 mm or 8 mm, which would decrease the number of positive CT results to 5% to 7%, or narrowing the definition of high-risk individuals who would be eligible for screening. An effective biomarker would also be an invaluable aid in the management of these indeterminate pulmonary nodules. Depending on their assay performance characteristics, biomarkers could guide the clinician toward reassurance, watchful waiting, or immediate biopsy or resection, and thus decrease the anxiety, cost, and uncertainty of lung cancer screening.


Lung cancer biomarkers may also reduce the problem of overdiagnosis in lung cancer screening. Although the NLST demonstrated that screening can decrease lung cancer mortality, a percentage of cancers diagnosed are likely indolent malignancies that may not progress if disregarded. At the New York University screening program, one-third of the cancers diagnosed were indolent adenocarcinomas, which were followed for a prolonged period before resection and were still stage I at the time of surgery. A biomarker that could a priori identify these indolent cancers may spare older patients or patients with other medical problems unnecessary surgeries.


The Biology of Lung Carcinogenesis


Continued progress in understanding the sequence of molecular changes underlying the progression from preneoplasia to invasive lung cancer has galvanized research into discovery and validation of lung cancer biomarkers for early detection. It has also raised the possibility of personalizing lung cancer treatment using biomarker profiles. The World Health Organization defines the various preneoplastic lesions of the bronchial epithelium as squamous dysplasia and carcinoma in situ, which progresses to squamous cell carcinoma; atypical adenomatous hyperplasia, which may precede adenocarcinoma; and diffuse idiopathic pulmonary neuroendocrine cell hyperplasia, which may progress to carcinoid. Small cell lung cancer (SCLC) is believed to arise from extensively molecularly damaged epithelium without going through recognizable preneoplastic stages.


Alterations in gene expression and chromosome structure known to be associated with malignant transformation have been demonstrated in these preneoplastic lesions, and the changes appear to be sequential; in particular, their frequency and number increase with increasing atypia. Some of the alterations found in preneoplastic lesions include hyperproliferation and loss of cell cycle control; abnormalities in the p53 pathway, the RAS genes, and genes in the genomic region of 3p14.2 and 3q26-29; aberrant gene promoter methylation; increased vascular growth; altered extracellular matrix; decreased retinoic acid and retinoid X receptor expression; and many other genetic and epigenetic changes.


Biomarker Validation


The validation of a biomarker for clinical use is challenging. Any biomarker considered for use in a clinical setting must satisfy a host of criteria related to ease of use and performance. The biomarker must be relatively noninvasive, require only small amounts of material needing a minimum of preparation, be quantifiable and reproducible in multiple populations and laboratories, have a proven clinical use with acceptable sensitivity and specificity for this use, be acceptable to the target population, and be cost-effective and reimbursed by health insurers. No markers have yet made it through these rigorous requirements, although many are in the pipeline. Appropriate study design will be crucial to bringing any of these markers to clinical use.


Guidelines for biomarker study design and statistical evaluation suggest that validation should be conducted using a prospective specimen collection retrospective blinded evaluation design. In this approach, specimens are collected prospectively from a longitudinal cohort that represents the target population. After the outcome status is determined, a nested case–control study can be designed. Cases and controls are selected randomly for biomarker studies, with the investigators blinded to the case–control status. Random sampling of cases and controls from within a well-defined cohort provides validity to the case–control design. An important element of this study design is that the validation population must be representative of the population in which the biomarker will be used, to minimize false positives. In the case of lung cancer, this means that individuals with a history of tobacco use and its related morbidities, including chronic obstructive pulmonary disease, cardiovascular disease, and other malignancies, must be included in the validation cohort. Ideally, the biomarker can be tested in longitudinal samples to ensure its accuracy in detecting early, preclinical disease. Measures of validity include sensitivity, specificity, negative predictive value, and positive predictive value (which can be summarized with a receiver operating characteristic [ROC] curve). The prevalence of the disease influences these measures, thus it is important that the biomarker validation process be applied to all possible populations in which the marker would be used. Lastly, when a potential marker has been validated as effective for early diagnosis, it should be evaluated in a screening trial with lung cancer mortality as the end point to prove that use of the biomarker decreases mortality and the validation studies were not hampered by problems of overdiagnosis, lead-time bias, or length bias. The Early Detection Research Network of the US National Cancer Institute has established guidelines for cancer biomarker development and validation.


Advances in Techniques for Biomarker Discovery


Currently, we see a profusion of potential biomarkers for lung cancer. Different histologic types, different stages of disease, and a variety of molecular pathways to transformation contribute to making the process of biomarker discovery for lung cancer complex. New high-throughput technologies allow researchers to look for and validate multiple biomarkers simultaneously. Microarrays are used to evaluate thousands of potential markers concurrently.


For example, circulating DNA (cDNA) microarrays identify thousands of genes that are differentially expressed in lung cancers, preneoplasias, and normal lung; antibody arrays evaluate multiple antigens or antibodies at once; and methylation arrays identify methylation of many different gene promoters simultaneously. Proteomics is the study of protein profiles in tissues and body fluids. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) and surface-enhanced laser desorption/ionization have been used to describe protein profiles and to identify individual protein markers in lung cancer. The ability to accurately measure quantitative transcriptome in individual cells with relatively small number of sequencing read makes single-cell RNA sequencing a popular technology for biomarker discovery. In recent years, important advances in the development and validation of these and other high-throughput technologies have raised the potential for great strides in biomarker discovery.


Specimen Types


One of the most important criteria for a successful biomarker is that the testing material be easily accessible. Current markers use multiple biologic sources. Tissue-based assays are generally the most invasive, but may be acceptable in some circumstances. The concept of field cancerization supports the theory that surrogate tissues—such as bronchial, buccal, and nasal brushings; endobronchial biopsy specimens; or even exhaled breath—may be used as markers of increased risk for lung cancer. Genetic and epigenetic changes in the bronchial epithelium or perhaps the nasal or buccal epithelium may mirror changes in the lower respiratory tract and suggest that a lesion seen on CT images represents malignancy. Although obtaining the tissue may require bronchoscopy, pairing molecular markers obtained from the airways with a high-risk profile and a lesion on CT images may increase the specificity of lung cancer screening. The potential use of tissue-based biomarkers is highly dependent on the accessibility of the specimens and the robustness of the assay offered. It may take additional time to refine airway epithelium-based biomarkers because banked samples are not as readily available as they are for tumor tissues or blood. Blood-based assays are attractive due to the ease of acquisition. This simplicity aids in the discovery process, the validation process, and the acceptance into clinical practice. Altered or methylated DNA, overexpressed messenger RNA (mRNA), microRNA (miRNA), proteins, peptides, metabolites, and even circulating tumor cells (CTCs) can all be detected in the circulating blood; however, there are significant challenges as well. Blood is a dynamic medium, which reflects various physiologic and pathologic states that can overwhelm the detection of an early stage, preclinical cancer.


Other biofluids—exhaled breath condensate, sputum, and urine—are also easily accessible samples for biomarker analysis. Each type of sample has its own appeal and its challenges. Exhaled breath is easily and painlessly obtained, and large volumes can be collected without detriment to the patient. Theoretically, the use of exhaled breath analysis may allow for a more specific lung cancer diagnosis. However, only volatile compounds can be detected and genetic material is sparse or absent. Sputum has the advantage of perhaps giving results specific to lung cancer, as it contains both bronchial epithelial cells and other secretions reflecting the local milieu of the lung. However, it is difficult to obtain adequate sputum samples from the lower airways, and samples are frequently exclusively saliva. Urine is an easily accessible biofluid, but it may be less specific to lung cancer. Lung cancer biomarker research using urine as the biologic sample is still in its infancy.




Lung Cancer Biomarkers for Early Detection


Given the many different genetic and epigenetic changes involved in malignant transformation in lung cancer, it is not surprising that innumerable potential biomarkers exist. With progress in understanding the biology of lung carcinogenesis, the development of high-throughput techniques for biomarker discovery, and increased focus on early detection of lung cancer, the field of lung cancer biomarker research has expanded at a phenomenal rate. As yet, no biomarker has been shown to have adequate sensitivity, specificity, reproducibility, and ease of use to be validated as a biomarker for the early detection of lung cancer. However, many studies of biomarkers for the early diagnosis of lung cancer have shown promising results ( Table 8.1 ).



TABLE 8.1

Biomarkers Evaluated for Detection of Lung Cancers






































































































































































































































































































































































































































































































































































































































































































































































































Author (y) Type of Marker Type of Specimen Marker(s) No. of Markers Platform No. in Training Set No. in Test Set Sensitivity a (%) Specificity a (%) AUC a
Cytology
Varella-Garcia et al. (2004) Chromosomal aneusomy and cytology Sputum Multitarget DNA FISH assay and cytology 2 FISH 33 NR 83 80 NR
Xin et al. (2005) Sputum cytometry Sputum DNA content and cytologic malignancy grade 2 Automated DNA image cytometry 2461 NR 80 93 0.87
Kemp et al. (2007) Sputum cytometry Sputum Lung sign: Cell nuclear features (DNA content, chromatin distribution) 13 features Automated DNA image cytometry 1123 NR 40 91 0.69
Roy et al. (2010) Nanoarchitectural alterations Buccal epithelium Disorder strength of cell nanoarchitecture L (d) 1 Partial wave spectroscopic microscopy 207 46 78 78 0.84
Noncoding RNAs
Xing et al. (2010) MicroRNA Sputum miR-205, miR-210, miR-708 (squamous) 3 qRT-PCR 96 122 73 96 0.87
Xie et al. (2010) MicroRNA Sputum miR-21 1 qRT-PCR 50 NR 70 100 0.90
Yu et al. (2010) MicroRNA Sputum miRNA signature for adenocarcinoma 7 qRT-PCR 72 122 81 92 0.90
Bianchi et al. (2011) MicroRNA Serum miRNA signature 34 qRT-PCR 64 64 71 90 0.89
Boeri et al. (2011) MicroRNA Plasma miRNA signature 15 miRNA array and qRT-PCR 20 15 80 90 0.85
Boeri et al. (2011) MicroRNA Plasma miRNA signature 13 miRNA array and qRT-PCR 19 16 75 100 0.88
Shen et al. (2011) MicroRNA Plasma miR-21, miR-126, miR-210, miR-486-5p 4 qRT-PCR 28 87 86 97 0.93
Shen et al. (2011) MicroRNA Plasma miR-21, miR-210, miR-486-5p 3 qRT-PCR 94 156 75 85 0.86
Chen et al. (2012) 179 MicroRNA Serum miRNA signature 10 qRT-PCR 310 310 93 90 0.97
Hennessey (2012) MicroRNA Serum miR-15b and miR-27b 2 qRT-PCR 50 130 100 84 0.98
Patnaik et al. (2012) MicroRNA Whole blood miRNA signature 96 Locked nucleic acid microarrays 45 NR 88 89 0.94
Liao et al. (2010) Small nucleolar RNA Plasma snoRD33, snoRD66, and snoRD76 3 qRT-PCR 85 NR 81 96 0.88
Genetic Changes and Gene Expression
Miura et al. (2006) mRNA Serum Human telomerase catalytic component and epidermal growth factor receptor 2 qRT-PCR 192 NR 89 73 NR
Li et al. (2007) Genetic deletions Sputum FHIT and HYAL2 2 FISH 74 NR 76 92 NR
Spira et al. (2007) mRNA Airway epithelium Gene expression signature 80 Affymetrix array (Santa Clara, CA, USA) 77 52 80 84 NR
Blomquist et al. (2009) Gene expression Bronchial epithelium Antioxidant, DNA repair, and transcription factor genes 14 Standardized RT-PCR 49 40 82 80 0.87
Showe et al. (2009) Gene expression PBMC Gene signature 29 Illumina human whole genome bead array 228 NR 91 80 NR
Zander et al. (2011) Gene expression Whole blood Gene expression profile 484 Illumina human whole genome bead array 77 156 97 89 0.97
DNA Methylation
Palmisano et al. (2000) DNA methylation Sputum P16, O 6 -MGMT 2 PCR 144 NR 100 n/a NR
Kim et al. (2004) DNA methylation Bronchoalveolar lavage p16, RARβ, H-cadherin, RASSF1A 4 MS-PCR 212 NR 68 NR NR
Grote et al. (2004) DNA methylation Bronchial aspirates APC 1 qMS-PCR 222 NR 39 99 NR
Grote et al. (2005) DNA methylation Bronchial aspirates p16(INK4a), RARB2 2 qMS-PCR 139 NR 69 87 NR
Belinsky et al. (2006) DNA methylation Sputum p16, MGMT, DAPK, RASSF1A, PAX5β, GATA5 6 Nested MS-PCR 190 NR 64 64 NR
Grote et al. (2006) DNA methylation Bronchial aspirates RASSF1A 1 qMS-PCR 203 NR 46 100 NR
Ostrow et al. (2010) DNA methylation Plasma DCC, Kif1a, NISCH, Rarb 4 qRT-PCR 37 183 73 71 0.64
Schmidt et al. (2010) 180 DNA methylation Bronchial aspirates SHOX2 1 PCR n/a 523 68 95 0.86
Begum et al. (2011) DNA methylation Serum APC, CDH1, MGMT, DCC, RASSF1A, AIM 6 qPCR 401 106 84 57 NR
Kneip et al. (2011) 181 DNA methylation Plasma SHOX2 1 qPCR 40 371 60 90 0.78
Richards et al. (2011) 182 DNA methylation Lung tissues TCF21 1 PCR 42 63 76 98 NR
Protein and Proteomic Markers
Khan et al. (2004) Protein Serum Serum amyloid A 1 ELISA 50 NR 60 64 NR
Rahman et al. (2005) 183 Proteomic profile Bronchial biopsies TMLS4, ACBP, CSTA, cytoC, MIF, ubiquitin, ACBP, Des-ubiquitin 8 MALDI-MS 51 60 66 88 0.77
Patz et al. (2007) Protein panel Serum CEA, RBP, α1-antitrypsin, SCCA 4 ELISA 100 97 78 75 NR
Yildiz et al. (2007) Proteomic profile Serum Proteomic signature 7 features MALDI-MS 185 106 58 86 0.82
Farlow et al. (2010) Protein panel Serum TNFα, CYFRA 21-1, IL-1ra, MMP-2, MCP-1, and sE selectin 6 Luminex (Austin, TX, USA) and ELISA 133 88 99 95 0.98
Gessner et al. (2010) Proteins (cytokines) Exhaled breath condensate VEGF, bFGF, angiogenin 3 Multiplex bead-based immunoassay 75 NR 100 95 0.99
Ostroff et al. (2010) Aptamers Serum Aptamer signature 12 Aptamers 985 341 89 83 0.90
Joseph et al. (2012) Protein Plasma Osteopontin velocity 1 ELISA 43 NR 80 88 0.88
Lee et al. (2012) 184 Proteomics Serum AIAT, CYFRA 21-1, IGF-1, RANTES, AFP 5 Luminex 347 49 80.3 99.3 0.99
Higgins et al. (2012) Protein Plasma Variant Ciz1 1 Western blot 170 160 95 74 0.90
Ajona et al. (2013) Complement fragment Plasma C4d 1 Immunocytochemistry 190 NR NR NR 0.73
Patz et al. (2013) Protein panel, clinical features Serum CEA, α1-antitrypsin, SCCA, nodule size 4 ELISA 509 399 80 89 NR
Li et al. (2013) Protein panel Serum Protein panel 13 Multiple reaction monitoring mass spectrometry 143 104 71 44 NR
Autoantibodies and Tumor-Associated Antigens
Zhong et al. (2005) Autoantibodies Plasma Phage peptides 5 Fluorescent protein microarray 41 40 90 95 0.98
Zhong et al. (2006) Autoantibodies Serum Phage peptides 5 ELISA 46 56 91 91 0.99
Qiu et al. (2008) Autoantibodies Serum Annexin I, 14-3-3 theta, LAMR1 3 Protein array NR 170 51 82 0.73
Rom et al. (2010) Tumor-associated antigens Serum Panel of tumor-associated antigens 10 ELISA 194 NR 81 97 0.90
Wu et al. (2010) Autoantibodies Serum Phage peptide clones 6 ELISA 20 180 92 92 0.96
Boyle et al. (2011) Autoantibodies Serum p53, NY-ESO-1, CAGE, GBU4-5, annexin 1, SOX2 6 ELISA 241 255 32 91 0.64
Lam et al. (2011) Autoantibodies Serum p53, NY-ESO-1, CAGE, GBU4-5, annexin 1, SOX2 6 ELISA NR 1376 39 87 NR
Chapman et al. (2012) Autoantibodies Serum p53, NY-ESO-1, CAGE, GBU4-5, SOX2, HuD, and MAGE A4 7 ELISA 501 836 41 93 NR
Pedchenko et al. (2013) Autoantibodies Serum Single-chain fragment variable antibodies to IgM autoantibodies 6 Fluorometric microvolume and homogeneous bridging MESA SCALE DISCOVERY 30 43 80 87 0.88
Volatile Organic Compounds
Phillips et al. (1999) VOC Exhaled breath VOC profile 22 GC/MS 108 100 81 NR
Philips et al. (2003) VOC Exhaled breath VOC profile 9 GC/MS 178 108 85 80 NR
Poli et al. (2005) VOC Exhaled breath VOC profile 13 GC/MS 146 72 93 NR
Mazzone et al. (2007) VOC Exhaled breath VOC pattern 36 sensors Colorimetric sensor array 100 43 73 72 NR
Bajtarevic et al. (2009) VOC Exhaled breath VOC profile 21 Proton transfer reaction MS/solid-phase microextraction, GC/MS 96 NR 71 100 NR
Ligor et al. (2009) VOC Exhaled breath VOC profile 8 Solid-phase microextraction, GC/MS 96 NR 51 100 NR
Fuchs et al. (2010) VOC Exhaled breath Aldehydes: pentanal, hexanal, octanal, and nonanal 4 GC/MS 36 NR 75 96 NR

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Nov 10, 2019 | Posted by in ONCOLOGY | Comments Off on Preclinical Biomarkers for the Early Detection of Lung Cancer

Full access? Get Clinical Tree

Get Clinical Tree app for offline access