Next Article in Journal
Does Anxiety Increase the Risk of all-Cause Dementia? An Updated Meta-Analysis of Prospective Cohort Studies
Previous Article in Journal
Nonimmune Hydrops Fetalis—Prenatal Diagnosis, Genetic Investigation, Outcomes and Literature Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Biomarkers in Early Diagnosis and Early Stage Lung Cancer: The Clinician’s Point of View

1
Department of Thoracic Surgery, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy
2
Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2020, 9(6), 1790; https://doi.org/10.3390/jcm9061790
Submission received: 4 June 2020 / Accepted: 5 June 2020 / Published: 9 June 2020
(This article belongs to the Section Oncology)

Abstract

:
Starting from the work of Ulivi and colleagues, we aim to summarize the research area of biomarkers for early diagnosis and early stage lung cancer.

It was with great interest that we read the article by Ulivi et al. [1] on the prognostic value of micro RNAs (miRNAs) as a potential prognostic marker for patients who had undergone surgery for stage I–II non-small cell lung cancer (NSCLC). Although the study was well designed, it did suffer from substantial limitations as it was lacking in validation and had little statistical strength. However, it has given us the opportunity to take stock of the “biomarker,” as seen through the eyes of a clinician focusing on “early diagnosis” and the diagnosis of molecular residual disease (MRD) [2,3] after surgery for early-stage cancer.
Lung cancer is a pandemic disease responsible for the highest rate of cancer mortality worldwide. The total number of deaths per year far exceeds the total deaths caused by the other three solid tumors, namely breast, colon, and prostate [4,5]. There are two fundamental causes of this high mortality. The first is the late diagnosis in 85% of patients (often at a stage where the cancer has become locally advanced and metastatic); the second is due to the particular biological phenotype of this cancer that affects patients who have undergone radical surgical resection. In at least 30% of such cases, survival is jeopardized [6]. As a result of this, in clinical practice, specific needs persist, such as the need to diagnose lung cancer at an early stage, as well as the need to stratify the level of risk of recurrence after surgery for an early-stage tumor, in order to prescribe adjuvant therapy if need be. In this context, the discovery of one or more molecular biomarkers could meet these needs [7] by entering the diagnostic work-up before and after a low dose chest CT scan.
As things currently stand, in practice, the low-dose chest CT scan (LDCT), a straightforward and sensitive procedure, is the only examination performed for preventive, diagnostic, and follow-up purposes for patients undergoing radical stage I surgery. In 2011, an American study [8], which constitutes a milestone in the clinical community, using LDCT revealed a reduction in mortality of 20% compared with chest X-rays in a small group of at-risk subjects (heavy smokers).
Subsequently, in 2012 [9] several clinical limitations were discussed that remain unresolved. These include the need to extend the selection criteria for population screening, the high cost of the tests performed, and the high number of false positives. For many years, progress in “precision medicine” [10,11,12] through the “omics sciences” [13,14,15] has yielded a myriad of potential biomarkers [16,17] and biological information fundamental to the discovery of lung cancer vulnerability. For many years, researchers have focused on biomarkers that could affect the physician’s strategic choice [18]. The importance of their work is inestimable. Indeed, for metastatic lung cancer, it is common standard practice to carry out liquid biopsy [12]—now an everyday reality—available to the oncologist. Biomarkers such as EGFR [19] and ALK [20] are fundamental in guiding the biological therapeutic and immunotherapeutic choice for cancer patients with adenocarcinoma (ADK) [21]. Similarly, squamous cell carcinoma in PDL-1 positive patients [22] is treated by immunotherapy.
These biomarkers bring additional crucial information to the standards of care in creating subgroups (taxa) of patients for whom the clinician will be able to set up the specific medical, biological, or immunotherapeutic treatment plan. In this context, they have created a taxonomic classification [15] of non-small cell lung cancer, based not on the tumor histology, but rather on the biological and genetic phenotype of each patient. This finding emphasizes the fact that lung cancer forms a complex and heterogeneous biological system [23,24,25,26] in which a single histology can represent several subgroups with biological microsystems that differ [27].
Assessing the published, peer-reviewed biomedical studies on biomarkers for early diagnosis and MRD post-surgery, one encounters an enormous amount of outstanding scientific work that reveals “signatures,” extracted from different biological fluids [28], which can potentially be employed in the clinic.
Below we will briefly highlight potential biomarkers, which have been analyzed on extraction from the blood, exhaled breath, and urine; three fluids that we believe to be clinically ideal for the choice of a future rapid, non-invasive, and scientifically robust test.
For early diagnosis, there are several potential biomarkers.
In the blood, autoantibodies and antigens have been evaluated [29,30,31,32], such as C4d [33]. Additionally, research has been conducted not only into miRNA [34] combined with an LDCT scan [35,36] but also into circulating tumor DNA [37,38] for which it will be essential to await the results of the Circulating Cell-free Genome Atlas Study [39]; the proteomic profile is also added to the potential signatures of early diagnosis [40,41].
In respiratory exhaled breath, an emerging research front, volatile organic compounds (VOCs), collected through simple spirometry, are being evaluated. The analysis can be performed by using gaseous mass spectrometers [42,43] to evaluate the molecular quality, and also by using artificial olfactory devices equipped with sensors that create volatile imprints, according to physical-chemical mechanisms that can differentiate healthy individuals from those with cancer [44]. It will be interesting to wait for the results of the Lung Cancer Indicator Detection (LuCID) study, which aims to validate the use of a high-throughput (www.clinicaltrials.NCT02612532) breath analysis technique.
In urine, which is abundant and easy to sample without any invasiveness, the use of liquid spectrometry [45] makes it easy to extract isolated metabolites [46] in protein panels [47]. Potentially useful for both the early diagnostic and prognostic phases [47,48,49], even though at present they are included in a discovery phase, we believe that this biological fluid can be combined with other biological fluids to create a simple, rapid and reproducible test [50].
Regarding post-surgery, for the diagnosis of MRD, the blood-based next generation of sequencing [51,52] (NGS) alongside comprehensive multiparameter analyses [53,54] have revealed multiple gene sequences [55,56], gene isolates [57], panels of serum proteins and autoantibody [58], panels of serum tumor antigen and autoantibodies [59], and circulating tumor cells [60]. We also note research published by E. Martinez-Terroba [61] et al. in which they identified and validated a protein-based signature, which is very promising in terms of technology used, simplicity and affordability, scientific robustness, and the potential for translation into clinical practice. Another study of notable scientific quality and simplicity is one published in 2014 by Harris [62].
The above shows the enormous dedication of researchers striving to validate specific biomarkers for both early diagnosis and post-surgery MRD. On the other hand, there is the absence of a valid and usable biomarker in the clinical context. Given the biology of lung cancer, we believe that in the future, multiple clinical-molecular-epidemiological profiles will be obtained that can identify and stratify the risk level of lung cancer. This expectation is yet to be met, due to the difficulty of translating most of the results obtained by the researchers into a clinical context. We believe in laboratory research in the clinical setting, at the patient’s bedside. It is our heartfelt hope that the individual, whether healthy or sick and who of course is central to everything, will one day be screened primarily through a non-invasive molecular test, involving sample collection of their biological fluids. The information arising from this will be combined with their medical history and epidemiological data and then possibly be followed by a low-dose CT scan.
In this regard, in our clinical institute, in both healthy patients and patients operated on for an early-stage tumor we are taking samples of blood, exhaled breath, and urine as part of a monocentric prospective study for the discovery of biomarkers. For healthy subjects, these three samples are taken in a hospital outpatient clinic; for those patients who have undergone surgery, samples are taken at the bedside. We have ascertained the logistical and technical ease of handling the three samples under the supervision of the research nurse and biologist. All the researchers involved meet regularly in our hospital and work closely with the clinicians to improve the technical aspects associated with the scientific workup of the project.
We would like to conclude by stressing the necessity to develop projects in the future, ideally designed in clinical contexts, to translate the results from research into the application at the bedside. It will also be crucial to process the results employing standard robust statistical methods and to adhere strictly to the five steps required for the validation of each potential biomarker [63,64]. It is undoubtedly crucial to persist in the relentless search for evidence suggestive of vulnerability to lung cancer in order to weaken this ruthless killer. Each country will need to collect all the data gathered and make it transparently available in the public domain. In this way, comparisons can be drawn between data from other countries for the common purpose of creating a worldwide Big Data database [65]. Artificial intelligence [66,67] will be used to process, overlap, and integrate the molecular, clinical, and epidemiological data, and machine learning [68,69] will be employed to produce realistic multiple diagnostic algorithms which the clinician can readily use for diagnostic, preventive, and prognostic purposes.

Author Contributions

Conceptualization: R.G.; writing—original draft preparation: R.G.; writing—review and editing: R.G., G.S., L.S.; supervision, R.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by the Italian Ministry of Health with Ricerca Corrente and 5 × 1000 funds.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ulivi, P.; Petracci, E.; Marisi, G.; Baglivo, S.; Chiari, R.; Billi, M.; Canale, M.; Pasini, L.; Racanicchi, S.; Vagheggini, A.; et al. Prognostic role of circulating miRNAs in Early-Stage Non-Small Cell Lung Cancer. J. Clin. Med. 2019, 8, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. McGuire, A.L.; Hughesman, C.B.; McConechy, M.K.; Melosky, B.; Lam, S.; Myers, R.; Yee, J.; Tang, E.; Yip, S. Optimizing molecular residual disease detection using liquid biopsy postoperatively in early stage lung cancer. Lung Cancer Manag. 2020, 9, LMT24. [Google Scholar] [CrossRef] [PubMed]
  3. Pantel, K.; Alix-Panabières, C. Liquid biopsy and minimal residual Disease-Latest advances and implications for cure. Nat. Rev. Clin. Oncol. 2019, 16, 409–424. [Google Scholar] [CrossRef]
  4. Fitzmaurice, C.; Abate, D.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdel-Rahman, O.; Abdelalim, A.; Abdoli, A.; Abdollahpour, I.; Abdulle, A.S.M.; et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and Disability-Adjusted Life-Years for 29 cancer groups, 1990 to 2017: A systematic analysis for the global burden of disease study. JAMA Oncol. 2019, 5, 1749–1768. [Google Scholar]
  5. Abubakar, I.I.; Tillmann, T.; Banerjee, A. Global, regional, and national Age–Sex specific All-Cause and Cause-Specific mortality for 240 causes of death, 1990–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2015, 385, 117–171. [Google Scholar]
  6. Goldstraw, P.; Crowley, J.; Chansky, K.; Giroux, D.J.; Groome, P.A.; Rami-Porta, R.; Postmus, P.E.; Rusch, V.; Sobin, L. The IASLC lung cancer staging project: Proposals for the revision of the tnm stage groupings in the forthcoming (seventh) edition of the TNM classification of malignant tumours. J. Thorac. Oncol. 2007, 2, 706–714. [Google Scholar] [CrossRef] [Green Version]
  7. Sears, C.R.; Mazzone, P.J. Biomarkers in lung cancer. Clin. Chest Med. 2020, 41, 115–127. [Google Scholar] [CrossRef] [PubMed]
  8. National Lung Screening Trial Research Team; Aberle, D.R.; Adams, A.M.; Berg, C.D.; Black, W.C.; Clapp, J.D.; Fagerstrom, R.M.; Gareen, I.F.; Gatsonis, C.; Marcus, P.M.; et al. The national lung screening trial research team reduced Lung-Cancer mortality with Low-Dose computed tomographic screening. N. Engl. J. Med. 2011, 365, 395–409. [Google Scholar]
  9. Bach, P.B.; Mirkin, J.; Oliver, T.K.; Azzoli, C.G.; Berry, D.; Brawley, O.W.; Byers, T.; Colditz, G.A.; Gould, M.K.; Jett, J.R.; et al. Benefits and harms of CT screening for lung cancer: A systematic review. JAMA 2012, 307, 2418–2429. [Google Scholar] [CrossRef] [Green Version]
  10. Schwaederle, M.; Daniels, G.A.; Piccioni, D.E.; Fanta, P.T.; Schwab, R.B.; Shimabukuro, K.A.; Parker, B.A.; Kurzrock, R. On the road to precision cancer medicine: Analysis of genomic biomarker actionability in 439 patients. Mol. Cancer Ther. 2015, 14, 1488–1494. [Google Scholar] [CrossRef] [Green Version]
  11. Yamashita, K.; Hosoda, K.; Nishizawa, N.; Katoh, H.; Watanabe, M. Epigenetic biomarkers of promoter DNA methylation in the new era of cancer treatment. Cancer Sci. 2018, 109, 3695–3706. [Google Scholar] [CrossRef] [PubMed]
  12. Heitzer, E.; Haque, I.S.; Roberts, C.E.S.; Speicher, M.R. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat. Rev. Genet. 2018, 20, 71–88. [Google Scholar] [CrossRef] [PubMed]
  13. De Anda-Jáuregui, G.; Hernández-Lemus, E. Computational Oncology in the Multi-Omics Era: State of the Art. Front. Oncol. 2020, 10, 423. [Google Scholar] [CrossRef] [PubMed]
  14. Seijo, L.M.; Peled, N.; Ajona, D.; Boeri, M.; Field, J.; Sozzi, G.; Pio, R.; Zulueta, J.J.; Spira, A.; Massion, P.P.; et al. Biomarkers in lung cancer screening: Achievements, promises, and challenges. J. Thorac. Oncol. 2019, 14, 343–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Robles, A.I.; Harris, C.C. Integration of multiple “OMIC” biomarkers: A precision medicine strategy for lung cancer. Lung Cancer 2016, 107, 50–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Mishra, A.; Verma, M. Cancer Biomarkers: Are we ready for the prime time? Cancers 2010, 2, 190–208. [Google Scholar] [CrossRef]
  17. Inage, T.; Nakajima, T.; Yoshino, I.; Yasufuku, K. Early lung cancer detection. Clin. Chest Med. 2018, 39, 45–55. [Google Scholar] [CrossRef]
  18. Mazzone, P.J.; Sears, C.R.; Arenberg, D.A.; Gaga, M.; Gould, M.K.; Massion, P.P.; Nair, V.S.; Powell, C.A.; Silvestri, G.A.; Vachani, A.; et al. Evaluating molecular biomarkers for the early detection of lung cancer: When is a biomarker ready for clinical use? An official American thoracic society policy statement. Am. J. Respir. Crit. Care Med. 2017, 196, e15–e29. [Google Scholar] [CrossRef]
  19. Duffy, M.J.; O’Byrne, K. Tissue and blood biomarkers in lung cancer: A review. Adv. Clin. Chem. 2018, 86, 1–21. [Google Scholar]
  20. Arbour, K.C.; Riely, G.J. Systemic therapy for locally advanced and metastatic Non-Small cell lung cancer: A review. JAMA 2019, 322, 764–774. [Google Scholar] [CrossRef]
  21. Hirsch, F.R.; Scagliotti, G.V.; Mulshine, J.L.; Kwon, R.; Curran, W.J.; Wu, Y.-L.; Paz-Ares, L. Lung cancer: Current therapies and new targeted treatments. Lancet 2017, 389, 299–311. [Google Scholar] [CrossRef]
  22. Paulsen, E.-E.; Kilvaer, T.K.; Khanehkenari, M.R.; Al-Saad, S.; Hald, S.M.; Andersen, S.; Richardsen, E.; Ness, N.; Busund, L.-T.; Bremnes, R.M.; et al. Assessing PDL-1 and PD-1 in Non–Small Cell Lung Cancer: A novel immunoscore approach. Clin. Lung Cancer 2017, 18, 220–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. De Sousa, V.M.L.; Carvalho, L. Heterogeneity in lung cancer. Pathobiology 2018, 85, 96–107. [Google Scholar] [CrossRef]
  24. Gerlinger, M.; Horswell, S.; Larkin, J.; Rowan, A.J.; Salm, M.; Varela, I.; Fisher, R.; McGranahan, N.; Matthews, N.; Santos, C.R.; et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 2014, 46, 225–233. [Google Scholar] [CrossRef] [PubMed]
  25. Bedard, P.L.; Hansen, A.R.; Ratain, M.J.; Siu, L.L. Tumour heterogeneity in the clinic. Nature 2013, 501, 355–364. [Google Scholar] [CrossRef] [Green Version]
  26. Marusyk, A.; Polyak, K. Tumor heterogeneity: Causes and consequences. Biochim. Biophys. Acta Rev. Cancer 2010, 1805, 105–117. [Google Scholar] [CrossRef] [Green Version]
  27. Gerlinger, M.; Rowan, A.J.; Horswell, S.; Math, M.; Larkin, J.; Endesfelder, D.; Grönroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012, 366, 883–892. [Google Scholar] [CrossRef] [Green Version]
  28. Hofman, P. The challenges of evaluating predictive biomarkers using small biopsy tissue samples and liquid biopsies from Non-Small cell lung cancer patients. J. Thorac. Dis. 2019, 11, S57–S64. [Google Scholar] [CrossRef]
  29. Edelsberg, J.; Weycker, D.; Atwood, M.; Hamilton-Fairley, G.; Jett, J.R. Cost-Effectiveness of an autoantibody test (EarlyCDT-Lung) as an aid to early diagnosis of lung cancer in patients with incidentally detected pulmonary nodules. PLoS ONE 2018, 13, e0197826. [Google Scholar] [CrossRef]
  30. Boyle, P.; Chapman, C.J.; Holdenrieder, S.; Murray, A.; Robertson, C.; Wood, W.C.; Maddison, P.; Healey, G.; Fairley, G.H.; Barnes, A.C.; et al. Clinical validation of an autoantibody test for lung cancer. Ann. Oncol. 2011, 22, 383–389. [Google Scholar] [CrossRef]
  31. Lam, S.; Boyle, P.; Healey, G.F.; Maddison, P.; Peek, L.; Murray, A.; Chapman, C.J.; Allen, J.; Wood, W.C.; Sewell, H.F.; et al. EarlyCDT-Lung: An immunobiomarker test as an aid to early detection of lung cancer. Cancer Prev. Res. 2011, 4, 1126–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Chen, D.-T.; Hsu, Y.-L.; Fulp, W.J.; Coppola, M.; Haura, E.B.; Yeatman, T.J.; Cress, W.D. Prognostic and predictive value of a Malignancy-Risk gene signature in Early-Stage Non–Small cell lung cancer. J. Natl. Cancer Inst. 2011, 103, 1859–1870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ajona, D.; Okrój, M.; Pajares, M.J.; Agorreta, J.; Lozano, M.D.; Zulueta, J.J.; Verri, C.; Roz, L.; Sozzi, G.; Pastorino, U.; et al. Complement C4d-Specific antibodies for the diagnosis of lung cancer. Oncotarget 2018, 9, 6346–6355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhang, Y.; Sui, J.; Shen, X.; Li, C.; Yao, W.; Hong, W.; Peng, H.; Pu, Y.; Yin, L.; Liang, G. Differential expression profiles of microRNAs as potential biomarkers for the early diagnosis of lung cancer. Oncol. Rep. 2017, 37, 3543–3553. [Google Scholar] [CrossRef] [Green Version]
  35. Sozzi, G.; Boeri, M.; Rossi, M.; Verri, C.; Suatoni, P.; Bravi, F.; Roz, L.; Conte, D.; Grassi, M.; Sverzellati, N.; et al. Clinical utility of a Plasma-Based miRNA signature classifier within computed tomography lung cancer screening: A correlative mild trial study. J. Clin. Oncol. 2014, 32, 768–773. [Google Scholar] [CrossRef]
  36. Montani, F.; Marzi, M.J.; Dezi, F.; Dama, E.; Carletti, R.M.; Bonizzi, G.; Bertolotti, R.; Bellomi, M.; Rampinelli, C.; Maisonneuve, P.; et al. MIR-Test: A blood test for lung cancer early detection. J. Natl. Cancer Inst. 2015, 107, djv063. [Google Scholar] [CrossRef] [Green Version]
  37. Chaudhuri, A.A.; Chabon, J.J.; Lovejoy, A.F.; Newman, A.M.; Stehr, H.; Azad, T.; Khodadoust, M.S.; Esfahani, M.S.; Liu, C.L.; Zhou, L.; et al. Early detection of molecular residual disease in localized lung cancer by circulating tumor DNA profiling. Cancer Discov. 2017, 7, 1394–1403. [Google Scholar] [CrossRef] [Green Version]
  38. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.V.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and localization of surgically resectable cancers with a Multi-Analyte blood test. Science 2018, 359, 926–930. [Google Scholar] [CrossRef] [Green Version]
  39. Oxnard, G.R.; Maddala, T.; Hubbell, E.; Aravanis, A.; Zhang, N.; Venn, O.; Valouev, A.; Shen, L.; Patel, S.; Jamshidi, A.; et al. Genome-Wide sequencing for early stage lung cancer detection from plasma Cell-Free DNA (cfDNA): The circulating cancer genome atlas (CCGA) study. J. Clin. Oncol. 2018, 36, LBA8501. [Google Scholar] [CrossRef]
  40. Gasparri, R.; Sedda, G.; Noberini, R.; Bonaldi, T.; Spaggiari, L. Clinical application of mass Spectrometry-Based proteomics in lung cancer early diagnosis. PROTEOM Clin. Appl. 2020. [Google Scholar] [CrossRef]
  41. Li, D.; Yang, W.; Zhang, Y.; Yang, J.Y.; Guan, R.; Xu, N.; Yang, M.Q. Genomic analyses based on pulmonary adenocarcinoma in situ reveal early lung cancer signature. BMC Med Genom. 2018, 11, 106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Mu, Y.; Zhou, Y.; Wang, Y.; Li, W.; Zhou, L.; Lu, X.; Gao, P.; Gao, M.; Zhao, Y.; Wang, Q.; et al. Serum metabolomics study of nonsmoking female patients with non-small cell lung cancer using gas Chromatography–Mass spectrometry. J. Proteome Res. 2019, 18, 2175–2184. [Google Scholar] [CrossRef] [PubMed]
  43. Callejón-Leblic, B.; García-Barrera, T.; Pereira-Vega, A.; Gómez-Ariza, J.L. Metabolomic study of serum, urine and bronchoalveolar lavage fluid based on gas chromatography mass spectrometry to delve into the pathology of lung cancer. J. Pharm. Biomed. Anal. 2019, 163, 122–129. [Google Scholar] [CrossRef] [PubMed]
  44. Gasparri, R.; Santonico, M.; Valentini, C.; Sedda, G.; Borri, A.; Petrella, F.; Maisonneuve, P.; Pennazza, G.; D’Amico, A.; Di Natale, C.; et al. Volatile signature for the early diagnosis of lung cancer. J. Breath Res. 2016, 10, 16007. [Google Scholar] [CrossRef]
  45. Heavner, D.L.; Richardson, J.D.; Morgan, W.T.; Ogden, M.W. Validation and application of a method for the determination of nicotine and five major metabolites in smokers’ urine by Solid-Phase extraction and liquid Chromatography-Tandem mass spectrometry. Biomed. Chromatogr. 2005, 19, 312–328. [Google Scholar] [CrossRef]
  46. Carrola, J.; Rocha, C.; Barros, A.; Gil, A.M.; Goodfellow, B.; Carreira, I.M.; Bernardo, J.; Gomes, A.; De Sousa, V.M.L.; Carvalho, L.; et al. Metabolic signatures of lung cancer in biofluids: NMR-Based metabonomics of urine. J. Proteome Res. 2011, 10, 221–230. [Google Scholar] [CrossRef]
  47. An, Z.; Chen, Y.; Zhang, R.; Song, Y.; Sun, J.; He, J.; Bai, J.; Dong, L.; Zhan, Q.; Abliz, Z. Integrated Ionization Approach for RRLC−MS/MS-based metabonomics: Finding potential biomarkers for lung cancer. J. Proteome Res. 2010, 9, 4071–4081. [Google Scholar] [CrossRef]
  48. Yang, Q.; Shi, X.; Wang, Y.; Wang, W.; He, H.; Lü, X.; Xu, G. Urinary metabonomic study of lung cancer by a fully automatic hyphenated hydrophilic interaction/RPLC-MS system. J. Sep. Sci. 2010, 33, 1495–1503. [Google Scholar] [CrossRef]
  49. Nolen, B.M.; Lomakin, A.; Marrangoni, A.; Velikokhatnaya, L.; Prosser, D.; Lokshin, A. Urinary protein biomarkers in the early detection of lung cancer. Cancer Prev. Res. 2014, 8, 111–119. [Google Scholar] [CrossRef] [Green Version]
  50. Mathe, E.A.; Patterson, A.; Haznadar, M.; Manna, S.K.; Krausz, K.W.; Bowman, E.D.; Shields, P.G.; Idle, J.; Smith, P.B.; Anami, K.; et al. Noninvasive urinary metabolomic profiling identifies diagnostic and prognostic markers in lung cancer. Cancer Res. 2014, 74, 3259–3270. [Google Scholar] [CrossRef] [Green Version]
  51. Lai, J.; Du, B.; Wang, Y.; Wu, R.; Yu, Z. Next-Generation sequencing of circulating tumor DNA for detection of gene mutations in lung cancer: Implications for precision treatment. OncoTargets Ther. 2018, 11, 9111–9116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Liang, W.; Zhao, Y.; Huang, W.; Gao, Y.; Xu, W.; Tao, J.; Yang, M.; Li, L.; Ping, W.; Shen, H.; et al. Non-Invasive diagnosis of Early-Stage lung cancer using High-Throughput targeted DNA methylation sequencing of circulating tumor DNA (ctDNA). Theranostics 2019, 9, 2056–2070. [Google Scholar] [CrossRef] [PubMed]
  53. Barrett, I.P. Cancer genome analysis informatics. In Choice Reviews Online; Barnes, M.R., Breen, G., Eds.; Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2010; Volume 628, pp. 75–102. [Google Scholar]
  54. Chang, J.T.-H.; Lee, Y.-M.; Huang, R.S. The impact of the Cancer Genome Atlas on lung cancer. Transl. Res. 2015, 166, 568–585. [Google Scholar] [CrossRef] [Green Version]
  55. Srivastava, M.; Khurana, P.; Sugadev, R. Lung cancer signature biomarkers: Tissue specific semantic similarity based clustering of Digital Differential Display (DDD) data. BMC Res. Notes 2012, 5, 617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Goncalves, R.; Bose, R. Using multigene tests to select treatment for Early-Stage breast cancer. J. Natl. Compr. Cancer Netw. 2013, 11, 174–182. [Google Scholar] [CrossRef]
  57. Okayama, H.; Schetter, A.J.; Ishigame, T.; Robles, A.I.; Kohno, T.; Yokota, J.; Takenoshita, S.; Harris, C.C. The Expression of Four Genes as a Prognostic Classifier for Stage I Lung Adenocarcinoma in 12 Independent Cohorts. Cancer Epidemiol. Biomarkers Prev. 2014, 23, 2884–2894. [Google Scholar] [CrossRef] [Green Version]
  58. Mazzone, P.J.; Wang, X.-F.; Han, X.; Choi, H.; Seeley, M.; Scherer, R.; Doseeva, V. Evaluation of a Serum Lung Cancer Biomarker Panel. Biomark. Insights 2018, 13. [Google Scholar] [CrossRef] [Green Version]
  59. 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. [Google Scholar] [CrossRef] [Green Version]
  60. Abbosh, C.; Birkbak, N.J.; Wilson, G.A.; Jamal-Hanjani, M.; Constantin, T.; Salari, R.; Le Quesne, J.; Moore, D.A.; Veeriah, S.; Rosenthal, R.; et al. Phylogenetic ctDNA analysis depicts Early-Stage lung cancer evolution. Nature 2017, 545, 446–451. [Google Scholar] [CrossRef]
  61. Martínez-Terroba, E.; Behrens, C.; De Miguel, F.J.; Agorreta, J.; Monsó, E.; Millares, L.; Sainz, C.; Mesa-Guzman, M.; Perez-Gracia, J.L.; Lozano, M.D.; et al. A novel protein-based prognostic signature improves risk stratification to guide clinical management in Early-Stage lung adenocarcinoma patients. J. Pathol. 2018, 245, 421–432. [Google Scholar] [CrossRef]
  62. Okayama, A.; Miyagi, Y.; Oshita, F.; Nishi, M.; Nakamura, Y.; Nagashima, Y.; Akimoto, K.; Ryo, A.; Hirano, H. Proteomic analysis of proteins related to prognosis of lung adenocarcinoma. J. Proteome Res. 2014, 13, 4686–4694. [Google Scholar] [CrossRef] [PubMed]
  63. Pepe, M.S.; Etzioni, R.; Feng, Z.; Potter, J.D.; Thompson, M.L.; Thornquist, M.; Winget, M.; Yasui, Y. Phases of biomarker development for early detection of cancer. J. Natl. Cancer Inst. 2001, 93, 1054–1061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Pepe, M.S.; Janes, H.; Li, C.I.; Bossuyt, P.M.; Feng, Z.; Hilden, J. Early-Phase studies of biomarkers: What target sensitivity and specificity values might confer clinical utility? Clin. Chem. 2016, 62, 737–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Global Burden of Disease Cancer Collaboration; Fitzmaurice, C.; Akinyemiju, T.F.; Al Lami, F.H.; Alam, T.; Alizadeh-Navaei, R.; Allen, C.; Alsharif, U.; Alvis-Guzman, N.; Amini, E.; et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2016: A systematic analysis for the global burden of disease study. JAMA Oncol. 2018, 4, 1553–1568. [Google Scholar]
  66. Topol, E.J. High-Performance medicine: The convergence of human and artificial intelligence. Nat. Med. 2019, 25, 44–56. [Google Scholar] [CrossRef]
  67. Miller, D.D.; Brown, E.W. Artificial intelligence in medical practice: The question to the answer? Am. J. Med. 2017, 131, 129–133. [Google Scholar] [CrossRef]
  68. Burki, T.K. Predicting lung cancer prognosis using machine learning. Lancet Oncol. 2016, 17, e421. [Google Scholar] [CrossRef]
  69. Goecks, J.; Jalili, V.; Heiser, L.M.; Gray, J.W. How machine learning will transform biomedicine. Cell 2020, 181, 92–101. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Gasparri, R.; Sedda, G.; Spaggiari, L. Biomarkers in Early Diagnosis and Early Stage Lung Cancer: The Clinician’s Point of View. J. Clin. Med. 2020, 9, 1790. https://doi.org/10.3390/jcm9061790

AMA Style

Gasparri R, Sedda G, Spaggiari L. Biomarkers in Early Diagnosis and Early Stage Lung Cancer: The Clinician’s Point of View. Journal of Clinical Medicine. 2020; 9(6):1790. https://doi.org/10.3390/jcm9061790

Chicago/Turabian Style

Gasparri, Roberto, Giulia Sedda, and Lorenzo Spaggiari. 2020. "Biomarkers in Early Diagnosis and Early Stage Lung Cancer: The Clinician’s Point of View" Journal of Clinical Medicine 9, no. 6: 1790. https://doi.org/10.3390/jcm9061790

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop