Next Article in Journal
Colorectal Cancer Anatomical Site and Sleep Quality
Previous Article in Journal
Effects of Cancer, Chemotherapy, and Cytokines on Subjective and Objective Cognitive Functioning Among Patients with Breast Cancer
Previous Article in Special Issue
Genomic Characterization of Concurrent Alterations in Non-Small Cell Lung Cancer (NSCLC) Harboring Actionable Mutations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Diagnostic and Predictive Biomarkers in Lung Cancer

by
Caterina Fumagalli
* and
Massimo Barberis
*
Division of Pathology, IEO, European Institute of Oncology, IRCCS, 20141 Milano, Italy
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(11), 2577; https://doi.org/10.3390/cancers13112577
Submission received: 19 May 2021 / Accepted: 23 May 2021 / Published: 25 May 2021
(This article belongs to the Special Issue Diagnostic and Predictive Biomarkers in Lung Cancer)
Lung cancer is the most frequent cause of cancer-related mortality worldwide. An early diagnosis, the identification of conditions predisposing to lung cancer, and the definition of the tumor genetic profile are crucial steps to improved patient outcomes. In the present special issue, “Diagnostic and Predictive Biomarkers in Lung Cancer”, these important topics are covered across fourteen peer-reviewed papers produced by recognized experts on the subject.
We have the pleasure of presenting a multicenter European study coordinated by Giulia Veronesi: “Lung Cancer Screening with Low-Dose Computed Tomography” [1]. The authors stated that the implementation of such screening is urgently needed in Europe in order to diagnose lung cancer at the early stages and reduce mortality rates. The group propose shared recommendations such as the implementation of a risk-prediction model to select high-risk populations or the management of lung nodules by utilizing volume and growth-rate data with more stringent cut-offs.
On a different topic, specific conditions such as idiopathic pulmonary fibrosis (IPF), a severe progressive interstitial lung disease, may be associated with lung cancer development. Indeed, up to 15% of IPF patients develop lung cancer within five years. Miriana D’Alessandro and colleagues [2,3] completed two studies focused on the identification of biomarkers linked to prognosis and therapy response in patients affected by idiopathic pulmonary fibrosis. In detail, the authors reported the utility of the most common, routinely available oncomarkers (namely NSE, CEA, Ca19.9, and Ca125) in the evaluation of IPF patients’ clinical courses [2]. They also identified the serum concentration of Krebs von den Lungen-6 (KL-6) as a helpful biomarker to be monitored in IPF patients during antifibrotic therapy, as stable levels were associated with stabilization of lung function parameters. Moreover, a KL-6 serum level increase was observed in IPF patients who developed lung cancer and was associated with poor prognosis [3]. Both these studies add valuable insight into the clinical management and follow up of IPF patients.
Furthermore, we should consider that 30% of patients are diagnosed at an early stage, and have resectable lung cancer. In these cases, a surgical approach of lobectomy/pneumonectomy could be applied. The interesting study by Marjanski and colleagues [4] evaluated a useful “low-tech” exercise of a 6 min walking test (6MWT) for prediction of complications and overall survival after pneumonectomy. As reported, patients with poor results in the 6MWT the day preceding the surgery were at high risk of postoperative cardiac complications and had poor overall survival.
Although great efforts have been made to improve screening and early tumor detection, up to 70% of patients are diagnosed at advanced stages and have unresectable disease. In advanced and metastatic contexts, the genomic portrait of the tumor takes on a pivotal role in therapy selection. Indeed, the detection of oncogene driver mutations in non-small-cell lung cancer (NSCLC) can be used to identify patients with potential survival benefit from targeted therapies, and PD-L1-positive staining in oncogenic-not-addicted tumors can help to identify patients as candidates for immunotherapy. However, the potential therapeutic options have moved beyond the standard-of-care EGFR/BRAF/ALK/ROS1 and PD-L1 evaluations. In this evolving context, where the number of actionable genetic alterations to be tested is constantly increasing but the tumor tissue may be scarce, a comprehensive genomic profile determined using next-generation sequencing (NGS) panels could be the most effective compromise. How to implement tumor genomic profiling in routine clinical settings is still debated; the Italian Association of Thoracic Oncology (AIOT) produced a position paper summarizing the results of a discussion from a Precision Medicine panel meeting on the challenges facing the introduction of a comprehensive genomic profile (CGP) into daily clinical practice [5]. The study conducted by De Luca and colleagues [6] presents a narrow RNA fusion panel used to investigate gene fusions and splicing events that could be successfully implemented in a routine diagnostic setting. Antonio Passaro and coworkers [7] discussed the characterization of concurrent driver alterations in NSCLC, which is helpful for personalized treatment selection and detection of resistance. Moreover, NGS analysis could increase understanding of the biology of tumor development and progression. As reported by Centoze [8], the four World Health Organization (WHO) classes of lung neuroendocrine neoplasms are characterized by specific molecular alterations that could predict the clinical course and guide therapy selection. Nevertheless, performing a CGP of NSCLCs may produce a plethora of molecular data that are tricky to manage and difficult to translate into clinical indications. To this end, we report the experience of a multidisciplinary molecular tumor board (MTB) as a valuable tool to support patients’ physicians to apply precision medicine strategies [9].
The field of biomarkers in NSCLC is protean: different targets, different methods, and different clinical contexts. Besides tissue-based markers, ctDNA, proteomic, epigenomic, and metabolomic markers have recently enlarged this constantly evolving panorama. In particular, the liquid biopsy, based on the detection of tumor-related biomarkers in body fluids such as peripheral blood, has emerged as a very promising and noninvasive diagnostic tool that can capture spatial and temporal intratumoral heterogeneity. Florian Janke and coworkers [10] described how liquid biomarkers can be tested to assess responses to therapy in advanced NSCLCs. The use of liquid biomarker panels might help to adjust treatment earlier than currently possible using radiographic tumor assessments, and thus avoid or shorten the side effects and adverse events associated with ineffective treatment. Paul Hofman [11] elegantly discussed the impact of next-generation sequencing in its application to liquid biopsies in treatment-naïve NSCLC patients. The author thoroughly investigated different issues regarding the implementation of liquid biopsy as a complementary or alternative option to tissue biopsy for treatment-naïve advanced NSCLC patients. Although this is a very exciting new area, NGS performed on circulating nucleic acids at the point of diagnosis may have limitations, and we should be aware that negative results could be due to the lower level of performance of molecular biology analyses performed via liquid biopsy compared with tissue biopsy.
The main goal in the clinical management of lung cancer patients is to schedule the most effective course of treatment for the individual patient, as different drugs are available and may be used in combination or sequentially to overcome resistance mechanisms. Towards this aim, different elements should be considered, including tumor-related and tumor-independent factors. Indeed, sex-based differences could also affect disease clinical courses. Pérez-Díez and coworkers [12] compared transcriptomic data obtained from lung adenocarcinomas that developed in male and female patients and highlighted different functional signatures that could influence tumor progression and response to therapy. Regarding the benefits of immunotherapy, nontumor factors such as the host immune system or microbiota could play a central role. To date, PD-L1 immunostaining alone may not be sufficient, but new biomarkers are emerging, such as tumor mutational burden (TMB) and tumor-infiltrating lymphocytes (TILs). On this topic, Sesma and colleagues [13] provide a review, identifying tumor mutational burden (TMB) and blood T-cell receptor TCRβ (TCRB) as promising predictive biomarkers for immunotherapy response.
Lastly, the evolving molecular landscape of neuroendocrine tumors of the lung, mainly small-cell lung cancers (SCLC) and large-cell neuroendocrine carcinomas (LCNEC) deserves a special mention for its therapeutic implications. Different chemotherapy schemes applied according to molecular biomarkers could offer new opportunities to patients with LCNEC. The study of Carazo and colleagues [14] reports an innovative genome-wide loss-of-function screen that may be useful for identifying biomarkers predictive of therapy response, especially for tumors with scarce therapeutic options. Applying this tool, the authors identified CREBBP-defective SCLC to be vulnerable to polo-like kinase 1 (PLK1) inhibition.
In conclusion, precision cancer medicine is merging the tumor genomic alteration landscape with available therapies, aiming to improve patient outcomes. The great challenge for the pathologist is to guide treatment accurately, which requires going beyond diagnosis and tumor classification to give clinically useful diagnostic, prognostic, and predictive parameters within a timeframe consistent with clinical needs.
This special issue of Cancers highlights the current state-of-the-art technology in diagnostic and predictive assays used for lung cancer treatment, with special emphasis on future prospects in early diagnosis, biomarker integration, and tumor response evaluation.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Veronesi, G.; Baldwin, D.R.; Henschke, C.I.; Ghislandi, S.; Iavicoli, S.; Oudkerk, M.; De Koning, H.J.; Shemesh, J.; Field, J.K.; Zulueta, J.J.; et al. Recommendations for Implementing Lung Cancer Screening with Low-Dose Computed Tomography in Europe. Cancers 2020, 12, 1672. [Google Scholar] [CrossRef] [PubMed]
  2. d’Alessandro, M.; Bergantini, L.; Torricelli, E.; Cameli, P.; Lavorini, F.; Pieroni, M.; Refini, R.M.; Sestini, P.; Bargagli, E. Systematic Review and Metanalysis of Oncomarkers in IPF Patients and Serial Changes of Oncomarkers in a Prospective Italian Real-Life Case Series. Cancers 2021, 13, 539. [Google Scholar] [CrossRef] [PubMed]
  3. d’Alessandro, M.; Bergantini, L.; Cameli, P.; Pieroni, M.; Refini, R.M.; Sestini, P.; Bargagli, E. Serum Concentrations of KL-6 in Patients with IPF and Lung Cancer and Serial Measurements of KL-6 in IPF Patients Treated with Antifibrotic Therapy. Cancers 2021, 13, 689. [Google Scholar] [CrossRef] [PubMed]
  4. Marjanski, T.; Wnuk, D.; Dziedzic, R.; Ostrowski, M.; Sawicka, W.; Marjanska, E.; Rzyman, W. Physiological Biomarkers Assessed by Low-Tech Exercise Tests Predict Complications and Overall Survival in Patients Undergoing Pneumonectomy Due to Lung Cancer. Cancers 2021, 13, 735. [Google Scholar] [CrossRef] [PubMed]
  5. Normanno, N.; Barberis, M.; De Marinis, F.; Gridelli, C.; On the Behalf of the Aiot Expert Panel. Molecular and Genomic Profiling of Lung Cancer in the Era of Precision Medicine: A Position Paper from the Italian Association of Thoracic Oncology (AIOT). Cancers 2020, 12, 1627. [Google Scholar] [CrossRef] [PubMed]
  6. De Luca, C.; Pepe, F.; Iaccarino, A.; Pisapia, P.; Righi, L.; Listì, A.; Greco, L.; Gragnano, G.; Campione, S.; De Dominicis, G.; et al. RNA-Based Assay for Next-Generation Sequencing of Clinically Relevant Gene Fusions in Non-Small Cell Lung Cancer. Cancers 2021, 13, 139. [Google Scholar] [CrossRef] [PubMed]
  7. Passaro, A.; Attili, I.; Rappa, A.; Vacirca, D.; Ranghiero, A.; Fumagalli, C.; Guarize, J.; Spaggiari, L.; de Marinis, F.; Barberis, M.; et al. Genomic Characterization of Concurrent Alterations in Non-Small Cell Lung Cancer (NSCLC) Harboring Actionable Mutations. Cancers 2021, 13, 2172. [Google Scholar] [CrossRef] [PubMed]
  8. Centonze, G.; Biganzoli, D.; Prinzi, N.; Pusceddu, S.; Mangogna, A.; Tamborini, E.; Perrone, F.; Busico, A.; Lagano, V.; Cattaneo, L.; et al. Beyond Traditional Morphological Characterization of Lung Neuroendocrine Neoplasms: In Silico Study of Next-Generation Sequencing Mutations Analysis across the Four World Health Organization Defined Groups. Cancers 2020, 12, 2753. [Google Scholar] [CrossRef] [PubMed]
  9. Fumagalli, C.; Guerini-Rocco, E.; Barberis, M. Making the Most of Complexity to Create Opportunities: Comprehensive Genomic Profiling and Molecular Tumor Board for Patients with Non-Small Cell Lung Cancer (NSCLC). Cancers 2021, 13, 609. [Google Scholar] [CrossRef] [PubMed]
  10. Janke, F.; Bozorgmehr, F.; Wrenger, S.; Dietz, S.; Heussel, C.P.; Heussel, G.; Silva, C.F.; Rheinheimer, S.; Feisst, M.; Thomas, M.; et al. Novel Liquid Biomarker Panels for A Very Early Response Capturing of NSCLC Therapies in Advanced Stages. Cancers 2020, 12, 954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Hofman, P. Next-Generation Sequencing with Liquid Biopsies from Treatment-Naïve Non-Small Cell Lung Carcinoma Patients. Cancers 2021, 13, 2049. [Google Scholar] [CrossRef] [PubMed]
  12. Pérez-Díez, I.; Hidalgo, M.R.; Malmierca-Merlo, P.; Andreu, Z.; Romera-Giner, S.; Farràs, R.; de la Iglesia-Vayá, M.; Provencio, M.; Romero, A.; García-García, F. Functional Signatures in Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis of Sex-Based Differences in Transcriptomic Studies. Cancers 2021, 13, 143. [Google Scholar] [CrossRef] [PubMed]
  13. Sesma, A.; Pardo, J.; Cruellas, M.; Gálvez, E.M.; Gascón, M.; Isla, D.; Martínez-Lostao, L.; Ocáriz, M.; Paño, J.R.; Quílez, E.; et al. From Tumor Mutational Burden to Blood T Cell Receptor: Looking for the Best Predictive Biomarker in Lung Cancer Treated with Immunotherapy. Cancers 2020, 12, 2974. [Google Scholar] [CrossRef] [PubMed]
  14. Carazo, F.; Bértolo, C.; Castilla, C.; Cendoya, X.; Campuzano, L.; Serrano, D.; Gimeno, M.; Planes, F.J.; Pio, R.; Montuenga, L.M.; et al. DrugSniper, a Tool to Exploit Loss-Of-Function Screens, Identifies CREBBP as a Predictive Biomarker of VOLASERTIB in Small Cell Lung Carcinoma (SCLC). Cancers 2020, 12, 1824. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fumagalli, C.; Barberis, M. Diagnostic and Predictive Biomarkers in Lung Cancer. Cancers 2021, 13, 2577. https://doi.org/10.3390/cancers13112577

AMA Style

Fumagalli C, Barberis M. Diagnostic and Predictive Biomarkers in Lung Cancer. Cancers. 2021; 13(11):2577. https://doi.org/10.3390/cancers13112577

Chicago/Turabian Style

Fumagalli, Caterina, and Massimo Barberis. 2021. "Diagnostic and Predictive Biomarkers in Lung Cancer" Cancers 13, no. 11: 2577. https://doi.org/10.3390/cancers13112577

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