Pathology in non-small cell lung cancer: evolving scenario
Review Article

Pathology in non-small cell lung cancer: evolving scenario

Pasquale Pisapia, Francesco Pepe, Antonino Iaccarino, Giancarlo Troncone, Umberto Malapelle

Department of Public Health, University of Naples “Federico II”, Naples 80131, Italy

Contributions: (I) Conception and design: P Pisapia, G Troncone, U Malapelle; (II) Administrative support: None; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: None; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Giancarlo Troncone. Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, Naples 80131, Italy. Email: giancarlo.troncone@unina.it.

Abstract: In 70–80% of advanced non small cell lung cancer (NSCLC) patients the only available material for both morphological diagnosis and molecular biomarkers assessment for the better treatment choice is represented by small tissue samples (small biopsies or cytological specimens). In order to avoid inadequate results due to the low amount of nucleic acids input and the difficulty to obtain tissue re-biopsies, the so called “liquid biopsy” could play a relevant role in this setting of patients. Tumoral DNA can be extracted from different body fluids [urine, saliva/sputum, cerebrospinal fluid (CSF), pleural effusion (PE)] of the patient or from supernatant sample of cytological preparations. In this review we analyzed the different the different “liquid biopsies” approaches.

Keywords: Non small cell lung cancer (NSCLC); liquid biopsy; next generation sequencing (NGS); epidermal growth factor receptor (EGRF); tyrosine kinase inhibitor (TKI)


Received: 13 May 2019; Accepted: 20 May 2019; Published: 05 June 2019.

doi: 10.21037/pcm.2019.05.04


Introduction

Lung cancer represents the most leading cause of mortality for cancer worldwide (1). To date, for non small cell lung cancer (NSCLC) patients there is a significant improvement in the target therapeutic strategies (2-8). For this reason, the College of American Pathologists (CAP), the International Association for the Study of Lung Cancer (IASLC), and the Association for Molecular Pathology (AMP), the National Comprehensive Cancer Network (NCCN) and the American Society of Clinical Oncology (ASCO) guidelines, defined a number of genes to necessarily test in advanced NSCLC patients, including epidermal growth factor receptor (EGRF), anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1 receptor tyrosine kinase (ROS1) and V-Raf murine sarcoma viral oncogene homolog B (BRAF) for treatment with tyrosine kinase inhibitors (TKIs) (9-11). In this evolving scenario, molecular predictive pathology plays a key role in the management of NSCLC patients (12). An important limitation in advanced NSCLC patients is represented by the low quantity of tissue specimens available for both morphological and molecular purposes (13). In particular, in a high percentage of these patients, the only material available to fulfill morphological and molecular requests is represented by small tissue biopsy and cytological samples, and in 30% of cases no tissue specimens are available (14-17). To overcome this limitation, a valid approach is the possibility to adopt liquid sources to assess the molecular status of these patients, the so called “liquid biopsies” (18-20). Among the different analytes that can be obtained from blood samples and the possibility to assess the molecular status of the different relevant biomarkers on other liquid specimens, to date the only approved analyte for TKI administration in NSCLC patients is represented by circulating tumor DNA (ctDNA) extracted from plasma to assess EGFR status (13,18). In particular, the analysis of ctDNA extracted from plasma is recommended in patients without tissue availability (insufficient tumor tissue or impossibility to obtain a tissue specimen) and to overcome problems relative to discomfort and risks of biopsies (13,18,21-26). An important limitation in this approach is represented by the low amount of ctDNA in the bloodstream (<0.5% of the total cell free DNA), that can be related to false negative results (13,14). Noteworthy, we demonstrated a high level of concordance between ctDNA and tissue results by using a next generation sequencing (NGS) approach with a small gene panel [SiRe®, Genedin (a spin-off of the Department of Public Health, University of Naples “Federico II”), Rome, Italy] that covers 568 clinical relevant mutations in 6 genes (EGFR, KRAS, NRAS, BRAF, PDGFRα and c-kit) (27-29). However, blood samples feature a number of analytes (e.g. circulating tumor cells, exosomes, platelet RNA and circulating tumor RNA), that showed an important role in NSCLC treatment decision making (30-33). In addition to blood samples, other fluids, that can be considered “liquid biopsies”, have the potential to give to molecular pathologists and clinicians the relevant information on the genetic assessment in NSCLC patients in order to administrate TKIs.

In this review, we focalized the attention on the advanced in the liquid biopsies approach, in particular paid attention on these other liquid sources different from blood samples.


Urine

The glomerular filtration represents a natural “centrifugation process” of plasma and allows, due to the low size of DNA fragments, the presence of tumor DNA in urine (20,34,35). A major advantage of urine samples is represented by the non-invasiveness in the collection than in blood samples (20). A major disadvantage is represented by the higher activity of both DNA and RNA hydrolyzing enzymes that contributed to degrade both analytes (36,37). Reckamp et al. firstly adopted in the TIGER-X trial, a phase 1/2 clinical study of a third generation TKI (rociletinib) in previously treated patients with a EGFR mutation in advanced NSCLC patients, urine sample to identify EGFR mutations (38). By using mutation enrichment PCR coupled with NGS detection, on n=60 urine samples with matched tissue specimens, the Authors showed a sensitivity of the EGFR exon 20 p.T790M, exon 21 p.L858R and exon 19 deletions of 72%, 75% and 67% when considering all urine volumes, and of 93%, 80% and 83%, respectively when a recommended urine volume of 90–100 mL was collected; the specificity was of 96%, 100% and 94% respectively (38). Of interest, the Authors emphasized the complementary role of urine and plasma with tissue. In particular, the authors identified in n=12 cases the EGFR exon 20 p.T790M (n=10 in urine and plasma samples, n=1 only in plasma and n=1 only in urine) previously undetectable or with inadequate results in tissue samples. Among these, n=9 showed a significant decrease of urine concentration of EGFR exon 20 p.T790M after 21 days from rociletinib administration (38). Li et al. analyzed n=160 urine, plasma and tissue samples from NSCLC patients, by digital droplet PCR (ddPCR), at various stages (39). On the overall, the Authors reported a high overall agreement between urine and tissue in particular in advanced stages (42% stages I/II vs. 93% stages III/IV) and between urine and plasma (75% stages I/II vs. 100% stages III/IV) (39). Hu et al. compared, by using ddPCR, urine samples with tissue specimens from n=213 NSCLC patients who underwent surgery (stages I–III) and with a known mutation for EGFR (n=111 EGFR exon 21 p.L858R and n=102 EGFR exon 19 deletions) (40). On the overall, only n=130 showed an EGFR positive result in urine samples (61%; 65.8% for EGFR exon 21 p.L858R and 55.9% for EGFR exon 19 deletions) (40).Interestingly, Authors demonstrated a reduction in urine of EGFR mutations in concordant cases after treatment (40). Zhang et al. performed ddPCR on n=160 urine samples, with matched tissue specimens, in advanced NSCLC patients (41). The authors showed an agreement on EGFR exon 21 p.L858R, EGFR exon 19 deletions and EGFR exon 20 p.T790M of 79%, 81% and 100% respectively (41). Considering all the EGFR mutations the positive predictive value (PPV) and negative predictive value (NPV) were 100% and 53.6% respectively (41). Wu et al. analyzed n=50 urine samples, in advanced (IIIB/IV) stage of disease with a NGS approach (42). The authors showed a concordance rate between tissue and urine of 70% (42). Wang et al. tested by a ddPRC approach n=200 urine and tissue samples from advanced stages NSCLC patients (43). The authors focused their attention on KRAS mutations, for prognostic purpose (43). On the overall, 78% (109/140) of cases showed KRAS mutation on both samples; no false positive results emerged in urine samples (n=60 negative cases for both samples) (43). The authors, also, emphasized that the identification and concentration of KRAS mutant DNA in urine is predictor of worse outcome (43). Similarly, Xie et al. analyzed n=150 (n=100 KRAS mutated cases and n=50 KRAS wild type cases) cases to investigate the potential role of urine analysis for prognostic purposes (44). Taking tissue results as gold standard, the overall agreement for the KRAS positive group was 77%; instead in the wild type group the concordance rate was 92% (44). The authors addressed an increasing number of KRAS positive cases in urine (before tissue positive and urine negative) when serial urine analysis was performed (93%) (44). In Table 1 are summarized results on urine samples.

Table 1

Studies focusing on the adoption of urine samples

First author N of urine samples (type of matched samples) Technology Concordance rate Sensitivity Specificity
Reckamp et al. (38) 60 (tissue) NGS p.T790M (93.0%), p.L858R (80.0%), del 19 (83.0%) p.T790M (96.0%), p.L858R (100.0%), del 19 (94.0%)
Li et al. (39) 160 (tissue) ddPCR p.L858R (97.0%)*, del 19 (89.0%)*, overall (93.0%)*
160 (plasma) p.L858R (100.0%)*, del 19 (100.0%)*, overall (100.0%)*
Hu et al. (40) 213 (tissue) ddPCR p.L858R (65.8%), del 19 (55.9%), overall (61.0%)
Zhang et al. (41) 160 (tissue) ddPCR p.L858R (79.0%), del 19 (81.0%), p.T790M (100.0%) 80.0% 100.0%
Wu et al. (42) 50 (tissue) NGS 70.0%
Wang et al. (43) 200 (tissue) ddPCR 70.0% 100.0%
Xie et al. (44) 150 (tissue) ddPCR 77.0%, 93.0%** 92.0%

*, advanced stages (III/IV); **, evaluation after serial urine analysis. ddPCR, digital droplet PCR; del, deletions; N, number; NGS, next generation sequencing.


Saliva

Saliva contains a wide range of different proteins, nucleic acids, electrolytes, and hormones that derived from different organs (45). For this reason, saliva can represent a source for biomarkers assessment as shown by Streckfus et al., who detected in this specimen c-erbB-2 in breast cancer patients (46). In the experience by Wu et al., in addition to urine samples the Authors analyzed n=50 saliva samples (42). Interestingly, the authors showed a concordance rate between tissue and saliva of 74% (42). Pu et al. analyzed n=17 saliva samples, collected from NSCLC patients before and after surgery, by using an electric field–induced release and measurement (EFIRM) technology (47). As gold standard, the Authors adopted the Food and Drug Administration (FDA) approved Cobas assay on matched tissue samples (47). Of interest, all cases harbored an EGFR exon 19 deletion or EGFR exon 21 p.L858R and all wild-type cases were confirmed in saliva samples (47). Only n=1 EGFR exon 18 p.G719X was not found in saliva by the EFIRM assay, due to the absence of the specific probes (47). The Authors evidenced only n=1 false positive result (EGFR exon 21 p.L858R) on plasma sample (47). Hubers et al. performed on n=10 EGFR tissue mutated cases and n=20 without EGFR mutation (n=10 lung cancer patients EGFR wild-type cases and n=10 chronic obstructive pulmonary disease (COPD) patients), at least four different tests [Cycleave PCR, co-amplification at lower denaturation temperature-PCR (COLD-PCR), Pangaea Biotech SL Technology (PST), and high resolution melting (HRM)] (48). No false positive results were evaluated in saliva samples; moreover a sensitivity of 50% was the higher evidenced (48). In Table 2 are summarized results on saliva samples.

Table 2

Studies focusing on the adoption of saliva samples

First author N of saliva samples (type of matched samples) Technology Concordance rate Sensitivity Specificity
Wu et al. (42) 50 (tissue) NGS 74.0%
Pu et al. (46) 17 (tissue) EFIRM 100.0%* 100.0%* 100.0%*
Hubers et al. (47) 10 (tissue) Cycleave PCR 30.0% 100.0%
COLD-PCR 40.0% 100.0%
PST 50.0% 100.0%
HRM 30.0% 100.0%

*, considering only EGFR exon 19 deletion and EGFR exon 21 p.L858R. COLD-PCR, co-amplification at lower denaturation temperature-PCR; del, deletion; EFIRM, electric field-induced release and measurement; EGFR, epidermal growth factor receptor; HRM, high resolution melting; N, number; PST, PangaeaBiotech SL Technology.


Cerebrospinal fluid (CSF)

The incidence of leptomeningeal metastases (LMs) in NSCLC patients is about 3% to 5% with an increasing in EGFR-mutated patients (49,50). Although lumbar puncture is an invasive procedure, CSF is a useful tool to obtain cell free DNA (cfDNA) in NSCLC patients with brain metastasis (20,51). Li et al. performed a NGS approach on n=26 CSF samples in EGFR mutated NSCLC patients with LM (52). In all cases the mutation was correctly evidenced in cfDNA extracted from CSF specimens (52). Additionally, mutations were found in matched precipitate of CFS and plasma samples (52). Interestingly, the detection rate of EGFR exon 20 p.T790M in CSF cfDNA was higher (7/23, 30.4%) than in plasma (5/23, 21.7%) (52). In another experience by the same group, Zheng et al. analyzed n=11 CSF and plasma samples in ALK rearrangement positive patients (53). On the overall, the concordance rate was higher (9/11, 81.8%) in CSF samples than in plasma (5/11, 45.5%) (53). Ying et al. performed a capture-based targeted sequencing on n=72 matched CSF and plasma samples (54). The authors showed a higher mutation detection rate in CSF than plasma when considering either any mutations (81.5% vs. 62.5%) or EGFR mutations (58.3% vs. 44.4%) (54). In a detailed analysis on EGFR mutated cases, the Authors evidenced that EGFR activating mutations were identified in 51.4% of CSF samples and in 38.9% of plasma samples (54). Zhao et al. focused the attention on n=7 NSCLC patients who failed EGFR TKI treatment (gefitinib) (55). In particular, different from matched plasma samples, the authors showed the persistence of EGFR mutated clones in all CSF samples respect to plasma (2/7, 28.6%) due to the low penetration of gefitinib of blood–brain barrier (55). Kawahara et al. evidenced a high sensitivity, specificity and accuracy (87.5%, 100%, 91.7% respectively) for the detection of EGFR mutations, including EGFR exon 20 p.T790M, by using cobas®EGFR Mutation Test v2 (56). Yang et al. performed amplification refractory mutation system (ARMS)-PCR assay on n=30 lung adenocarcinomas with brain metastasis (57). The authors showed a PPV, NPV, sensitivity and specificity between tissue and CSF samples of 75%, 75%, 67% and 82% respectively; the concordance rate in EGFR mutated cases was 75% (57). Similarly, Rong et al. adopted ARMS-PCR on n=5 CSF of NSCLC patients with EGFR sensitizing mutations (58). In only n=3 (60%) cases the mutation was confirmed (58). In Table 3 are summarized results on CSF samples.

Table 3

Studies focusing on the adoption of CSF samples

First author N of CSF samples (type of matched samples) Technology Concordance rate Sensitivity Specificity
Li et al. (52) 26 (tissue) NGS 100.0%
Zheng et al. (53) 11 (tissue) NGS 81.8%
Ying et al. (54) 72 (plasma) NGS 47.7%*, 32.7%**
Kawahara et al. (56) 12 (tissue) RT-PCR 87.5% 100%
Yang et al. (57) 30 (tissue) ARMS-PCR 75.0% 67.0% 82.0%
Rong et al. (58) 5 (tissue) ARMS-PCR 60.0%

*, considering EGFR activating mutations; **, considering all mutations. ARMS-PCR, amplification refractory mutation system-PCR; CSF, cerebrospinal fluid; N, number; NGS, next generation sequencing; RT-PCR, real time PCR.


Pleural effusion (PE)

Malignant pleural effusions (MPEs) are often observed in advanced NSCLC patients (59). Although thoracentesis is an invasive procedure, it is necessary for diagnostic, therapeutic and molecular purposes (60-62). Kimura et al. firstly reported the possibility to detect EGFR sensitizing mutation by analyzing DNA extracted from PE, and the correlation with response (partial response in this paper) with TKI treatment (63). Liu et al. performed the EGFR analysis on n=41 matching metastatic pleural tumor tissues (MPTTs), MPE supernatant and MPE cell blocks (CBs) by using ARMS-PCR (64). On the overall, confronting the MPTT and MPE supernatant results, the EGFR mutation detection sensitivity and specificity in MPE supernatants was 84.2% and 90.9%, respectively (64). A higher sensitivity (94.7%) but lower specificity (81.8%) was observed when considering both MPE samples (64). Lin et al. analyzed n=22 matched MPE supernatant, MPE cell pellet and tissue biopsy samples by using HRM (65). Considering tissue biopsy results as gold standard, no false positive results were evaluated among MPE supernatant samples (specificity 100%), whereas only n=1 false negative results was evidenced (sensitivity 92.3%) (65). A higher number of false negative results (4/22, 18.2%) was evidenced in MPE cell pellet samples (65). Similarly, Kawahara et al. analyzed n=74 (n=29 EGFR wild type and n=45 EGFR mutated cases) matched MPE supernatant, MPE cell pellet and tissue biopsy samples by using TaqMan Mutation Detection assay or fluorescence resonance energy transfer-based preferential homoduplex formation assay (F-PHFA) (66). The Authors showed a sensitivity and specificity on MPE supernatant of 44.4% and 100%, respectively (66). Lee et al. performed the EGFR analysis by using a peptide nucleic acid (PNA) mediated PCR clamping on two groups of lung adenocarcinoma (ADC) patients (67). In particular, the Authors analyzed cfDNA extracted from PE in previously genotyped TKI-naïve patients (n=32; n=19 EGFR mutated patients and n=13 wild type cases) and TKI-acquired resistance patients (n=18) (67). In the first group a concordance between the two samples of 88% was obtained; in particular in cfDNA extracted from PE n=2 out of 19 EGFR mutations were missed and in the wild type group n=2 additional mutations were found (67). In the second group only n=1 EGFR mutation was missed in cfDNA extracted from PE. Of interest, in n=11 (61%) cases in addition to the initial EGFR sensitizing mutation, an additional EGFR exon 20 p.T790M was found (67). On the overall, considering the EGFR sensitizing mutations the concordance rate in this second group was of 94% (67). In Table 4 are summarized results on PE samples.

Table 4

Studies focusing on the adoption of PE samples

First author N of PE samples (type of matched samples) Technology Concordance rate Sensitivity Specificity
Liu et al. (64) 41 (tissue) ARMS-PCR 76.2% 84.2% 90.9%
Lin et al. (65) 22 (tissue) HRM 92.3% 100.0%
Kawahara et al. (66) 74 (tissue) RT-PCR 44.4% 100.0%
Lee et al. (67) 32 (tissue) RT-PCR 88.0% 89.5% 84.6%
18 (tissue) 94.0%

ARMS-PCR, amplification refractory mutation system-PCR; HRM, high resolution melting; N, number; PE, pleural effusion; RT-PCR, real time PCR.


Future perspectives and conclusions

In addition to “body liquid biopsies”, another source of cfDNA for molecular purposes is represented by the supernatant after cytological preparations. This neglected material is normally discarded in cytological laboratories. However, several studies demonstrated the suitability of cfDNA extracted from supernatant obtained from cytological preparations for NGS analysis (68-74).

In conclusion, the so called “liquid biopsies” represent a valid material, not only in alternative to tissue based testing, to better define the molecular status of biomarkers in NSCLC patients, in order to define the best treatment choice (75,76).


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editors Alfredo Addeo and Giuseppe Banna for the series “Non-Small Cell Lung Cancer (NSCLC)” published in Precision Cancer Medicine. The article has undergone external peer review.

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/pcm.2019.05.04). The series “Non-Small Cell Lung Cancer (NSCLC)” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin 2012;62:10-29. [Crossref] [PubMed]
  2. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009;361:947-57. [Crossref] [PubMed]
  3. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2012;13:239-46. [Crossref] [PubMed]
  4. Yang JC, Wu YL, Schuler M, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 2015;16:141-51. [Crossref] [PubMed]
  5. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:113-25. [Crossref] [PubMed]
  6. Shaw AT, Kim DW, Nakagawa K, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 2013;368:2385-94. [Crossref] [PubMed]
  7. Peters S, Camidge DR, Shaw AT, et al. Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:829-38. [Crossref] [PubMed]
  8. Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med 2014;371:1963-71. [Crossref] [PubMed]
  9. Lindeman NI, Cagle PT, Aisner DL, et al. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch Pathol Lab Med 2018;142:321-46. [Crossref] [PubMed]
  10. Kalemkerian GP, Narula N, Kennedy EB, et al. Molecular Testing Guideline for the Selection of Patients With Lung Cancer for Treatment With Targeted Tyrosine Kinase Inhibitors: American Society of Clinical Oncology Endorsement of the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology Clinical Practice Guideline Update. J Clin Oncol 2018;36:911-9. [Crossref] [PubMed]
  11. Ettinger DS, Aisner DL, Wood DE, et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 5.2018. J Natl Compr Canc Netw 2018;16:807-821. [Crossref] [PubMed]
  12. Walk EE. The role of pathologists in the era of personalized medicine. Arch Pathol Lab Med 2009;133:605-10. [PubMed]
  13. Pisapia P, Malapelle U, Troncone G. Liquid Biopsy and Lung Cancer. Acta Cytol 2018; [Epub ahead of print]. [Crossref] [PubMed]
  14. Malapelle U, Pisapia P, Rocco D, et al. Next generation sequencing techniques in liquid biopsy: focus on non-small cell lung cancer patients. Transl Lung Cancer Res 2016;5:505-10. [Crossref] [PubMed]
  15. Ofiara LM, Navasakulpong A, Ezer N, et al. The importance of a satisfactory biopsy for the diagnosis of lung cancer in the era of personalized treatment. Curr Oncol 2012;19:S16-23. [Crossref] [PubMed]
  16. Wang S, Yu B, Ng CC, et al. The suitability of small biopsy and cytology specimens for EGFR and other mutation testing in non-small cell lung cancer. Transl Lung Cancer Res 2015;4:119-25. [PubMed]
  17. Malapelle U, Bellevicine C, De Luca C, et al. EGFR mutations detected on cytology samples by a centralized laboratory reliably predict response to gefitinib in non-small cell lung carcinoma patients. Cancer Cytopathol 2013;121:552-60. [Crossref] [PubMed]
  18. Rolfo C, Mack PC, Scagliotti GV, et al. Liquid Biopsy for Advanced Non-Small Cell Lung Cancer (NSCLC): A Statement Paper from the IASLC. J Thorac Oncol 2018;13:1248-68. [Crossref] [PubMed]
  19. Crowley E, Di Nicolantonio F, Loupakis F, et al. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 2013;10:472-84. [Crossref] [PubMed]
  20. Siravegna G, Marsoni S, Siena S, et al. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 2017;14:531-48. [Crossref] [PubMed]
  21. Sacher AG, Komatsubara KM, Oxnard GR. Application of Plasma Genotyping Technologies in Non-Small Cell Lung Cancer: A Practical Review. J Thorac Oncol 2017;12:1344-56. [Crossref] [PubMed]
  22. Chouaid C, Dujon C, Do P, et al. Feasibility and clinical impact of re-biopsy in advanced non small-cell lung cancer: a prospective multicenter study in a real-world setting (GFPC study 12-01). Lung Cancer 2014;86:170-3. [Crossref] [PubMed]
  23. Heerink WJ, de Bock GH, de Jonge GJ, et al. Complication rates of CT-guided trans thoracic lung biopsy: meta-analysis. Eur Radiol 2017;27:138-48. [Crossref] [PubMed]
  24. Sacher AG, Paweletz C, Dahlberg SE, et al. Prospective Validation of Rapid Plasma Genotyping for the Detection of EGFR and KRAS Mutations in Advanced Lung Cancer. JAMA Oncol 2016;2:1014-22. [Crossref] [PubMed]
  25. Cai W, Lin D, Wu C, et al. Intratumoral Heterogeneity of ALK-Rearranged and ALK/EGFR Co altered Lung Adenocarcinoma. J Clin Oncol 2015;33:3701-9. [Crossref] [PubMed]
  26. Jamal-Hanjani M, Wilson GA, McGranahan N, et al. Tracking the Evolution ofNon-Small-Cell Lung Cancer. N Engl J Med 2017;376:2109-21. [Crossref] [PubMed]
  27. Malapelle U, Mayo de-Las-Casas C, Rocco D, et al. Development of a gene panel for next-generation sequencing of clinically relevant mutations in cell-free DNA from cancer patients. Br J Cancer 2017;116:802-10. [Crossref] [PubMed]
  28. Pisapia P, Pepe F, Smeraglio R, et al. Cell free DNA analysis by SiRe(®) next generation sequencing panel in non small cell lung cancer patients: focus on basal setting. J Thorac Dis 2017;9:S1383-90. [Crossref] [PubMed]
  29. Pisapia P, Rocco D, Pepe F, et al. EGFR exon 19 deletion switch and development of p.L792Q mutation as a new resistance mechanism to osimertinib: a case report and literature review. Transl Cancer Res 2019;8:S64-9. [Crossref]
  30. Joosse SA, Pantel K. Tumor-Educated Platelets as Liquid Biopsy in Cancer Patients. Cancer Cell 2015;28:552-4. [Crossref] [PubMed]
  31. Raez LE, Manca P, Rolfo C, et al. ROS-1 Rearrangements in Circulating Tumor Cells. J Thorac Oncol 2018;13:e71-2. [Crossref] [PubMed]
  32. Reclusa P, Taverna S, Pucci M, et al. Exosomes as diagnostic and predictive biomarkers in lung cancer. J Thorac Dis 2017;9:S1373-82. [Crossref] [PubMed]
  33. Reclusa P, Laes JF, Malapelle U, et al. EML4-ALK translocation identification in RNA exosomal cargo (ExoALK) in NSCLC patients: a novel role for liquid biopsy. Transl Cancer Res 2019;8:S76-8. [Crossref]
  34. Su YH, Wang M, Brenner DE, et al. Human urine contains small, 150 to 250 nucleotide-sized, soluble DNA derived from the circulation and may be useful in the detection of colorectal cancer. J Mol Diagn 2004;6:101-7. [Crossref] [PubMed]
  35. Su YH, Wang M, Aiamkitsumrit B, et al. Detection of a K-ras mutation in urine of patients with colorectal cancer. Cancer Biomark 2005;1:177-82. [Crossref] [PubMed]
  36. Nadano D, Yasuda T, Kishi K. Measurement of deoxyribonuclease I activity inhuman tissues and body fluids by a single radial enzyme-diffusion method. Clin Chem 1993;39:448-52. [PubMed]
  37. Mall C, Rocke DM, Durbin-Johnson B, et al. Stability of miRNA in human urine supports its biomarker potential. Biomark Med 2013;7:623-31. [Crossref] [PubMed]
  38. Reckamp KL, Melnikova VO, Karlovich C, et al. A Highly Sensitive and Quantitative Test Platform for Detection of NSCLC EGFR Mutations in Urine and Plasma. J Thorac Oncol 2016;11:1690-700. [Crossref] [PubMed]
  39. Li F, Huang J, Ji D, et al. Utility of urinary circulating tumor DNA for EGFR mutation detection in different stages of non-small cell lung cancer patients. Clin Transl Oncol 2017;19:1283-91. [Crossref] [PubMed]
  40. Hu T, Shen H, Huang H, et al. Urinary circulating DNA profiling in non-small cell lung cancer patients following treatment shows prognostic potential. J Thorac Dis 2018;10:4137-46. [Crossref] [PubMed]
  41. Zhang H, He B, Cui J, et al. Comparison of circulating DNA from plasma and urine for EGFR mutations in NSCLC patients. Cancer Biomark 2018;23:427-36. [Crossref] [PubMed]
  42. Wu Z, Yang Z, Li CS, et al. Differences in the genomic profiles of cell-free DNA between plasma, sputum, urine, and tumor tissue in advanced NSCLC. Cancer Med 2019;8:910-9. [Crossref] [PubMed]
  43. Wang X, Meng Q, Wang C, et al. Investigation of transrenal KRAS mutation in late stage NSCLC patients correlates to disease progression. Biomarkers 2017;22:654-60. [PubMed]
  44. Xie F, Li P, Gong J, et al. Urinary cell-free DNA as a prognostic marker for KRAS-positive advanced-stage NSCLC. Clin Transl Oncol 2018;20:591-8. [Crossref] [PubMed]
  45. Gao K, Zhou H, Zhang L, et al. Systemic disease-induced salivary biomarker profiles in mouse models of melanoma and non-small cell lung cancer. PLoS One 2009;4:e5875. [Crossref] [PubMed]
  46. Streckfus C, Bigler L, Dellinger T, et al. The presence of soluble c-erbB-2 in saliva and serum among women with breast carcinoma: a preliminary study. Clin Cancer Res 2000;6:2363-70. [PubMed]
  47. Pu D, Liang H, Wei F, et al. Evaluation of a novel saliva-based epidermal growth factor receptor mutation detection for lung cancer: A pilot study. Thorac Cancer 2016;7:428-36. [Crossref] [PubMed]
  48. Hubers AJ, Heideman DA, Yatabe Y, et al. EGFR mutation analysis in sputum of lung cancer patients: a multi technique study. Lung Cancer 2013;82:38-43. [Crossref] [PubMed]
  49. Remon J, Le Rhun E, Besse B. Leptomeningeal carcinomatosis in non-small cell lung cancer patients: A continuing challenge in the personalized treatment era. Cancer Treat Rev 2017;53:128-37. [Crossref] [PubMed]
  50. Li YS, Jiang BY, Yang JJ, et al. Leptomeningeal Metastases in Patients with NSCLC with EGFR Mutations. J Thorac Oncol 2016;11:1962-9. [Crossref] [PubMed]
  51. Pan W, Gu W, Nagpal S, et al. Brain tumor mutations detected in cerebral spinal fluid. Clin Chem 2015;61:514-22. [Crossref] [PubMed]
  52. Li YS, Jiang BY, Yang JJ, et al. Unique genetic profiles from cerebrospinal fluid cell-free DNA in leptomeningeal metastases of EGFR-mutant non-small-cell lung cancer: a new medium of liquid biopsy. Ann Oncol. 2018;29:945-52. [Crossref] [PubMed]
  53. Zheng MM, Li YS, Jiang BY, et al. Clinical Utility of Cerebrospinal Fluid Cell-Free DNA as Liquid Biopsy for Leptomeningeal Metastases in ALK-Rearranged NSCLC. J Thorac Oncol 2019; [Epub ahead of print]. [Crossref] [PubMed]
  54. Ying S, Ke H, Ding Y, et al. Unique genomic profiles obtained from cerebrospinal fluid cell-free DNA of non-small cell lung cancer patients with leptomeningeal metastases. Cancer Biol Ther 2019;20:562-70. [Crossref] [PubMed]
  55. Zhao J, Ye X, Xu Y, et al. EGFR mutation status of paired cerebrospinal fluid and plasma samples in EGFR mutant non-small cell lung cancer with leptomeningeal metastases. Cancer Chemother Pharmacol 2016;78:1305-10. [Crossref] [PubMed]
  56. Kawahara A, Abe H, Murata K, et al. Screening system for epidermal growth factor receptor mutation detection in cytology cell-free DNA of cerebrospinal fluid based on assured sample quality. Cytopathology 2019;30:144-9. [Crossref] [PubMed]
  57. Yang H, Cai L, Zhang Y, et al. Sensitive detection of EGFR mutations in cerebrospinal fluid from lung adenocarcinoma patients with brain metastases. J Mol Diagn 2014;16:558-63. [Crossref] [PubMed]
  58. Rong J, Chunhua M, Yuan L, et al. Detected EGFR mutation in cerebrospinal fluid of lung adenocarcinoma patients with meningeal metastasis. Open Med (Wars) 2016;11:93-6. [Crossref] [PubMed]
  59. Antony VB, Loddenkemper R, Astoul P, et al. Management of malignant pleural effusions. Eur Respir J 2001;18:402-19. [Crossref] [PubMed]
  60. Dai Y, Morishita Y, Mase K, et al. Application of the p53 and K-ras gene mutation patterns for cytologic diagnosis of recurrent lung carcinomas. Cancer 2000;90:258-63. [Crossref] [PubMed]
  61. Huang MJ, Lim KH, Tzen CY, et al. EGFR mutations in malignant pleural effusion of non-small cell lung cancer: a case report. Lung Cancer 2005;49:413-5. [Crossref] [PubMed]
  62. Soh J, Toyooka S, Aoe K, et al. Usefulness of EGFR mutation screening in pleural fluid to predict the clinical outcome of gefitinib treated patients with lung cancer. Int J Cancer 2006;119:2353-8. [Crossref] [PubMed]
  63. Kimura H, Fujiwara Y, Sone T, et al. EGFR mutation status in tumour-derived DNA from pleural effusion fluid is a practical basis for predicting the response to gefitinib. Br J Cancer 2006;95:1390-5. [Crossref] [PubMed]
  64. Liu D, Lu Y, Hu Z, et al. Malignant pleural effusion supernatants are substitutes for metastatic pleural tumor tissues in EGFR mutation test in patients with advanced lung adenocarcinoma. PLoS One 2014;9:e89946. [Crossref] [PubMed]
  65. Lin J, Gu Y, Du R, et al. Detection of EGFR mutation in supernatant, cell pellets of pleural effusion and tumor tissues from non-small cell lung cancer patients by high resolution melting analysis and sequencing. Int J Clin Exp Pathol 2014;7:8813-22. [PubMed]
  66. Kawahara A, Fukumitsu C, Azuma K, et al. A Combined test using both cell sediment and supernatant cell-free DNA in pleural effusion shows increased sensitivity in detecting activating EGFR mutation in lung cancer patients. Cytopathology 2018;29:150-5. [Crossref] [PubMed]
  67. Lee JS, Hur JY, Kim IA, et al. Liquid biopsy using the supernatant of a pleural effusion for EGFR genotyping in pulmonary adenocarcinoma patients: a comparison between cell-free DNA and extracellular vesicle-derived DNA. BMC Cancer 2018;18:1236. [Crossref] [PubMed]
  68. Tian SK, Killian JK, Rekhtman N, et al. Optimizing Workflows and Processing of Cytologic Samples for Comprehensive Analysis by Next-Generation Sequencing: Memorial Sloan Kettering Cancer Center Experience. Arch Pathol Lab Med 2016; [Epub ahead of print]. [Crossref] [PubMed]
  69. Roy-Chowdhuri S. Molecular testing of residual cytology samples: Rethink, reclaim, repurpose. Cancer Cytopathol 2019;127:15-7. [Crossref] [PubMed]
  70. Rekhtman N, Roy-Chowdhuri S. Cytology Specimens: A Goldmine for Molecular Testing. Arch Pathol Lab Med 2016;140:1189-90. [Crossref] [PubMed]
  71. Roy-Chowdhuri S, Mehrotra M, Bolivar AM, et al. Salvaging the supernatant: next generation cytopathology for solid tumor mutation profiling. Mod Pathol 2018;31:1036-45. [Crossref] [PubMed]
  72. Guibert N, Tsukada H, Hwang DH, et al. Liquid biopsy of fine-needle aspiration supernatant for lung cancer genotyping. Lung Cancer 2018;122:72-5. [Crossref] [PubMed]
  73. Hannigan B, Ye W, Mehrotra M, et al. Liquid Biopsy Assay for Lung Carcinoma Using Centrifuged Supernatants from Fine Needle Aspiration Specimens. Ann Oncol 2019; [Epub ahead of print]. [Crossref] [PubMed]
  74. Ye W, Hannigan B, Zalles S, et al. Centrifuged supernatants from FNA provide a liquid biopsy option for clinical next-generation sequencing of thyroid nodules. Cancer Cytopathol 2019;127:146-60. [Crossref] [PubMed]
  75. Liquid Biopsy Holds Its Own in NSCLC. Cancer Discov 2019; [Epub ahead of print].
  76. Leighl NB, Page RD, Raymond VM, et al. Clinical Utility of Comprehensive Cell-Free DNA Analysis to Identify Genomic Biomarkers in Patients with Newly Diagnosed Metastatic Non-Small Cell Lung Cancer. Clin Cancer Res 2019; [Epub ahead of print]. [Crossref] [PubMed]
doi: 10.21037/pcm.2019.05.04
Cite this article as: Pisapia P, Pepe F, Iaccarino A, Troncone G, Malapelle U. Pathology in non-small cell lung cancer: evolving scenario. Precis Cancer Med 2019;2:17.

Download Citation