Next Article in Journal
Influence of Obesity and Fluid Balance on Operative Outcomes in Hepatic Resection
Previous Article in Journal
A Single Center Initial Experience with Robotic-Assisted Minimally Invasive Coronary Artery Bypass Surgery (RA-MIDCAB)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Preliminary Experience of Liquid Biopsy in Lung Cancer Compared to Conventional Assessment: Light and Shadows

by
Marco Montella
1,
Giovanni Ciani
2,
Vincenza Granata
3,
Roberta Fusco
4,
Francesca Grassi
2,*,
Andrea Ronchi
1,
Immacolata Cozzolino
1,
Renato Franco
1,
Federica Zito Marino
1,
Fabrizio Urraro
2,
Riccardo Monti
2,
Roberto Sirica
5,
Giovanni Savarese
5,
Ugo Chianese
2,
Angela Nebbioso
2,
Lucia Altucci
2,
Maria Teresa Vietri
2,
Valerio Nardone
2,
Alfonso Reginelli
2 and
Roberta Grassi
2,6
1
Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
2
Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
3
Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131 Naples, Italy
4
Medical Oncology Division, Igea SpA, 80013 Napoli, Italy
5
AMES-Centro Polidiagnostico Strumentale, SRL, 80013 Naples, Italy
6
Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, 20122 Milan, Italy
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2022, 12(11), 1896; https://doi.org/10.3390/jpm12111896
Submission received: 31 August 2022 / Revised: 8 November 2022 / Accepted: 10 November 2022 / Published: 12 November 2022
(This article belongs to the Section Methodology, Drug and Device Discovery)

Abstract

:
Purpose: To assess the qualitative relationship between liquid biopsy and conventional tissue biopsy. As a secondary target, we evaluated the relationship between the liquid biopsy results and the T stage, N stage, M stage, and compared to grading. Methods: The Local Ethics Committee of the “Università degli Studi della Campania Luigi Vanvitelli”, with the internal resolution number 24997/2020 of 12.11.2020, approved this spontaneous prospective study. According to the approved protocol, patients with lung cancer who underwent Fine-Needle Aspiration Cytology (FNAC), CT-guided biopsy, and liquid biopsy were enrolled. A Yates chi-square test was employed to analyze differences in percentage values of categorical variables. A p-value < 0.05 was considered statistically significant. Data analysis was performed using the Matlab Statistic Toolbox (The MathWorks, Inc., Natick, MA, USA). Results: When a genetic mutation is present on the pathological examination, this was also detected on the liquid biopsy. ROS1 and PDL1 mutations were found in 2/29 patients, while EGFR Exon 21 was identified in a single patient. At liquid biopsy, 26 mutations were identified in the analyzed samples. The mutations with the highest prevalence rate in the study populations were: ALK (Ile1461Val), found in 28/29 patients (96.6%), EML4 (Lys398Arg), identified in 16/29 (55.2%) patients, ALK (Asp1529Glu), found in 14/29 (48.3%) patients, EGFR (Arg521Lys), found in 12/29 (41.4%) patients, ROS (Lys2228Gln), identified in 11/29 (37.9%) patients, ROS (Arg167Gln) and ROS (Ser2229Cys), identified in 10/29 (34.5%) patients, ALK (Lys1491Arg) and PIK3CA (Ile391Met), identified in 8/29 (27.6%) patients, ROS (Thr145Pro), identified in 6/29 (20.7%) patients, and ROS (Ser1109Leu), identified in 4/29 (13.8%) patients. No statistically significant differences can be observed in the mutation rate between the adenocarcinoma population and the squamous carcinoma population (p > 0.05, Yates chi-square test). Conclusions: We showed that, when a genetic mutation was detected in pathological examination, this was always detected by liquid biopsy, demonstrating a very high concordance rate of genomic testing between tissues and their corresponding mutations obtained by liquid biopsy, without cases of false-negative results. In addition, in our study, liquid biopsy highlighted 26 mutations, with the prevalence of ALK mutation in 96.6% of patients, supporting the idea that this approach could be an effective tool in cases with insufficient tumor tissue specimens or in cases where tissue specimens are not obtainable.

1. Introduction

The assessment of tumor genetic alterations is currently crucial in oncological patient management and treatment decisions [1,2,3,4,5,6,7,8,9,10]. In fact, in the era of personalized medicine, an accurate genetic tumor assessment allows to identify the more precise treatment, to define the treatment time, and to standardize the follow-up time [11,12,13,14,15,16,17,18,19,20,21]. Usually, evaluation of tumor mutational status is performed by means of a section of the primary or secondary lesion [22,23,24,25]. However, this approach shows several weaknesses, such as the invasive nature of the procedure, the inaccessibility when the lesion is in a deep site, and the risk of an inadequate sample or a partial assessment, considering intra-tumor heterogeneity, especially spatial heterogeneity, when a single biopsy is tested [26]. Moreover, this approach does not allow a dynamic follow-up of cancer molecular modifications that could occur during treatment [26]. In addition, in oncological patients, a critical point is related to the possibility of an early cancer diagnosis. It is known that patients have a higher cure rate and five-year survival if diagnosed at early stages [2,27,28,29,30,31]. To perform large-scale tumor screening among healthy people and to obtain an accurate mutational cancer status, several researchers and companies have turned their attention toward liquid biopsy [31,32,33,34,35,36,37]. Blood includes several categories of biological substances, such as circulating cells, extracellular vesicles, platelets, protein, mRNA, miRNA, and cell-free DNA (cfDNA) [36]. In oncological patients, a cfDNA portion, known as circulating tumor DNA (ctDNA), is liberated by lesions due to active release, necrosis, and apoptosis [37]. The alterations in ctDNA can be considered as a biomarker, and these can allow to identify cancer patients from a group of healthy individuals, to evaluate the cancer mutational status, to assess the response to treatment, and to identify groups of patients with a poor prognosis. This approach could manage the highest level of personalized medicine [38,39,40,41,42,43,44,45,46,47,48,49]. Compared to conventional tissue biopsy, liquid biopsy is more feasible and less invasive and is more comprehensive to assess tumor heterogeneity since all tumor parts release ctDNA [38].
Worldwide, lung cancer is one of the leading causes of morbidity and mortality among oncological patients [50,51,52,53]. Despite the progress in the treatment, also thanks to the benefits from immunotherapy, there is a necessity to identify new robust biomarkers that could predict response, resistance, and/or toxicity to treatment [54,55,56,57]. Up to now, the European Medicines Agency (EMA) [58] and the FDA [59] have approved epidermal growth factor receptor (EGFR) mutation testing using ctDNA in non-small-cell lung cancer (NSCLC) patients. For patients treated with immune checkpoint inhibitors for NSCLC, several studies showed that ctDNA could be an early marker of therapeutic efficacy and it could better predict survival outcomes [60].
ctDNA is highly fragmented, ranging from 100 to 10,000 bp. It is challenging to isolate ctDNA from the blood for quantitation since the small fragments are easy to lose or degrade [26]. Although the concentration of ctDNA should increase with the stage and tumor size, the total percentage of ctDNA in the blood is extremely low, putting many requirements on the sample processing procedure [26]. Thus, ctDNA assays used for early cancer diagnosis should be highly sensitive. However, highly sensitive assays are always expensive, making large-scale practical applications unrealistic. For late-stage cancer tumor typing, the sensitivity can be moderate because the concentration of ctDNA is much larger [26]. Additionally, it has been shown that both the concentration and stability of ctDNA could be influenced by the form, release, degradation, and clearance of cfDNA [26]. Until now, very few studies have discussed the clearance rate and biological mechanism of ctDNA. Another significant obstacle at present is the lack of biological knowledge and experimental evidence to support the quantitative relationship between ctDNA and early cancer development [26].
Another critical issue is the lack of biological knowledge and experimental evidence to support the qualitative and quantitative relationship between ctDNA and conventional pathological tissue.
This research is a part of an ongoing study on liquid biopsy in lung cancer. We assessed as a primary endpoint the qualitative relationship between ctDNA and conventional cytological sampling biopsy. As a secondary target, we evaluated the relationship between the liquid biopsy results and the T stage, N stage, M stage, and grading.

2. Methods

2.1. Patient Selection

The Local Ethics Committee of the “Università degli Studi della Campania Luigi Vanvitelli”, with internal resolution No. 24997/2020 of 12.11.2020, approved this prospective study.
According to the approved protocol, patients with histologically confirmed lung cancer who underwent tissue CT-guided biopsy and liquid biopsy were enrolled.
The study was performed in accordance with relevant guidelines and regulations.

2.2. Subjects’ Selection

The eligibility of the patients has been assessed by the investigators. Inclusion criteria included: (a) age ≥ 18 years, (b) suitable mental health conditions, (c) ability to sign a specific informed consent form, (d) diagnosis of NSCLC with histological confirmation, (e) computed tomography staging, and (f) blood sample to perform liquid biopsy.
Exclusion criteria included: (a) age less than 18 years, (b) pregnancy, (c) absolute contraindication to CT study (previous adverse reactions to contrast medium), (d) inability to sign a specific informed consent form, (e) refusal to provide a blood sample for liquid biopsy, and (f) absence of conventional biopsy.
Having to compare the proportions of genetic mutations between two groups, we used the following statistical inference model: inference for two proportions. To obtain a statistical power of 80% and a statistical significance of 0.1, and an error of the second type of 0.2, starting from a difference in incidence between the proportions of genetic mutations between the two groups of 10%, it will be necessary to include at least 116 cases (58 per group).
In this study, we report the preliminary results of the first 29 enrolled patients.

3. Sampling Protocol

The study included patients with a newly detected pulmonary nodule, who had been scheduled to undergo transthoracic CT-guided Fine-Needle Aspiration Cytology (FNAC) and a final cytological diagnosis of NSCLC. The patients were thoroughly informed of risks, complications, and the possible inadequacy of the rate of the FNAC procedures and gave their written informed consent to undergo the procedure.
A 23-G, 150 mm needle connected to a syringe mounted on the holder was used in all cases.
Thereafter, a radiologist, with the collaboration of a cytopathologist, performed a CT-guided percutaneous transthoracic FNAC using a Revolution Discovery 64-slice CT scanner (General Electric, Boston, MA, USA). ROSE (Rapid On-Site Evaluation) was always performed to provide indications on the number of passes and the choice of vials in CT-guided cases. According to our protocol [13], a total of 1 to 4 passes were performed for each patient. A total of 1 to 2 smears were air-dried and DiffQuik-stained for ROSE, and additional smears were alcohol-fixed and Papanicolaou-stained. Residual material in the hub of the needle from the first pass, as well as material from the additional passes, was suspended in formalin for cell block (CB) preparation.
All the patients were clinically monitored in the recovery room, and 2 h after the procedure, a chest radiography was performed to diagnose possible complications.

4. Morphophenotypical Assessment

Cytological diagnoses were matched with the corresponding histological diagnoses, when available, and all diagnoses were conducted according to the last WHO classification of tumors criteria (WHO 2021). All cytological and histological slides were evaluated by two experienced pathologists with specific expertise in cytology.
Immunocytochemistry (ICC) was carried out on CB sections for diagnostic purposes to classify pulmonary non-small-cell carcinomas. ICC was performed on the Ventana platform (VENTANA BenchMark Ultra system; Ventana, Tucson, Arizona) and included the following antibodies: TTF1 (thyroid transcription factor 1), p63, Napsin, p40, cytokeratin 7 (CK7), PD-L1 (programmed death ligand-1), ALK (anaplastic lymphoma kinase), and ROS1. The ICC markers used were TTF1, Napsin, and CK7 for adenocarcinoma, and p63, p40, and CK 5/6 for squamous cell carcinoma.

5. Molecular and Predictive Tests

The smears were used for the extraction of DNA for next-generation sequencing, whereas CB sections were used for ICC (ALK, ROS1, and PD-L1). Next-generation sequencing was performed to analyze epidermal growth factor receptor (EGFR) and BRAF mutational status in all cases of lung adenocarcinoma. In all cases, smears were used for the extraction of DNA by the Qiamp DNA Micro Kit (Qiagen, Hilden, Germany), according to the manufacturer’s instructions. Extracted DNA was eluted in 20 μL of elution buffer and subsequently quantified by a Qubit 2.0 Fluorometer (Life Technologies, Carlsbad, CA, USA) using the Qubit dsDNA HS Assay kit, according to the manufacturer’s recommendations. The extracted DNA was stored at −20 °C.

6. Liquid Biopsy Analysis

-
Blood Samples and ctDNA Extraction
Here, 5 mL of blood was collected by ethylenediaminetetraacetic acid (EDTA) blood collection tubes, the same day as CT-guided Fine-Needle Aspiration Cytology.
To remove blood cells, blood was centrifuged at 1800× g for 10 min at 4 °C. Then, the supernatant was centrifuged at 16,000× g for 10 min at 4 °C to remove any remaining cells. Circulating tumor DNA was extracted from 2 mL of plasma, by digestion in 100 μL of proteinase K buffer for 10 min at 37 °C, followed by purification with the Plasma XS kit with the given protocol. The purified ctDNA was quantified by a Picogreen fluorescence assay using the provided lambada DNA standards.
-
ctDNA Sequencing and Analysis
The 5-biotinylated probe solution was provided as a capture probe, and the baits target cancer-related genes. Hybridization, target amplification, barcode library preparation, and size selection were performed according to the manufacturer’s protocols. Libraries were prepared with the TruSigt Oncology 500 ctDNA kit, based on target enrichment that analyzes 523 cancer-relative genes. The assay detected several classes of genetic mutations, such as small nucleotide variants (SNVs), indels, splice variants, and biomarkers such as tumor mutational burden (TMB) and microsatellite instability (MSI). Sequencing was performed on an Illumina NovaSeq 6000 (San Diego, CA, USA) platform. The analysis was performed with the TruSight Oncology local app on a Dragen server.

Statistical Analysis

A Yates chi-square test was employed to analyze differences in percentage values of categorical variables.
A p-value < 0.05 was considered statistically significant.
Data analysis was performed using the MATLAB Statistical Toolbox (The MathWorks, Inc., Natick, MA, USA).

7. Results

We assessed 29 patients with NSCLC. In 21/29 patients, the lesion was adenocarcinoma, while 8/29 patients had squamous carcinoma. Tumor lesions were localized in the peripheral area in 86.2% (25/29 patients), while 4/29 (13.8%) lesions were localized in the central area.
Figure 1 describes the distribution of the location of tumors and the type of carcinoma.
Table 1 reports the TNM and stage distribution in our population.
All the genetic mutations, detected by pathological analysis, were found by liquid biopsy. There were no false negatives at liquid biopsy compared to pathological analysis.
Table 2 reports the mutations found at pathological analysis: ROS1 and PDL1 mutations were found in 2/29 (at immunocytochemical analysis) patients, while EGFR Exon 21 was identified in a single patient (at RT-PCR analysis). Mutations were observed in the adenocarcinoma group.
At liquid biopsy, 26 mutations were identified in the analyzed samples. Figure 2 reports the mutation status at liquid biopsy for a single patient.
The mutations with the highest prevalence rate in the study populations were:
-
ALK (Ile1461Val), found in 28/29 (96.6%) patients;
-
EML4 (Lys398Arg), identified in 16/29 (55.2%) patients;
-
ALK (Asp1529Glu), found in 14/29 (48.3%) patients;
-
EGFR (Arg521Lys), found in 12/29 (41.4%) patients;
-
ROS (Lys2228Gln), identified in 11/29 (37.9%) patients;
-
ROS (Arg167Gln) and ROS (Ser2229Cys), identified in 10/29 (34.5%) patients;
-
ALK (Lys1491Arg) and PIK3CA (Ile391Met), identified in 8/29 (27.6%) patients;
-
ROS (Thr145Pro), identified in 6/29 (20.7%) patients;
-
ROS (Ser1109Leu), identified in 4/29 (13.8%) patients;
-
KRAS (Gly12Cys) was found in 2/29 (6.9%) patients; and
-
EGFR (Arg255Gln), EGFR (Val592Ile), EGFR (Ala647Thr), EGFR (Leu858Arg), ALK (Pro254Thr), ALK (Trp288Ser), ALK (Glu797Lys), PIK3CA (Arg19Ile), PIK3CA (Arg852Pro), PIK3CA (Met1043Ile), ROS (Glu1902Lys), ROS (Leu567Val), ROS (Phe1153Leu), and ROS (Trp847Leu) were identified only in one patient.
Table 3 reports the mutation status at liquid biopsy compared, respectively, to T, N, M, and grading.
No statistically significant differences were observed among the mutation status and the disease status (p > 0.05, Yates chi-square test).
Figure 3 reports the number of mutated patients compared between the adenocarcinoma population and the squamous carcinoma population. No statistically significant differences were observed in the number of each mutation between the two populations (p > 0.05, Yates chi-square test, Table 3).
EGFR (Arg255Gln), EGFR (Ala647Thr), EGFR (Leu858Arg), KRAS (Gly12Cys), ALK (Pro254Thr), ALK (Trp288Ser), ALK (Glu797Lys), ROS (Glu1902Lys), PIK3CA (Met1043Ile), ROS (Leu567Val), and ROS (Phe1153Leu) mutations were present only in patients with adenocarcinoma.
In the adenocarcinoma group, the main mutations were ALK (Ile1461Val), in 95.24% of patients, EML4 (Lys398Arg) in 47.62% of the patients, and EGFR (Arg521Lys) and ROS (Arg167Gln) in 42.86% of the patients (Table 3).
EGFR (Val592Ile), PIK3CA (Arg19Ile), PIK3CA (Arg852Pro), and ROS (Trp847Leu) mutations were present only in the squamous carcinoma population.
In the squamous carcinoma group, the main mutations were ALK (Ile1461Val), in 100% of patients, and EML4 (Lys398Arg) in 75% of the patients (Table 4).
Figure 4 shows the CB section of NSCLC adenocarcinoma and the immunocytochemistry evaluation.

8. Discussion

At present, there are two significant settings in which the liquid biopsy could provide an improvement to NSCLC patients: at the first molecular diagnosis and at the progression during targeted therapy [61]. A precise assessment of genetic mutations, at both baseline and progression, is critical for patient management, and to understand the process that causes therapies’ resistance [62]. Although repetitive tissue biopsies may represent the tools to monitor the tumor evolution, the feasibility of these procedures is not applicable for all patients. Therefore, several liquid biopsy platforms have been established as a complementary tool to conventional tissue biopsy and as a feasible means of identifying acquired resistance mechanisms [63,64,65,66,67,68]. Today, liquid biopsy is proposed in the new College of American Pathologists (CAP)/International Association for the Study of Lung Cancer (IASLC)/Association for Molecular Pathology (AMP) guideline for molecular testing of patients with NSCLC [69]. This approach cannot be a substitute of conventional tissue biopsy, but instead an alternative in cases with insufficient tumor tissue specimens or when a tissue specimen is not accessible [61]. In addition, circulating biomarkers are hypothetically more likely to reveal tumor burden and are more useful in representing cancer heterogeneity. Despite these benefits, there are several issues regarding sensitivity and utility, so due to its function in clinical settings, these should be evaluated in supplementary studies.
In this ongoing prospective study, at the time of writing, we assessed 29 patients with NSCLC: 21/29 with adenocarcinoma and 8/29 with squamous carcinoma. Tumor lesions were localized in the peripheral area in 86.2% (25/29 patients) and in 13.8% (4/29) in the central area. According to our results, all the genetic mutations detected in pathological analysis were found by liquid biopsy. There were no false negatives at liquid biopsy compared to pathological analysis. Although the great reliability of the results obtained in cytology has already been proven [70,71], we are well-aware that molecular analysis on cytological samples is not without limitations, as the tumor cell quality, quantity, and purity can greatly vary from specimen to specimen [72]. Despite this, cytological specimens are the main samples used for the diagnosis of advanced lung cancer and expression rates in cytological samples are not statistically different from histological samples [73].
The mutations with the highest prevalence rate in the study populations were: ALK (Ile1461Val), found in 28/29 patients (96.6%), EML4 (Lys398Arg), identified in 16/29 (55.2%) patients, ALK (Asp1529Glu), found in 14/29 (48.3%) patients, EGFR (Arg521Lys), found in 12/29 (41.4%) patients, ROS (Lys2228Gln), identified in 11/29 (37.9%) patients, ROS (Arg167Gln) and ROS (Ser2229Cys), identified in 10/29 (34.5%) patients, ALK (Lys1491Arg) and PIK3CA (Ile391Met), identified in 8/29 (27.6%) patients, ROS (Thr145Pro), identified in 6/29 (20.7%) patients, and ROS (Ser1109Leu), identified in 4/29 (13.8%) patients. KRAS (Gly12Cys) was found in 2/29 (6.9%) patients, and EGFR (Arg255Gln), EGFR (Val592Ile), EGFR (Ala647Thr), EGFR (Leu858Arg), ALK (Pro254Thr), ALK (Trp288Ser), ALK (Glu797Lys), PIK3CA (Arg19Ile), PIK3CA (Arg852Pro), PIK3CA (Met1043Ile), ROS (Glu1902Lys), ROS (Leu567Val), ROS (Phe1153Leu), and ROS (Trp847Leu) were identified only in one patient.
These results are similar to the data obtained by Kong et al. [74]. In addition, in our study, liquid biopsy was able to highlight 26 mutations, with the prevalence of the ALK (Ile1461Val) mutation in 96.6% of patients. These data disagree with several studies on conventional biopsy, which confirmed the presence of ALK fusion genes in 2–7% of NSCLC, arising more commonly in nonsmokers and almost exclusively in tumors of non-squamous histology, although this result was obtained by liquid biopsy [56]. Regarding the adenocarcinoma group, the main mutations were ALK (Ile1461Val) in 95.24% of patients, EML4 (Lys398Arg) in 47.62% of the patients, and EGFR (Arg521Lys) and ROS (Arg167Gln) in 42.86% of the patients. In the squamous carcinoma group, the main mutations were ALK (Ile1461Val) in 100% of patients and EML4 (Lys398Arg) in 75% of the patients. The EGFR (Val592Ile), PIK3CA (Arg19Ile), PIK3CA (Arg852Pro), and ROS (Trp847Leu) mutations were assessed only in patients with squamous carcinoma. Our results suggested that liquid biopsy has the ability to offer much of the data needed for clinical application of targeted therapy. The discovery of oncogenic driver mutations of the EGFR gene and the authorization of EGFR inhibitors have modified the therapy in NSCLC [75]. Patients with EGFR exon 19 deletions or exon 21 L858R mutations are suitable for treatment with afatinib, gefitinib, dacomitinib, erlotinib, and osimertin [69]. Most of the FDA-approved EGFR mutation detection tests are based on conventional biopsies. However, several plasma-based assays have been introduced for providing a non-invasive tool for patients not suitable for tissue biopsy. Liquid biopsy allows the clinicians to assess therapy effectiveness and disease progression over time. Liu et al. [75] assessed the blood samples of 24 NSCLC patients and 6 age-matched healthy donors, comparing the EGFR mutation profile detected from CTCs and cfDNA to matched tumor tissues. They showed that the results from this non-invasive EGFR mutation analysis were encouraging, and this combined workflow could represent a valuable means for informing therapy selection and for monitoring treatment of patients with NSCLC [75].
We found no statistically significant differences among the mutation status and the disease status (p > 0.05, Yates chi-square test) and no statistically significant differences could be observed in the number of each mutation between patients with adenocarcinoma and patients with squamous carcinoma (p > 0.05, Yates chi-square test), probably due to the small sample size of the populations. Unlike our results, Van der Linden et al. showed that by using a variant allele frequency threshold of 1%, somatic variants were detected in 23.5% of patients, with a median variant allele fraction of 3.65%. By using this threshold, they could discriminate early-stage lung cancer patients from the control group [76].
The detection of cfDNA offers prospects for screening, diagnosis, treatment assessment, and disease surveillance. Increasing evidence has highlighted the clinical utility of detecting mutations in cfDNA, and the amount of cfDNA in circulation has been correlated with the tumor burden [77]. The liquid biopsy offers advantages to the treatment-naive patient by saving tissue for additional assessment, comprising immunohistochemistry. In addition, this approach is simple and less expensive, providing intra-tumor heterogeneity and high sensitivity for detecting tumor burden [77]. However, not all tumors release adequate quantities of DNA. Treatment-naive patients with indolent, slow-growing tumors could be at higher risk of false-negative results in plasma compared to patients with a more disseminated cancer [75].
This study has several limits. Firstly, the small sample size makes accurate comparisons problematic, and therefore the analysis should be performed with a larger number of patients to verify and to validate the findings, and this represents a future end-point of our study. Secondly, the analysis was focused on the first diagnosis, so we had no data on genetic mutations during progression. Finally, the meanings of the 26 mutations that we found and what role these might play in patient management were not discussed and should be explored in future studies.

9. Conclusions

In this ongoing prospective study, we assessed 29 patients with NSCLC: 21/29 with adenocarcinoma and 8/29 with squamous carcinoma. All the genetic mutations detected by pathological analysis were found by liquid biopsy. There were no false negatives at liquid biopsy compared to pathological analysis. In addition, in our study, liquid biopsy was able to highlight 26 mutations, with the prevalence of the ALK mutation in 96.6% of patients, supporting the idea that this approach could be an effective tool in cases with insufficient tumor tissue specimens or in cases where tissue specimens are not obtainable. Additionally, circulating biomarkers are more likely to reveal tumor burden and are more useful in representing cancer heterogeneity. Despite these advantages, there are a number of issues concerning sensitivity and utility and the role of liquid biopsy in clinical settings that require further investigations.

Author Contributions

Formal analysis, R.F. (Roberta Fusco); investigation, M.M., G.C., V.G., F.G., A.R. (Andrea Ronchi), I.C., R.F. (Renato Franco), F.Z.M., F.U., R.M., R.S., G.S., U.C., A.N., L.A., M.T.V., V.N., A.R. (Alfonso Reginelli), and R.G.; methodology, M.M., G.C., V.G., R.F. (Roberta Fusco), F.G., A.R. (Andrea Ronchi), I.C., F.Z.M., F.U., R.M., R.S., U.C., A.N., L.A., M.T.V., V.N., A.R. (Alfonso Reginelli), and R.G.; project administration, R.F. (Renato Franco) and G.S.; writing—original draft, V.G.; writing—review and editing, V.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The Local Ethics Committee of the “Università degli Studi della Campania Luigi Vanvitelli”, with internal resolution No. 24997/2020 of 12.11.2020, approved this prospective study. The study was performed in accordance with relevant guidelines and regulations.

Informed Consent Statement

Each enrolled patient signed the informed Consent.

Data Availability Statement

All data are reported in this manuscript.

Acknowledgments

The authors are grateful to Alessandra Trocino, librarian at the National Cancer Institute of Naples, Italy. Moreover, the authors are grateful to Maria Serio for the collaboration and research support.

Conflicts of Interest

The authors have no conflict of interest to be disclosed. The authors confirm that the article is not under consideration for publication elsewhere. Each author has participated sufficiently to take public responsibility for the manuscript content.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. World Health Organization (WHO). Global Health Estimates 2020: Deaths by Cause, Age, Sex, by Country and by Region, 2000–2019; WHO: Geneva, Switzerland, 2020; Available online:https://www.who.int/data/gho/data/themes/mortality-and-global-health-estimates/ghe-leading-causes-of-death (accessed on 11 December 2020).
  3. Granata, V.; Grassi, R.; Fusco, R.; Belli, A.; Cutolo, C.; Pradella, S.; Grazzini, G.; La Porta, M.; Brunese, M.C.; De Muzio, F.; et al. Diagnostic evaluation and ablation treatments assessment in hepatocellular carcinoma. Infect. Agents Cancer 2021, 16, 53. [Google Scholar] [CrossRef]
  4. Barabino, M.; Gurgitano, M.; Fochesato, C.; Angileri, S.A.; Franceschelli, G.; Santambrogio, R.; Mariani, N.M.; Opocher, E.; Carrafiello, G. LI-RADS to categorize liver nodules in patients at risk of HCC: Tool or a gadget in daily practice? Radiol. Med. 2021, 126, 5–13. [Google Scholar] [CrossRef]
  5. Gabelloni, M.; Di Nasso, M.; Morganti, R.; Faggioni, L.; Masi, G.; Falcone, A.; Neri, E. Application of the ESR iGuide clinical decision support system to the imaging pathway of patients with hepatocellular carcinoma and cholangiocarcinoma: Preliminary findings. Radiol. Med. 2020, 125, 531–537. [Google Scholar] [CrossRef] [PubMed]
  6. Granata, V.; Grassi, R.; Fusco, R.; Setola, S.V.; Belli, A.; Ottaiano, A.; Nasti, G.; La Porta, M.; Danti, G.; Cappabianca, S.; et al. Intrahepatic cholangiocarcinoma and its differential diagnosis at MRI: How radiologist should assess MR features. Radiol. Med. 2021, 126, 1584–1600. [Google Scholar] [CrossRef] [PubMed]
  7. Gatti, M.; Calandri, M.; Bergamasco, L.; Darvizeh, F.; Grazioli, L.; Inchingolo, R.; Ippolito, D.; Rousset, S.; Veltri, A.; Fonio, P.; et al. Characterization of the arterial enhancement pattern of focal liver lesions by multiple arterial phase magnetic resonance imaging: Comparison between hepatocellular carcinoma and focal nodular hyperplasia. Radiol. Med. 2020, 125, 348–355. [Google Scholar] [CrossRef] [PubMed]
  8. Orlacchio, A.; Chegai, F.; Roma, S.; Merolla, S.; Bosa, A.; Francioso, S. Degradable starch microspheres transarterial chemoembolization (DSMs-TACE) in patients with unresectable hepatocellular carcinoma (HCC): Long-term results from a single-center 137-patient cohort prospective study. Radiol. Med. 2020, 125, 98–106. [Google Scholar] [CrossRef] [PubMed]
  9. Vietri, M.T.; Caliendo, G.; Casamassimi, A.; Cioffi, M.; De Paola, M.L.; Napoli, C.; Molinari, A.M. A novel PALB2 truncating mutation in an Italian family with male breast cancer. Oncol. Rep. 2015, 33, 1243–1247. [Google Scholar] [CrossRef] [Green Version]
  10. Vietri, M.T.; Caliendo, G.; Schiano, C.; Casamassimi, A.; Molinari, A.M.; Napoli, C.; Cioffi, M. Analysis of PALB2 in a cohort of Italian breast cancer patients: Identification of a novel PALB2 truncating mutation. Fam. Cancer 2015, 14, 341–348. [Google Scholar] [CrossRef]
  11. Granata, V.; Fusco, R.; Avallone, A.; Catalano, O.; Filice, F.; Leongito, M.; Palaia, R.; Izzo, F.; Petrillo, A. Major and ancillary magnetic resonance features of LI-RADS to assess HCC: An overview and update. Infect. Agents Cancer 2017, 12, 23. [Google Scholar] [CrossRef]
  12. Granata, V.; Fusco, R.; Avallone, A.; Filice, F.; Tatangelo, F.; Piccirillo, M.; Grassi, R.; Izzo, F.; Petrillo, A. Critical analysis of the major and ancillary imaging features of LI-RADS on 127 proven HCCs evaluated with functional and morphological MRI: Lights and shadows. Oncotarget 2017, 8, 51224–51237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cozzolino, I.; Ronchi, A.; Messina, G.; Montella, M.; Morgillo, F.; Vicidomini, G.; Tirino, V.; Grimaldi, A.; Marino, F.Z.; Santini, M.; et al. Adequacy of Cytologic Samples by Ultrasound-Guided Percutaneous Transthoracic Fine-Needle Aspiration Cytology of Peripheral Pulmonary Nodules for Morphologic Diagnosis and Molecular Evaluations: Comparison with Computed Tomography-Guided Percutaneous Transthoracic Fine-Needle Aspiration Cytology. Arch. Pathol. Lab. Med. 2020, 144, 361–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Granata, V.; Fusco, R.; Filice, S.; Catalano, O.; Piccirillo, M.; Palaia, R.; Izzo, F.; Petrillo, A. The current role and future prospectives of functional parameters by diffusion weighted imaging in the assessment of histologic grade of HCC. Infect. Agents Cancer 2018, 13, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. De Filippo, M.; Ziglioli, F.; Russo, U.; Pagano, P.; Brunese, L.; Bertelli, E.; Pagnini, F.; Maestroni, U. Radiofrequency ablation (RFA) of T1a renal cancer with externally cooled multitined expandable electrodes. Radiol. Med. 2020, 125, 790–797. [Google Scholar] [CrossRef]
  16. Arrigoni, F.; Bruno, F.; Gianneramo, C.; Palumbo, P.; Zugaro, L.; Zoccali, C.; Barile, A.; Masciocchi, C. Evolution of the imaging features of osteoid osteoma treated with RFA or MRgFUS during a long-term follow-up: A pictorial review with clinical correlations. Radiol. Med. 2020, 125, 578–584. [Google Scholar] [CrossRef]
  17. Fusco, R.; Granata, V.; Sansone, M.; Rega, D.; Delrio, P.; Tatangelo, F.; Romano, C.; Avallone, A.; Pupo, D.; Giordano, M.; et al. Validation of the standardized index of shape tool to analyze DCE-MRI data in the assessment of neo-adjuvant therapy in locally advanced rectal cancer. Radiol. Med. 2021, 126, 1044–1054. [Google Scholar] [CrossRef]
  18. Fusco, R.; Granata, V.; Mazzei, M.A.; Meglio, N.D.; Roscio, D.D.; Moroni, C.; Monti, R.; Cappabianca, C.; Picone, C.; Neri, E.; et al. Quantitative imaging decision support (QIDSTM) tool consistency evaluation and radiomic analysis by means of 594 metrics in lung carcinoma on chest CT scan. Cancer Control 2021, 28, 1073274820985786. [Google Scholar] [CrossRef]
  19. Granata, V.; Fusco, R.; Avallone, A.; De Stefano, A.; Ottaiano, A.; Sbordone, C.; Brunese, L.; Izzo, F.; Petrillo, A. Radiomics-Derived Data by Contrast Enhanced Magnetic Resonance in RAS Mutations Detection in Colorectal Liver Metastases. Cancers 2021, 13, 453. [Google Scholar] [CrossRef] [PubMed]
  20. Granata, V.; Fusco, R.; Risi, C.; Ottaiano, A.; Avallone, A.; De Stefano, A.; Grimm, R.; Grassi, R.; Brunese, L.; Izzo, F.; et al. Diffusion-Weighted MRI and Diffusion Kurtosis Imaging to Detect RAS Mutation in Colorectal Liver Metastasis. Cancers 2020, 12, 2420. [Google Scholar] [CrossRef]
  21. Granata, V.; Fusco, R.; Catalano, O.; Piccirillo, M.; De Bellis, M.; Izzo, F.; Petrillo, A. Percutaneous ablation therapy of hepatocellular carcinoma with irreversible electroporation: MRI findings. AJR Am. J. Roentgenol. 2015, 204, 1000–1007. [Google Scholar] [CrossRef] [PubMed]
  22. Avallone, A.; Pecori, B.; Bianco, F.; Aloj, L.; Tatangelo, F.; Romano, C.; Granata, V.; Marone, P.; Leone, A.; Botti, G.; et al. Critical role of bevacizumab scheduling in combination with pre-surgical chemo-radiotherapy in MRI-defined high-risk locally advanced rectal cancer: Results of the BRANCH trial. Oncotarget 2015, 6, 30394–30407. [Google Scholar] [CrossRef] [Green Version]
  23. Trombadori, C.M.L.; DAngelo, A.; Ferrara, F.; Santoro, A.; Belli, P.; Manfredi, R. Radial Scar: A management dilemma. Radiol. Med. 2021, 126, 774–785. [Google Scholar] [CrossRef]
  24. Argalia, G.; Tarantino, G.; Ventura, C.; Campioni, D.; Tagliati, C.; Guardati, P.; Kostandini, A.; Marzioni, M.; Giuseppetti, G.M.; Giovagnoni, A. Shear wave elastography and transient elastography in HCV patients after direct-acting antivirals. Radiol. Med. 2021, 126, 894–899. [Google Scholar] [CrossRef] [PubMed]
  25. Trimboli, P.; Castellana, M.; Virili, C.; Havre, R.F.; Bini, F.; Marinozzi, F.; D’Ambrosio, F.; Giorgino, F.; Giovanella, L.; Prosch, H.; et al. Performance of contrast-enhanced ultrasound (CEUS) in assessing thyroid nodules: A systematic review and meta-analysis using histological standard of reference. Radiol. Med. 2020, 125, 406–415. [Google Scholar] [CrossRef]
  26. Chen, M.; Zhao, H. Next-generation sequencing in liquid biopsy: Cancer screening and early detection. Hum. Genom. 2019, 13, 34. [Google Scholar] [CrossRef] [Green Version]
  27. Izzo, F.; Granata, V.; Fusco, R.; D’Alessio, V.; Petrillo, A.; Lastoria, S.; Piccirillo, M.; Albino, V.; Belli, A.; Tafuto, S.; et al. Clinical Phase I/II Study: Local Disease Control and Survival in Locally Advanced Pancreatic Cancer Treated with Electrochemotherapy. J. Clin. Med. 2021, 10, 1305. [Google Scholar] [CrossRef]
  28. Fusco, R.; Petrillo, M.; Granata, V.; Filice, S.; Sansone, M.; Catalano, O.; Petrillo, A. Magnetic Resonance Imaging Evaluation in Neoadjuvant Therapy of Locally Advanced Rectal Cancer: A Systematic Review. Radiol. Oncol. 2017, 51, 252–262. [Google Scholar] [CrossRef]
  29. Laurelli, G.; Falcone, F.; Gallo, M.S.; Scala, F.; Losito, S.; Granata, V.; Cascella, M.; Greggi, S. Long-Term Oncologic and Reproductive Outcomes in Young Women with Early Endometrial Cancer Conservatively Treated: A Prospective Study and Literature Update. Int. J. Gynecol. Cancer 2016, 26, 1650–1657. [Google Scholar] [CrossRef]
  30. Fusco, R.; Sansone, M.; Granata, V.; Setola, S.V.; Petrillo, A. A systematic review on multiparametric MR imaging in prostate cancer detection. Infect. Agents Cancer 2017, 12, 57. [Google Scholar] [CrossRef] [Green Version]
  31. Rega, D.; Pace, U.; Scala, D.; Chiodini, P.; Granata, V.; Fares Bucci, A.; Pecori, B.; Delrio, P. Treatment of splenic flexure colon cancer: A comparison of three different surgical procedures: Experience of a high volume cancer center. Sci. Rep. 2019, 9, 10953. [Google Scholar] [CrossRef]
  32. Kwapisz, D. The first liquid biopsy test approved. is it a new era of mutation testing for non-small cell lung cancer? Ann. Transl. Med. 2017, 5, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Crowley, E.; Di Nicolantonio, F.; Loupakis, F.; Bardelli, A. Liquid biopsy: Monitoring cancer-genetics in the blood. Nat. Rev. Clin. Oncol. 2013, 10, 472. [Google Scholar] [CrossRef] [PubMed]
  34. Yoshioka, Y.; Kosaka, N.; Konishi, Y.; Ohta, H.; Okamoto, H.; Sonoda, H.; Nonaka, R.; Yamamoto, H.; Ishii, H.; Mori, M.; et al. Ultra-sensitive liquid biopsy of circulating extracellular vesicles using exoscreen. Nat. Commun. 2014, 5, 3591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Pantel, K.; Alix-Panabières, C. Real-time liquid biopsy in cancer patients: Fact or fiction? Cancer Res. 2013, 73, 6384–6388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Labib, M.; Mohamadi, R.M.; Poudineh, M.; Ahmed, S.U.; Ivanov, I.; Huang, C.-L.; Moosavi, M.; Sargent, E.H.; Kelley, S.O. Single-cell mrna cytometry via sequence- specific nanoparticle clustering and trapping. Nat. Chem. 2018, 10, 489–495. [Google Scholar] [CrossRef] [PubMed]
  37. Stroun, M.; Lyautey, J.; Lederrey, C.; Olson-Sand, A.; Anker, P. About the possible origin and mechanism of circulating DNA: Apoptosis and active DNA release. Clin. Chim. Acta 2001, 313, 139–142. [Google Scholar] [CrossRef]
  38. Siena, S.; Sartore-Bianchi, A.; Garcia-Carbonero, R.; Karthaus, M.; Smith, D.; Tabernero, J.; Van Cutsem, E.; Guan, X.; Boedigheimer, M.; Ang, A.; et al. Dynamic molecular analysis and clinical correlates of tumor evolution within a phase ii trial of panitumumab-based therapy in metastatic colorectal cancer. Ann. Oncol. 2017, 29, 119–126. [Google Scholar] [CrossRef]
  39. Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223. [Google Scholar] [CrossRef]
  40. Granata, V.; Fusco, R.; Setola, S.V.; De Muzio, F.; Dell’ Aversana, F.; Cutolo, C.; Faggioni, L.; Miele, V.; Izzo, F.; Petrillo, A. CT-Based Radiomics Analysis to Predict Histopathological Outcomes Following Liver Resection in Colorectal Liver Metastases. Cancers 2022, 14, 1648. [Google Scholar] [CrossRef]
  41. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Setola, S.V.; Grassi, R.; Grassi, F.; Ottaiano, A.; Nasti, G.; Tatangelo, F.; et al. Radiomics textural features by MR imaging to assess clinical outcomes following liver resection in colorectal liver metastases. Radiol. Med. 2022, 127, 461–470. [Google Scholar] [CrossRef]
  42. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Setola, S.V.; Dell’Aversana, F.; Ottaiano, A.; Nasti, G.; Grassi, R.; Pilone, V.; et al. EOB-MR Based Radiomics Analysis to Assess Clinical Outcomes following Liver Resection in Colorectal Liver Metastases. Cancers 2022, 14, 1239. [Google Scholar] [CrossRef] [PubMed]
  43. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Setola, S.V.; Dell’ Aversana, F.; Ottaiano, A.; Avallone, A.; Nasti, G.; Grassi, F.; et al. Contrast MR-Based Radiomics and Machine Learning Analysis to Assess Clinical Outcomes following Liver Resection in Colorectal Liver Metastases: A Preliminary Study. Cancers 2022, 14, 1110. [Google Scholar] [CrossRef] [PubMed]
  44. Rampado, O.; Depaoli, A.; Marchisio, F.; Gatti, M.; Racine, D.; Ruggeri, V.; Ruggirello, I.; Darvizeh, F.; Fonio, P.; Ropolo, R. Effects of different levels of CT iterative reconstruction on low-contrast detectability and radiation dose in patients of different sizes: An anthropomorphic phantom study. Radiol. Med. 2021, 126, 55–62. [Google Scholar] [CrossRef] [PubMed]
  45. Shin, N.; Choi, J.A.; Choi, J.M.; Cho, E.S.; Kim, J.H.; Chung, J.J.; Yu, J.S. Sclerotic changes of cavernous hemangioma in the cirrhotic liver: Long-term follow-up using dynamic contrast-enhanced computed tomography. Radiol. Med. 2020, 125, 1225–1232. [Google Scholar] [CrossRef] [PubMed]
  46. Bottari, A.; Silipigni, S.; Carerj, M.L.; Cattafi, A.; Maimone, S.; Marino, M.A.; Mazziotti, S.; Pitrone, A.; Squadrito, G.; Ascenti, G. Dual-source dual-energy CT in the evaluation of hepatic fractional extracellular space in cirrhosis. Radiol. Med. 2020, 125, 7–14. [Google Scholar] [CrossRef]
  47. Tangvarasittichai, O.; Jaiwang, W.; Tangvarasittichai, S. The plasma DNA concentration as a potential breast cancer screening marker. Indian J. Clin. Biochem. 2015, 30, 55–58. [Google Scholar] [CrossRef] [Green Version]
  48. Lehmann-Werman, R.; Neiman, D.; Zemmour, H.; Moss, J.; Magenheim, J.; Vaknin-Dembinsky, A.; Rubertsson, S.; Nellgård, B.; Blennow, K.; Zetterberg, H.; et al. Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proc. Natl. Acad. Sci. USA 2016, 113, E1826–34. [Google Scholar] [CrossRef] [Green Version]
  49. Thakur, B.K.; Zhang, H.; Becker, A.; Matei, I.; Huang, Y.; Costa-Silva, B.; Zheng, Y.; Hoshino, A.; Brazier, H.; Xiang, J.; et al. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 2014, 24, 766. [Google Scholar] [CrossRef] [Green Version]
  50. Granata, V.; Fusco, R.; Costa, M.; Picone, C.; Cozzi, D.; Moroni, C.; La Casella, G.V.; Montanino, A.; Monti, R.; Mazzoni, F.; et al. Preliminary Report on Computed Tomography Radiomics Features as Biomarkers to Immunotherapy Selection in Lung Adenocarcinoma Patients. Cancers 2021, 13, 3992. [Google Scholar] [CrossRef]
  51. Granata, V.; Grassi, R.; Miele, V.; Larici, A.R.; Sverzellati, N.; Cappabianca, S.; Brunese, L.; Maggialetti, N.; Borghesi, A.; Fusco, R.; et al. Structured Reporting of Lung Cancer Staging: A Consensus Proposal. Diagnostics 2021, 11, 1569. [Google Scholar] [CrossRef]
  52. Nardone, V.; Reginelli, A.; Grassi, R.; Boldrini, L.; Vacca, G.; D’Ippolito, E.; Annunziata, S.; Farchione, A.; Belfiore, M.P.; Desideri, I.; et al. Delta radiomics: A systematic review. Radiol. Med. 2021, 126, 1571–1583. [Google Scholar] [CrossRef] [PubMed]
  53. Bracci, S.; Dolciami, M.; Trobiani, C.; Izzo, A.; Pernazza, A.; D’Amati, G.; Manganaro, L.; Ricci, P. Quantitative CT texture analysis in predicting PD-L1 expression in locally advanced or metastatic NSCLC patients. Radiol. Med. 2021, 126, 1425–1433. [Google Scholar] [CrossRef] [PubMed]
  54. Merlotti, A.; Bruni, A.; Borghetti, P.; Ramella, S.; Scotti, V.; Trovò, M.; Chiari, R.; Lohr, F.; Ricardi, U.; Bria, E.; et al. Sequential chemo-hypofractionated RT versus concurrent standard CRT for locally advanced NSCLC: GRADE recommendation by the Italian Association of Radiotherapy and Clinical Oncology (AIRO). Radiol. Med. 2021, 126, 1117–1128. [Google Scholar] [CrossRef] [PubMed]
  55. Sarkar, B.; Ganesh, T.; Munshi, A.; Manikandan, A.; Roy, S.; Krishnankutty, S.; Chitral, L.; Sathiya, J.; Pradhan, A.; Mohanti, B.K. Rotational positional error-corrected linear set-up margin calculation technique for lung stereotactic body radiotherapy in a dual imaging environment of 4-D cone beam CT and ExacTrac stereoscopic imaging. Radiol. Med. 2021, 126, 979–988. [Google Scholar] [CrossRef] [PubMed]
  56. Agazzi, G.M.; Ravanelli, M.; Roca, E.; Medicina, D.; Balzarini, P.; Pessina, C.; Vermi, W.; Berruti, A.; Maroldi, R.; Farina, D. CT texture analysis for prediction of EGFR mutational status and ALK rearrangement in patients with non-small cell lung cancer. Radiol. Med. 2021, 126, 786–794. [Google Scholar] [CrossRef]
  57. Kim, B.H.; Kim, J.S.; Kim, K.H.; Moon, H.J.; Kim, S. Clinical significance of radiation dose-volume parameters and functional status on the patient-reported quality of life changes after thoracic radiotherapy for lung cancer: A prospective study. Radiol. Med. 2021, 126, 466–473. [Google Scholar] [CrossRef]
  58. European Medicines Agency. EMA. Tarceva. 2018. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/tarceva (accessed on 24 July 2022).
  59. US Food FDA and Drug Administration. Premarket Approval P150044 Cobas EGFR MUTATION TEST V2. 2018. Available online: https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?id=P150044 (accessed on 24 July 2022).
  60. Goldberg, S.B.; Narayan, A.; Kole, A.J.; Decker, R.H.; Teysir, J.; Carriero, N.J.; Lee, A.; Nemati, R.; Nath, S.K.; Mane, S.M.; et al. Early assessment of lung cancer immunotherapy response via circulating tumor DNA. Clin. Cancer Res. 2018, 24, 1341. [Google Scholar] [CrossRef] [Green Version]
  61. Rolfo, C.; Mack, P.C.; Scagliotti, G.V.; Baas, P.; Barlesi, F.; Bivona, T.G.; Herbst, R.S.; Mok, T.S.; Peled, N.; Pirker, R.; et al. Liquid Biopsy for Advanced Non-Small Cell Lung Cancer (NSCLC): A Statement Paper from the IASLC. J. Thorac. Oncol. 2018, 13, 1248–1268. [Google Scholar] [CrossRef] [Green Version]
  62. Sorber, L.; Zwaenepoel, K.; Deschoolmeester, V.; Van Schil, P.E.; Van Meerbeeck, J.; Lardon, F.; Rolfo, C.; Pauwels, P. Circulating cell-free nucleic acids and platelets as a liquid biopsy in the provision of personalized therapy for lung cancer patients. Lung Cancer 2017, 107, 100–107. [Google Scholar] [CrossRef]
  63. Garcia, J.; Wozny, A.S.; Geiguer, F.; Delherme, A.; Barthelemy, D.; Merle, P.; Tissot, C.; Jones, F.S.; Johnson, C.; Xing, X.; et al. Profiling of circulating tumor DNA in plasma of non-small cell lung cancer patients, monitoring of epidermal growth factor receptor p.T790M mutated allelic fraction using beads, emulsion, amplification, and magnetics companion assay and evaluation in future application in mimicking circulating tumor cells. Cancer Med. 2019, 8, 3685–3697. [Google Scholar] [CrossRef]
  64. Otsubo, K.; Sakai, K.; Takeshita, M.; Harada, D.; Azuma, K.; Ota, K.; Akamatsu, H.; Goto, K.; Horiike, A.; Kurata, T.; et al. Genetic Profiling of Non-Small Cell Lung Cancer at Development of Resistance to First- or Second-Generation EGFR-TKIs by CAPP-Seq Analysis of Circulating Tumor DNA. Oncologist 2019, 24, 1022–1026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liheng, M.; Guofan, X.; Balzano, R.F.; Yuying, L.; Weifeng, H.; Ning, Y.; Yayun, J.; Mouyuan, L.; Guglielmi, G. The value of DTI: Achieving high diagnostic performance for brain metastasis. Radiol. Med. 2021, 126, 291–298. [Google Scholar] [CrossRef] [PubMed]
  66. Falcinelli, L.; Mendichi, M.; Chierchini, S.; Tenti, M.V.; Bellavita, R.; Saldi, S.; Ingrosso, G.; Reggioli, V.; Bini, V.; Aristei, C. Pulmonary function in stereotactic body radiotherapy with helical tomotherapy for primary and metastatic lung lesions. Radiol. Med. 2021, 126, 163–169. [Google Scholar] [CrossRef] [PubMed]
  67. Farchione, A.; Larici, A.R.; Masciocchi, C.; Cicchetti, G.; Congedo, M.T.; Franchi, P.; Gatta, R.; Lo Cicero, S.; Valentini, V.; Bonomo, L.; et al. Exploring technical issues in personalized medicine: NSCLC survival prediction by quantitative image analysis-usefulness of density correction of volumetric CT data. Radiol. Med. 2020, 125, 625–635. [Google Scholar] [CrossRef]
  68. Zhang, G.; Yang, Z.; Gong, L.; Jiang, S.; Wang, L.; Zhang, H. Classification of lung nodules based on CT images using squeeze-and-excitation network and aggregated residual transformations. Radiol. Med. 2020, 125, 374–383. [Google Scholar] [CrossRef] [PubMed]
  69. Lindeman, N.I.; Cagle, P.T.; Aisner, D.L.; Arcila, M.E.; Beasley, M.B.; Bernicker, E.H.; Colasacco, C.; Dacic, S.; Hirsch, F.R.; Kerr, K.; 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. J. Thorac. Oncol. 2018, 13, 323–358. [Google Scholar] [CrossRef] [Green Version]
  70. Vietri, M.T.; Caliendo, G.; D’Elia, G.; Resse, M.; Casamassimi, A.; Minucci, P.B.; Dello Ioio, C.; Cioffi, M.; Molinari, A.M. Five Italian Families with Two Mutations in BRCA Genes. Genes (Basel) 2020, 11, 1451. [Google Scholar] [CrossRef] [PubMed]
  71. Zito Marino, F.; Rossi, G.; Cozzolino, I.; Montella, M.; Micheli, M.; Bogina, G.; Munari, E.; Brunelli, M.; Franco, R. Multiplex fluorescence in situ hybridisation to detect anaplastic lymphoma kinase and ROS proto-oncogene 1 receptor tyrosine kinase rearrangements in lung cancer cytological samples. J. Clin. Pathol. 2020, 73, 96–101. [Google Scholar] [CrossRef] [PubMed]
  72. VanderLaan, P.A. Non-small cell lung cancer predictive biomarker testing via immunocytochemistry: Ways of future past? Cancer Cytopathol. 2019, 127, 278–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ekin, Z.; Nart, D.; Savaş, P.; Veral, A. Comparison of PD-L1, EGFR, ALK, and ROS1 Status Between Surgical Samples and Cytological Samples in Non-Small Cell Lung Carcinoma. Balkan Med. J. 2021, 38, 287–295. [Google Scholar] [CrossRef] [PubMed]
  74. Kong, F.; Li, Y.; Guo, R.; Yang, L.; Di, J.; He, L.; Wang, Z.; Liu, D.; Li, X. Liquid biopsy assay for pulmonary adenocarcinoma using supernatants from core-needle biopsy specimens. Thorac. Cancer 2022, 13, 1822–1826. [Google Scholar] [CrossRef]
  75. Liu, H.E.; Vuppalapaty, M.; Wilkerson, C.; Renier, C.; Chiu, M.; Lemaire, C.; Che, J.; Matsumoto, M.; Carroll, J.; Crouse, S.; et al. Detection of EGFR Mutations in cfDNA and CTCs, and Comparison to Tumor Tissue in Non-Small-Cell-Lung-Cancer (NSCLC) Patients. Front. Oncol. 2020, 10, 572895. [Google Scholar] [CrossRef]
  76. Van der Linden, M.; Van Gaever, B.; Raman, L.; Vermaelen, K.; Demedts, I.; Surmont, V.; Himpe, U.; Lievens, Y.; Ferdinande, L.; Dedeurwaerdere, F.; et al. Application of an Ultrasensitive NGS-Based Blood Test for the Diagnosis of Early-Stage Lung Cancer: Sensitivity, a Hurdle Still Difficult to Overcome. Cancers 2022, 14, 2031. [Google Scholar] [CrossRef]
  77. Kapeleris, J.; Ebrahimi Warkiani, M.; Kulasinghe, A.; Vela, I.; Kenny, L.; Ladwa, R.; O’Byrne, K.; Punyadeera, C. Clinical Applications of Circulating Tumour Cells and Circulating Tumour DNA in Non-Small Cell Lung Cancer—An Update. Front. Oncol. 2022, 12, 859152. [Google Scholar] [CrossRef]
Figure 1. Distribution of location of tumors and type of carcinoma.
Figure 1. Distribution of location of tumors and type of carcinoma.
Jpm 12 01896 g001
Figure 2. Mutation status at liquid biopsy for a single patient (A = adenocarcinoma and S = squamous carcinoma).
Figure 2. Mutation status at liquid biopsy for a single patient (A = adenocarcinoma and S = squamous carcinoma).
Jpm 12 01896 g002
Figure 3. Number of mutated patients in the adenocarcinoma and squamous carcinoma groups.
Figure 3. Number of mutated patients in the adenocarcinoma and squamous carcinoma groups.
Jpm 12 01896 g003
Figure 4. (A) CB section of NSCLC adenocarcinoma (40× magnification, scale bar 50 µm, H/E staining). (BD) Immunocytochemistry evaluation: (B) ALK-negative (Clone D5F3), (C) ROS1-positive (Clone SP384), and (D) PDL1-positive & gt; 50% (Clone SP263) (ICC assay on Ventana Benchmark XT, 40× magnification, scale bar 50 µm).
Figure 4. (A) CB section of NSCLC adenocarcinoma (40× magnification, scale bar 50 µm, H/E staining). (BD) Immunocytochemistry evaluation: (B) ALK-negative (Clone D5F3), (C) ROS1-positive (Clone SP384), and (D) PDL1-positive & gt; 50% (Clone SP263) (ICC assay on Ventana Benchmark XT, 40× magnification, scale bar 50 µm).
Jpm 12 01896 g004
Table 1. TNM and stage distribution.
Table 1. TNM and stage distribution.
ValueT (No. Patients)N (No. Patients)M (No. Patients)Stage (No. Patients)
0 2021
1151811
275 4
343 6
43 8
Table 2. Mutations found in pathological analysis.
Table 2. Mutations found in pathological analysis.
MutationNumber of Mutations Found in Adenocarcinoma Patients
EGFR Exon 211
ROS12
ALK0
PDL12
Table 3. Mutation status at liquid biopsy compared, respectively, to T, N, and M, and to grading.
Table 3. Mutation status at liquid biopsy compared, respectively, to T, N, and M, and to grading.
EGFRKRAS (Gly12Cys)2ALKEML4 (Lys398Arg)2PIK3CAROS
Mutation Status Compared to T Stage
Negative171408103
2360261
3340332
4230011
Positive181157512
2417516
3104112
4103322
p-Value, Yates’ Chi-Square0.970.831.00.600.920.91
Mutation status compared to N stage
Negative0111809155
1010100
2150240
3330112
Positive0922011515
1101011
2405325
3003221
p-Value, Yates’ Chi-Square0.540.331.00.990.940.57
Mutation status compared to M stage
Negative01020010164
1570343
Positive01112111517
1318545
p-Value, Yates’ Chi-Square0.760.931.00.940.360.58
Mutation status compared to grading
Negative15110493
2230430
3360241
4570343
Positive16011728
2214014
3306425
4318545
p-Value, Yates’ Chi-Square0.940.911.00.600.770.91
Table 4. Mutation status in adenocarcinoma and squamous carcinoma groups.
Table 4. Mutation status in adenocarcinoma and squamous carcinoma groups.
MutationAdenocarcinoma%Squamous Carcinoma%p-Value, Yates’ Chi-Square Test
EGFR (Arg255Gln)14.76000.61
EGFR (Arg521Lys)942.86337.50.93
EGFR (Val592Ile)00.00112.50.61
EGFR (Ala647Thr)14.76000.61
EGFR (Leu858Arg)14.76000.61
KRAS (Gly12Cys) 229.52000.93
ALK (Asp1529Glu)838.106750.17
ALK (Ile1461Val)2095.2481000.66
ALK (Lys1491Arg)628.572250.79
ALK (Pro254Thr)14.76000.61
ALK (Trp288Ser)14.76000.61
ALK (Glu797Lys)14.76000.61
EML4 (Lys398Arg)21047.626750.36
PIK3CA (Arg19Ile)00.00112.50.61
PIK3CA (Ile391Met)628.572250.79
PIK3CA (Arg852Pro)00.00112.50.61
PIK3CA (Met1043Ile)14.76000.61
ROS (Glu1902Lys)14.76000.61
ROS (Lys2228Gln)838.10337.50.69
ROS (Leu567Val)14.76000.61
ROS (Phe1153Leu)14.76000.61
ROS (Ser1109Leu)314.29112.50.63
ROS (Ser2229Cys)733.33337.50.82
ROS (Thr145Pro)523.81112.50.87
ROS (Trp847Leu)00.00112.50.61
ROS (Arg167Gln)942.86112.50.27
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Montella, M.; Ciani, G.; Granata, V.; Fusco, R.; Grassi, F.; Ronchi, A.; Cozzolino, I.; Franco, R.; Zito Marino, F.; Urraro, F.; et al. Preliminary Experience of Liquid Biopsy in Lung Cancer Compared to Conventional Assessment: Light and Shadows. J. Pers. Med. 2022, 12, 1896. https://doi.org/10.3390/jpm12111896

AMA Style

Montella M, Ciani G, Granata V, Fusco R, Grassi F, Ronchi A, Cozzolino I, Franco R, Zito Marino F, Urraro F, et al. Preliminary Experience of Liquid Biopsy in Lung Cancer Compared to Conventional Assessment: Light and Shadows. Journal of Personalized Medicine. 2022; 12(11):1896. https://doi.org/10.3390/jpm12111896

Chicago/Turabian Style

Montella, Marco, Giovanni Ciani, Vincenza Granata, Roberta Fusco, Francesca Grassi, Andrea Ronchi, Immacolata Cozzolino, Renato Franco, Federica Zito Marino, Fabrizio Urraro, and et al. 2022. "Preliminary Experience of Liquid Biopsy in Lung Cancer Compared to Conventional Assessment: Light and Shadows" Journal of Personalized Medicine 12, no. 11: 1896. https://doi.org/10.3390/jpm12111896

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