scholarly journals Liquid Biopsy for Disease Monitoring in Non-Small Cell Lung Cancer: The Link between Biology and the Clinic

Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 1912
Author(s):  
Maria Gabriela O. Fernandes ◽  
Catarina Sousa ◽  
Joana Pereira Reis ◽  
Natália Cruz-Martins ◽  
Conceição Souto Moura ◽  
...  

Introduction: Cell-free DNA (cfDNA) analysis offers a non-invasive method to identify sensitising and resistance mutations in advanced Non-Small Cell Lung Cancer (NSCLC) patients. Next-generation sequencing (NGS) of circulating free DNA (cfDNA) is a valuable tool for mutations detection and disease′s clonal monitoring. Material and methods: An amplicon-based targeted gene NGS panel was used to analyse 101 plasma samples of advanced non-small cell lung cancer (NSCLC) patients with known oncogenic mutations, mostly EGFR mutations, serially collected at different clinically relevant time points of the disease. Results: The variant allelic frequency (VAF) monitoring in consecutive plasma samples demonstrated different molecular response and progression patterns. The decrease in or the clearance of the mutant alleles was associated with response and the increase in or the emergence of novel alterations with progression. At the best response, the median VAF was 0% (0.0% to 3.62%), lower than that at baseline, with a median of 0.53% (0.0% to 9.9%) (p = 0.004). At progression, the VAF was significantly higher (median 4.67; range: 0.0–36.9%) than that observed at the best response (p = 0.001) and baseline (p = 0.006). These variations anticipated radiographic changes in most cases, with a median time of 0.86 months. Overall, the VAF evolution of different oncogenic mutations predicts clinical outcomes. Conclusion: The targeted NGS of circulating tumour DNA (ctDNA) has clinical utility to monitor treatment response in patients with advanced lung adenocarcinoma.

Author(s):  
Jun Lu ◽  
Wei Zhang ◽  
Lele Zhang ◽  
Yuqing Lou ◽  
Ping Gu ◽  
...  

Abstract Background Anlotinib has been demonstrated to be effective in advanced non-small cell lung cancer (NSCLC) patients. The underlying value of proteomics for anlotinib study remains unclear. Methods In this study, plasma samples from 28 anlotinib-treated NSCLC patients (including 14 responders and 14 non-responders) were performed proteomics analysis. LC-MS/MS analysis was performed on those samples with different time points including baseline, best response and progression disease. Bioinformatics analysis was performed to understand the underlying value of those differential proteins. Results Proteomics analysis suggested the differential proteins from responders after anlotinib administration potential play a role in the molecular mechanism characterization and biomarker screening. The differential proteins between responders and non-responders at baseline mainly contribute to biomarker screening. Integrative analysis indicated 43 proteins could be used as underlying biomarkers for clinical practice. Lastly, we selected ARHGDIB and demonstrated that it has potential predictive value for anlotinib. Conclusions This study not only offered the first insight that the proteomic technology potentially be used for anlotinib molecular mechanism characterization, but also provided a basis for anlotinb biomarker screening via proteomics in the future.


Cancers ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 3804
Author(s):  
Riziero Esposito Abate ◽  
Daniela Frezzetti ◽  
Monica Rosaria Maiello ◽  
Marianna Gallo ◽  
Rosa Camerlingo ◽  
...  

Lung cancer (LC) is the main cause of death for cancer worldwide and non-small cell lung cancer (NSCLC) represents the most common histology. The discovery of genomic alterations in driver genes that offer the possibility of therapeutic intervention has completely changed the approach to the diagnosis and therapy of advanced NSCLC patients, and tumor molecular profiling has become mandatory for the choice of the most appropriate therapeutic strategy. However, in approximately 30% of NSCLC patients tumor tissue is inadequate for biomarker analysis. The development of highly sensitive next generation sequencing (NGS) technologies for the analysis of circulating cell-free DNA (cfDNA) is emerging as a valuable alternative to assess tumor molecular landscape in case of tissue unavailability. Additionally, cfDNA NGS testing can better recapitulate NSCLC heterogeneity as compared with tissue testing. In this review we describe the main advantages and limits of using NGS-based cfDNA analysis to guide the therapeutic decision-making process in advanced NSCLC patients, to monitor the response to therapy and to identify mechanisms of resistance early. Therefore, we provide evidence that the implementation of cfDNA NGS testing in clinical research and in the clinical practice can significantly improve precision medicine approaches in patients with advanced NSCLC.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e21505-e21505
Author(s):  
Gulfem Guler ◽  
David Haan ◽  
Yuhong Ning ◽  
Jeremy Ku ◽  
Erin McCarthy ◽  
...  

e21505 Background: Liquid biopsies are gaining prominence for not only cancer diagnosis but also patient monitoring. Mutational signals derived from cell-free DNA (cfDNA) show promise to assess response to cancer treatment, including immunotherapy. However, reliance of these methods on mutational data from tissue biopsies limit their applicability when a tumor biopsy is unavailable, or when mutational landscape of tumor changes under the selective pressures of cancer drug treatment. Epigenomic approaches have the potential to address these shortcomings. Methods: Blood draws were obtained from a cohort of non-small cell lung cancer (NSCLC) patients (n = 19) who went on to anti-PD1 treatment prior to therapy start and while on therapy. cfDNA was isolated from plasma and was subsequently processed to generate 5hmC genome-wide profiles. Results: We analyzed cfDNA from NSCLC patients undergoing anti-PD1 therapy to investigate whether immunotherapy response can be detected from plasma. Using a predictive model trained on lung cancer and non-cancer samples, we were able to detect changes in prediction scores in patient treated with immunotherapy that were consistent with RECIST. Patients with progressive disease (n = 3), determined by RECIST, had prediction scores that increased while they received treatment. On the other hand, majority of the patients that exhibited partial response to treatment (n = 12) had predictive scores that decreased with treatment, again consistent with RECIST. Furthermore, score changes consistent with RECIST was observed one cycle prior to the RECIST timepoint in all except one patient, where an extra blood draw after baseline was available (n = 7). Annotation of the regions that account for differential scoring identified enhancer, 5’UTR and promoter regions. Comparison of partial responders to patients with progressive disease revealed genes involved in metastasis, oncogenes and tumor suppressors that change in opposing directions between these patient groups, consistent with the underlying biology. Conclusions: Our results suggest that 5hmC profiles from cfDNA can be used to determine immunotherapy response in non-small cell lung cancer patients. Compared with mutation based liquid biopsy methods to assess response, epigenomics-based methods have the advantage of being agnostic to starting tumor mutations, and not relying on a mutational analysis from tumor biopsy. Future work will help determine applicability of this method to other kinds of therapies and cancer types.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e21578-e21578
Author(s):  
Feng Liang ◽  
Sisi Liu ◽  
Ya Jiang ◽  
Xiuxiu Xu ◽  
Qiuxiang Ou ◽  
...  

e21578 Background: Programmed cell death 1 (PD-L1) is the first FDA-approved predictive biomarker for non-small cell lung cancer (NSCLC) patients treated with PD-(L)1 blockade therapy. Herein, we aim to identify potential anti-PD-L1 treatment-related biomarkers through evaluating the correlation between the PD-L1 expression level, clinical characteristics, and the mutational profile of a large Chinese NSCLC cohort. Methods: Genomic profiling of tumor biopsies from a total of 808 Chinese NSCLC patients, including 651 adenocarcinomas (ADCs) and 157 squamous cell carcinomas (SCCs), was performed using next-generation sequencing by targeting 425 cancer-relevant genes. Immunohistochemical analysis was used to evaluate PD-L1 protein expression using PD-L1 antibodies including DAKO 22C3 ( N= 695) and DAKO 28-8 ( N= 113), respectively. Results: The PD-L1 positive ( > 1%) rate was 49.2% in ADCs and 52.9% in SCCs, respectively. PD-L1 expression (22C3) was associated with the male gender( p< 0.01) and lymph node metastasis ( p= 0.048) in ADCs but not in SCC patients. PD-L1 expression (22C3) was inversely correlated with KRAS wildtype ( p< 0.001) and EGFR exon 19 deletion( p< 0.01) in ADC, while it was negatively associated with TP53 oncogenic mutations ( p= 0.049) in SCC. Copy number variation analysis revealed that MDM2 amplification ( p= 0.027), 1q gain ( p= 0.012), and 5q deletion ( p< 0.01) negatively correlated with PD-L1 expression, whereas PD-L1 and PD-L2 amplification ( p< 0.001 and p< 0.0001) were positively associated with PD-L1 expression in ADCs. In SCCs, PD-L1 expression (22C3) was negatively associated with copy number gain in EGFR ( p= 0.040), MDM2 ( p= 0.044), 14q ( p= 0.032), and 20q ( p= 0.026), along with PTPRD loss (p = 0.015) and 19p deletion (p = 0.025). However, it was positively associated with 9p amplification ( p< 0.01) and 13q deletion ( p= 0.019). Plus, KIF5B- RET ( p= 0.006) appeared to be inversely related to the PD-L1 expression levels (22C3) in ADCs alone. In addition, these predicted biomarkers were used to delineate the receiver operating characteristic (ROC) calculation to discriminate between PD-L1 low and high (22C3, 50%) with an AUC score of 0.779. Lastly, PD-L1 expression (28-8) did not show significant correlation with any detected oncogenic mutations, but negatively correlated with NKX2-1 gain ( p= 0.0379) and 9q deletion ( p= 0.0379) in ADCs. Conclusions: This study revealed the correlation between PD-L1 protein expression, clinical features, and mutational traits in NSCLC patients, and provided a classifier for PD-L1 expression prediction.


2016 ◽  
Vol 5 (5) ◽  
pp. 511-516 ◽  
Author(s):  
Mónica Garzón ◽  
Sergi Villatoro ◽  
Cristina Teixidó ◽  
Clara Mayo ◽  
Alejandro Martínez ◽  
...  

2019 ◽  
Vol 47 (11) ◽  
pp. 5593-5600
Author(s):  
Lanxiang Ma ◽  
Jie Du ◽  
Yongjie Sui ◽  
Shuili Wang

Objectives Plasma free DNA is a promising new tumor biomarker, which may have applications in clinical diagnosis and treatment of non-small cell lung cancer (NSCLC). Methods Plasma free DNA was collected from 120 healthy controls and 116 patients with NSCLC before and after treatment. Results The mean plasma free DNA levels in 116 NSCLC patients (200.70 ± 88.54 ng/mL) were higher than those of 120 healthy controls (18.65 ± 6.30 ng/mL). Further analysis showed that the mean serum free DNA level in stage I/II NSCLC patients was 172.75 ± 72.87 ng/mL, significantly lower than that of stage III/IV patients (221.88 ± 93.86 ng/mL). Following surgery and effective chemotherapy, the plasma free DNA levels of NSCLC patients decreased significantly. Conclusions Through quantitation of plasma free DNA, this study established proof-of-concept for a highly sensitive and specific detection method, which can be used for diagnosis, prognosis and treatment monitoring in NSCLC patients.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A64-A64
Author(s):  
Kamil Sklodowski ◽  
Vito Dozio ◽  
Silvia Lopez-Lastra ◽  
Andrés Lanzós ◽  
Kristina Beeler ◽  
...  

BackgroundImmune checkpoint inhibitors have improved clinical responses and overall survival for patients with non-small cell lung cancer (NSCLC). However, the response is not equal and known NSCLC biomarkers are not sufficient in predicting therapy outcome. Deep proteomic analysis of NSCLC patient‘s plasma treated with anti-PD-1-blockade using a state-of-the-art data independent acquisition mass spectrometry (DIA-MS) is a powerful and unbiased way of identifying protein signatures associated with disease stage or response to treatment. However, to unravel these associations large-scale omics data should be analyzed with respect to available clinical information. To achieve this goal, we have used an approach previously applied by Uhlen et al., 20171 for transcriptomic datasets. In this approach survival data is used to set the most optimal thresholds for candidate biomarkers.Methods125 plasma samples were analyzed by capillary flow liquid chromatography coupled to DIA-MS. Data were extracted with latest SpectronautTM and proteins were quantified. Each recorded protein intensity was used as a threshold for two groups of samples for which Kaplan-Meier estimates were generated using ‘survival’2 package in R. Benjamini-Hochberg correction was applied and p-values with corresponding intensity cut-offs were extracted to generate panels of potential biomarkers.Results125 plasma samples (in total 75 baseline and 50 after 8-weeks treatment) from advanced NSCLC patients treated with an anti-PD-1 inhibitor following at least 1 prior line of treatment were analyzed. 727 unique proteins were quantified across all samples. Data analysis was performed separately for each line of treatment and treatment status resulting in more than 100’000 p-values. For each group, panels of proteins with best performance in separating progression free survivals were defined at FDR of 0.10, giving 64 unique proteins which were mapped to acute phase response, platelet degranulation and complement activation. Several of these proteins were listed in the Early Detection Research Network database of the National Cancer Institute, and one of them – LYPD3, was a potential therapeutic target in a preclinical study for NSCL treatment.3 Selected proteins were then used to cluster patients into cohorts that showed association with the response to therapy.ConclusionsDeep proteomic profiling of plasma samples using DIA-MS in conjunction with clinical outcome enables a holistic and stringent analysis of potential circulating biomarkers. Such analysis generates functional insights into the plasma proteome that enable deeper understanding and comprehensive integration of clinical data with proteomics markers at different disease stages and treatment phases.ReferencesUhlen M, Zhang C, Lee S, Sjöstedt E, Fagerberg L, Bidkhori G, Benfeitas R, Arif M, Liu Z, Edfors F, Sanli K, von Feilitzen K, Oksvold P, Lundberg E, Hober S, Nilsson P, Mattsson J, Schwenk J.Therneau TM, Grambsch PM. Modeling Survival Data: Extending the Cox Model. Springer. 2000, New York, ISBN 0-387-98784-3.Willuda J, Linden L, Lerchen H, Kopitz C, Stelte-Ludwig B, Pena C, Lange C, Golfier S, Kneip C, Carrigan P E, Mclean K, Schuhmacher J, von Ahsen O, Müller J, Dittmer F, Beier R, El Sheikh S, Tebbe J, Leder G, Apeler H, Jautelat R, Ziegelbauer K, Kreft B, Preclinical Antitumor Efficacy of BAY 1129980-a Novel Auristatin-Based Anti-C4.4A (LYPD3) Antibody-Drug Conjugate for the Treatment of Non-Small Cell Lung Cancer. Mol Caner Ther 2017;16(5):893–904


Sign in / Sign up

Export Citation Format

Share Document