Longitudinal Copy-Number Alteration Analysis in Plasma Cell-Free DNA of Neuroendocrine Neoplasms is a Novel Specific Biomarker for Diagnosis, Prognosis, and Follow-up

Gitta Boons, Timon Vandamme, Laura Mariën, Willem Lybaert, Geert Roeyen, Tim Rondou, Konstantinos Papadimitriou, Katrien Janssens, Bart Op de Beeck, Marc Simoens, Wim Demey, Isabel Dero, Guy Van Camp, Marc Peeters, Ken Op de Beeck, Gitta Boons, Timon Vandamme, Laura Mariën, Willem Lybaert, Geert Roeyen, Tim Rondou, Konstantinos Papadimitriou, Katrien Janssens, Bart Op de Beeck, Marc Simoens, Wim Demey, Isabel Dero, Guy Van Camp, Marc Peeters, Ken Op de Beeck

Abstract

Purpose: As noninvasive biomarkers are an important unmet need for neuroendocrine neoplasms (NEN), biomarker potential of genome-wide molecular profiling of plasma cell-free DNA (cfDNA) was prospectively studied in patients with NEN.

Experimental design: Longitudinal plasma samples were collected from patients with well-differentiated, metastatic gastroenteropancreatic and lung NEN. cfDNA was subjected to shallow whole-genome sequencing to detect genome-wide copy-number alterations (CNA) and estimate circulating tumor DNA (ctDNA) fraction, and correlated to clinicopathologic and survival data. To differentiate pancreatic NENs (PNEN) from pancreatic adenocarcinomas (PAAD) using liquid biopsies, a classification model was trained using tissue-based CNAs and validated in cfDNA.

Results: One hundred and ninety-five cfDNA samples from 43 patients with NEN were compared with healthy control cfDNA (N = 100). Plasma samples from patients with PNEN (N = 21) were used for comparison with publicly available PNEN tissue (N = 98), PAAD tissue (N = 109), and PAAD cfDNA (N = 96). Thirty percent of the NEN cfDNA samples contained ctDNA and 44% of the patients had at least one ctDNA-positive (ctDNA+) sample. CNAs detected in cfDNA were highly specific for NENs and the classification model could distinguish PAAD and PNEN cfDNA samples with a sensitivity, specificity, and AUC of 62%, 86%, and 79%, respectively. ctDNA-positivity was associated with higher World Health Organization (WHO) grade, primary tumor location, and higher chromogranin A and neuron-specific enolase values. Overall survival was significantly worse for ctDNA+ patients and increased ctDNA fractions were associated with poorer progression-free survival.

Conclusions: Sequential genome-wide profiling of plasma cfDNA is a novel, noninvasive biomarker with high specificity for diagnosis, prognosis, and follow-up in metastatic NENs.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
OS analysis for ctDNA+ (blue curve) and ctDNA− (red curve) patients with significance values, in the whole cohort (A) and the subset of PNENs (B). NR, not reached.
Figure 2.
Figure 2.
Frequency plots for detected CNAs in cfDNA of patients with PNEN (N = 13; A), and tumor tissue of patients with PNEN (N = 98; B). Gains are indicated in green and losses in red.
Figure 3.
Figure 3.
Evolution of CNA profiles determined by ichorCNA between sample at inclusion (top) and last sample (bottom) for patient P01 (A) and P05 (B). Samples from P01 were taken 8 months apart and from P05 3 weeks apart.
Figure 4.
Figure 4.
Illustration of the effect of tumor fraction on PFS probability (red curve, with 95% CI in blue) for P01, according to our joint model, whereby all tumor fraction measurements (*) after treatment initiation were consecutively added (A–D). When a higher tumor fraction value was added to the measurements, the survival probability decreased.
Figure 5.
Figure 5.
Tumor fraction evolution during disease course (black curve) of patients P06 (A), P17 (B), and P04 (C). All patients started with everolimus and SSA treatment on day 0. In addition, the DSUM was indicated (gray area), as well as investigator-assessed imaging results (triangle) and treatment breaks or switches (line or box). PD, progressive disease.

References

    1. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, et al. . Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol 2017;3:1335–42.
    1. Rindi G, Klimstra DS, Abedi-Ardekani B, Asa SL, Bosman FT, Brambilla E, et al. . A common classification framework for neuroendocrine neoplasms: an International Agency for Research on Cancer (IARC) and World Health Organization (WHO) expert consensus proposal. Mod Pathol 2018;31:1770–86.
    1. Marotta V, Zatelli MC, Sciammarella C, Ambrosio MR, Bondanelli M, Colao A, et al. . Chromogranin A as circulating marker for diagnosis and management of neuroendocrine neoplasms: more flaws than fame. Endocr Relat Cancer 2018;25:R11–29.
    1. Hofland J, Zandee WT, de Herder WW. Role of biomarker tests for diagnosis of neuroendocrine tumours. Nat Rev Endocrinol 2018;14:656–69.
    1. van Adrichem RCS, Kamp K, Vandamme T, Peeters M, Feelders RA, de Herder WW. Serum neuron-specific enolase level is an independent predictor of overall survival in patients with gastroenteropancreatic neuroendocrine tumors. Ann Oncol 2015;27:746–7.
    1. Oberg K, Modlin IM, De Herder W, Pavel M, Klimstra D, Frilling A, et al. . Consensus on biomarkers for neuroendocrine tumour disease. Lancet Oncol 2015;16:e435–e46.
    1. Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. . Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer 2017;17:223–38.
    1. Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA, et al. . Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun 2017;8:1324.
    1. Raman L, Van der Linden M, Van der Eecken K, Vermaelen K, Demedts I, Surmont V, et al. . Shallow whole-genome sequencing of plasma cell-free DNA accurately differentiates small from non-small cell lung carcinoma. Genome Med 2020;12:35.
    1. Boons G, Vandamme T, Peeters M, Beyens M, Driessen A, Janssens K, et al. . Cell-free DNA from metastatic pancreatic neuroendocrine tumor patients contains tumor-specific mutations and copy number variations. Front Oncol 2018;8:467.
    1. Zakka K, Nagy R, Drusbosky L, Akce M, Wu C, Alese OB, et al. . Blood-based next-generation sequencing analysis of neuroendocrine neoplasms. Oncotarget 2020;11:1749–57.
    1. Boons G, Vandamme T, Peeters M, Van Camp G, Op de Beeck K. Clinical applications of (epi)genetics in gastroenteropancreatic neuroendocrine neoplasms: Moving towards liquid biopsies. Rev Endocr Metab Disord 2019;20:333–51.
    1. Lawrence B, Blenkiron C, Parker K, Tsai P, Fitzgerald S, Shields P, et al. . Recurrent loss of heterozygosity correlates with clinical outcome in pancreatic neuroendocrine cancer. NPJ Genom Med 2018;3:18.
    1. Tang LH, Basturk O, Sue JJ, Klimstra DS. A practical approach to the classification of WHO grade 3 (G3) well differentiated neuroendocrine tumor (WD-NET) and poorly differentiated neuroendocrine carcinoma (PD-NEC) of the pancreas. Am J Surg Pathol 2016;40:1192–202.
    1. Boons G, Vandamme T, Ibrahim J, Roeyen G, Driessen A, Peeters D, et al. . PDX1 DNA methylation distinguishes two subtypes of pancreatic neuroendocrine neoplasms with a different prognosis. Cancers 2020;12:1461.
    1. Banck MS, Kanwar R, Kulkarni AA, Boora GK, Metge F, Kipp BR, et al. . The genomic landscape of small intestine neuroendocrine tumors. J Clin Invest 2013;123:2502–8.
    1. Scarpa A, Chang DK, Nones K, Corbo V, Patch A-M, Bailey P, et al. . Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 2017;543:65–71.
    1. Simbolo M, Mafficini A, Sikora KO, Fassan M, Barbi S, Corbo V, et al. . Lung neuroendocrine tumours: deep sequencing of the four World Health Organization histotypes reveals chromatin-remodelling genes as major players and a prognostic role for TERT, RB1, MEN1 and KMT2D. J Pathol 2017;241:488–500.
    1. Wei T, Zhang J, Li J, Chen Q, Zhi X, Tao W, et al. . Genome-wide profiling of circulating tumor DNA depicts landscape of copy number alterations in pancreatic cancer with liver metastasis. Mol Oncol 2020;14:1966–77.
    1. Goethals S, De Wilde A, Lesage K, Smits E, Pauwels P, Peeters M. Tumorbank@uza: A collection of tissue, fluid samples and associated data of oncology patients for the use in translational research. Open J Bioresources 2018;5:4.
    1. Hudson TJ, Anderson W, Artez A, Barker AD, Bell C, Bernabé RR, et al. . International network of cancer genome projects. Nature 2010;464:993–8.
    1. Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv: 2013.
    1. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. . The sequence alignment/Map format and SAMtools. Bioinformatics 2009;25:2078–9.
    1. Ha G, Roth A, Lai D, Bashashati A, Ding J, Goya R, et al. . Integrative analysis of genome-wide loss of heterozygosity and monoallelic expression at nucleotide resolution reveals disrupted pathways in triple-negative breast cancer. Genome Res 2012;22:1995–2007.
    1. Sing T, Sander O, Beerenwinkel N, Lengauer T. ROCR: visualizing classifier performance in R. Bioinformatics 2005;21:3940–1.
    1. Rizopoulos D. JM: An R package for the joint modelling of longitudinal and time-to-event data. J Stat Softwa 2010;35:1–33.
    1. Gao B, Baudis M. Signatures of discriminative copy number aberrations in 31 cancer subtypes. bioRxiv 2021Apr 15 [epub ahead of print].
    1. Bronkhorst AJ, Ungerer V, Holdenrieder S. The emerging role of cell-free DNA as a molecular marker for cancer management. Biomol Detect Quantific 2019;17:100087.
    1. Pessoa LS, Heringer M, Ferrer VP. ctDNA as a cancer biomarker: A broad overview. Crit Rev Oncol Hematol 2020;155:103109.
    1. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. . Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24.
    1. Thierry AR, El Messaoudi S, Gahan PB, Anker P, Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev 2016;35:347–76.
    1. Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, Constantin T, Salari R, et al. . Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446–51.
    1. Öberg K, Califano A, Strosberg JR, Ma S, Pape U, Bodei L, et al. . A meta-analysis of the accuracy of a neuroendocrine tumor mRNA genomic biomarker (NETest) in blood. Ann Oncol 2020;31:202–12.
    1. Ococks E, Frankell AM, Soler NM, Grehan N, Northrop A, Coles H, et al. . Longitudinal tracking of 97 esophageal adenocarcinomas using liquid biopsy sampling. Ann Oncol 2021;32:522–32.
    1. Xie M, Lu C, Wang J, McLellan MD, Johnson KJ, Wendl MC, et al. . Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014;20:1472–8.
    1. Vandamme T, Beyens M, Boons G, Schepers A, Kamp K, Biermann K, et al. . Hotspot DAXX, PTCH2 and CYFIP2 mutations in pancreatic neuroendocrine neoplasms. Endocr Relat Cancer 2019;26:1–12.
    1. Mouliere F, Chandrananda D, Piskorz AM, Moore EK, Morris J, Ahlborn LB, et al. . Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med 2018;10:eaat4921.
    1. Neveling K, Mensenkamp AR, Derks R, Kwint M, Ouchene H, Steehouwer M, et al. . BRCA testing by single-molecule inversion probes. Clin Chem 2017;63:503–12.
    1. Steeghs EMP, Kroeze LI, Tops BBJ, van Kempen LC, Elst AT, Kastner-van Raaij AWM, et al. . Comprehensive routine diagnostic screening to identify predictive mutations, gene amplifications, and microsatellite instability in FFPE tumor material. BMC Cancer 2020;20:291.
    1. Jensen RT, Bodei L, Capdevila J, Couvelard A, Falconi M, Glasberg S, et al. . Unmet needs in functional and nonfunctional pancreatic neuroendocrine neoplasms. Neuroendocrinology 2019;108:26–36.
    1. Botling J, Lamarca A, Bajic D, Norlén O, Lönngren V, Kjaer J, et al. . High-grade progression confers poor survival in pancreatic neuroendocrine tumors. Neuroendocrinology 2020;110:891–8.
    1. Alexandraki KI, Tsoli M, Kyriakopoulos G, Angelousi A, Nikolopoulos G, Kolomodi D, et al. . Current concepts in the diagnosis and management of neuroendocrine neoplasms of unknown primary origin. Minerva Endocrinol 2019;44:378–86.
    1. Vandamme T, Beyens M, Op de Beeck K, Dogan F, van Koetsveld PM, Pauwels P, et al. . Long-term acquired everolimus resistance in pancreatic neuroendocrine tumours can be overcome with novel PI3K-AKT-mTOR inhibitors. Br J Cancer 2016;114:650–8.
    1. Beyens M, Vandamme T, Peeters M, Van Camp G, Op de Beeck K. Resistance to targeted treatment of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer 2019;26:R109.

Source: PubMed

3
Subscribe