Circulating Tumor DNA as a Biomarker in Patients With Stage III and IV Wilms Tumor: Analysis From a Children's Oncology Group Trial, AREN0533

Laura M Madanat-Harjuoja, Lindsay A Renfro, Kelly Klega, Brett Tornwall, Aaron R Thorner, Anwesha Nag, David Dix, Jeffrey S Dome, Lisa R Diller, Conrad V Fernandez, Elizabeth A Mullen, Brian D Crompton, Laura M Madanat-Harjuoja, Lindsay A Renfro, Kelly Klega, Brett Tornwall, Aaron R Thorner, Anwesha Nag, David Dix, Jeffrey S Dome, Lisa R Diller, Conrad V Fernandez, Elizabeth A Mullen, Brian D Crompton

Abstract

Purpose: The utility of circulating tumor DNA (ctDNA) analyses has not been established in the risk stratification of Wilms tumor (WT). We evaluated the detection of ctDNA and selected risk markers in the serum and urine of patients with WT and compared findings with those of matched diagnostic tumor samples.

Patients and methods: Fifty of 395 children with stage III or IV WT enrolled on Children's Oncology Group trial AREN0533 had banked pretreatment serum, urine, and tumor available. Next-generation sequencing was used to detect ctDNA. Copy-number changes in 1q, 16q, and 1p, and single-nucleotide variants in serum and urine were compared with tumor biopsy data. Event-free survival (EFS) was compared between patients with and without ctDNA detection.

Results: ctDNA was detected in the serum of 41/50 (82%) and in the urine in 13/50 (26%) patients. Agreement between serum ctDNA detection and tumor sequencing results was as follows: 77% for 1q gain, 88% for 16q deletions, and 70% for 1p deletions, with ĸ-coefficients of 0.56, 0.74, and 0.29, respectively. Sequencing also demonstrated that single-nucleotide variants detected in tumors could be identified in the ctDNA. There was a trend toward worse EFS in patients with ctDNA detected in the serum (4-year EFS 80% v 100%, P = .14).

Conclusion: ctDNA demonstrates promise as an easily accessible prognostic biomarker with potential to detect tumor heterogeneity. The observed trend toward more favorable outcome in patients with undetectable ctDNA requires validation. ctDNA profiling should be further explored as a noninvasive diagnostic and prognostic tool in the risk-adapted treatment of patients with WT.

Trial registration: ClinicalTrials.gov NCT00379340.

Figures

FIG 1.
FIG 1.
Detection of ctDNA by identification of copy-number variants in serum and urine of patients with WT. (A) Violin plot of ctDNA levels in serum and urine samples restricted to cases with ctDNA above the limit of detection by ULP-WGS. Dashed vertical line within each violin plot indicates the median and the dotted lines indicate the boundaries between the first and second quartiles and the third and fourth quartiles. Probability densities are reflected by the upper and lower borders of the violin plot. Individual ctDNA data are indicated by black dots. (B) Summary of CNAs in chromosomes 1p, 1q, and 16q by case across sample types. Tissue is indicated at the top of the plot, data type indicated in the column label, and vertical black bars indicate the percent of total evaluable samples containing the variant (or with detectable ctDNA). Red blocks indicate copy gains, blue indicates copy losses, white indicates that a variant was not detected, dark gray indicates an unevaluable sample (no tissue or insufficient DNA for sequencing), and light gray indicates that no ctDNA was detectable in the sample. Percent ctDNA content indicated by number with higher ctDNA content shaded in dark teal and lower ctDNA by light teal. Cases plotted in (C-E) include “a” in the patient identifier. (C-E) Genome-wide plots represent the log2 ratio for each data point with blue data equivalent to two copies of the genomic location, teal equal to copy-number loss, and red equal to copy-number gains. Chromosomal segmental medians are also plotted as horizontal lines with light teal segments representing likely subclonal events. Arrows indicate CNAs that are not present in all matched samples from the patient. Case identifiers are present at the top of the panels. CNA, copy-number alteration; ctDNA, circulating tumor DNA; WT, Wilms tumor.
FIG 2.
FIG 2.
SNVs identified in tumors and matched ctDNA samples. Plotted are somatic variants identified in genes frequently mutated in WTs. Open circles indicate variants identified in tumors, red circles indicate variants identified in serum, and open triangles identified from urine. The size of the symbols reflects the relative allelic fraction of the identified events. Vertical bar plots reflect the fraction of ctDNA identified in the serum (red bars) and urine (open bars with black outline). Horizontal blue bars summarize the number of variants identified in the indicated gene. aA case with two variants in the DICER1 gene identified in both the tumor and the serum. ctDNA, circulating tumor DNA; SNV, single-nucleotide variant; WT, Wilms tumor.
FIG 3.
FIG 3.
EFS and OS in patients with and without detectable ctDNA in the serum: (A) EFS and (B) OS by ctDNA detection in the serum of patients with stage III and IV WT. ctDNA, circulating tumor DNA; EFS, event-free survival; OS, overall survival; WT, Wilms tumor.

References

    1. Murphy WM, Grignon DJ, Perlman EJ: Kidney Tumors in Children, Atlas of Tumor Pathology. Washington DC, Armed Forces Institute of Pathology, 2004, pp 57-64
    1. Malogolowkin M, Cotton CA, Green DM, et al. : Treatment of Wilms tumor relapsing after initial treatment with vincristine, actinomycin D, and doxorubicin. A report from the National Wilms Tumor Study Group. Pediatr Blood Cancer 50:236-241, 2008
    1. Spreafico F, Pritchard Jones K, Malogolowkin MH, et al. : Treatment of relapsed Wilms tumors: Lessons learned. Expert Rev Anticancer Ther 9:1807-1815, 2009
    1. Mullen EA, Chi YY, Hibbitts E, et al. : Impact of surveillance imaging modality on survival after recurrence in patients with favorable-histology Wilms tumor: A report from the Children's Oncology Group. J Clin Oncol 36:3396-3403, 2018
    1. Gratias EJ, Dome JS, Jennings LJ, et al. : Association of chromosome 1q gain with inferior survival in favorable-histology Wilms tumor: A report from the Children's Oncology Group. J Clin Oncol 34:3189-3194, 2016
    1. Grundy PE, Breslow NE, Li S, et al. : Loss of heterozygosity for chromosomes 1p and 16q is an adverse prognostic factor in favorable-histology Wilms tumor: A report from the National Wilms Tumor Study Group. J Clin Oncol 23:7312-7321, 2005
    1. Dix DB, Fernandez CV, Chi YY, et al. : Augmentation of therapy for combined loss of heterozygosity 1p and 16q in favorable histology Wilms tumor: A Children's Oncology Group AREN0532 and AREN0533 study report. J Clin Oncol 37:2769-2777, 2019
    1. Bettegowda C, Sausen M, Leary RJ, et al. : Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 6:224ra24, 2014
    1. Abbou SD, Shulman DS, DuBois SG, et al. : Assessment of circulating tumor DNA in pediatric solid tumors: The promise of liquid biopsies. Pediatr Blood Cancer 66:e27595, 2019
    1. Weiser DA, West-Szymanski DC, Fraint E, et al. : Progress toward liquid biopsies in pediatric solid tumors. Cancer Metastasis Rev 38:553-571, 2019
    1. Marcuello M, Vymetalkova V, Neves RPL, et al. : Circulating biomarkers for early detection and clinical management of colorectal cancer. Mol Aspects Med 69:107-122, 2019
    1. Chabon JJ, Hamilton EG, Kurtz DM, et al. : Integrating genomic features for non-invasive early lung cancer detection. Nature 580:245-251, 2020
    1. Alimirzaie S, Bagherzadeh M, Akbari MR: Liquid biopsy in breast cancer: A comprehensive review. Clin Genet 95:643-660, 2019
    1. Haber DA, Velculescu VE: Blood-based analyses of cancer: Circulating tumor cells and circulating tumor DNA. Cancer Discov 4:650-661, 2014
    1. Chabon JJ, Simmons AD, Lovejoy AF, et al. : Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun 7:11815, 2016
    1. Corcoran RB, Chabner BA: Application of cell-free DNA analysis to cancer treatment. N Engl J Med 379:1754-1765, 2018
    1. Dawson S, Tsui D, Murtaza M, et al. : Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med 368:1199-1209, 2013
    1. Bosse KR, Buongervino S, Lane M, Hyman A, et al. : Serial profiling of ctDNA identifies clinically actionable genomic evolution in high-risk neuroblastoma. Cancer Res, 79, 2019 (suppl 13; abstr 3105)
    1. Charlton J, Williams RD, Weeks M, et al. : Methylome analysis identifies a Wilms tumor epigenetic biomarker detectable in blood. Genome Biol 15:434, 2014
    1. Chicard M, Colmet-Daage L, Clement N, et al. : Whole-exome sequencing of cell-free DNA reveals temporo-spatial heterogeneity and identifies treatment-resistant clones in neuroblastoma. Clin Cancer Res 24:939-949, 2018
    1. Jimenez I, Chicard M, Colmet-Daage L, et al. : Circulating tumor DNA analysis enables molecular characterization of pediatric renal tumors at diagnosis. Int J Cancer 144:68-79, 2019
    1. Klega K, Imamovic-Tuco A, Ha G, et al. : Detection of somatic structural variants enables quantification and characterization of circulating tumor DNA in children with solid tumors. JCO Precis Oncol
    1. Miguez ACK, Barros BDF, de Souza JES, et al. : Assessment of somatic mutations in urine and plasma of Wilms tumor patients. Cancer Med 9:5948-5959, 2020
    1. Shulman DS, Klega K, Imamovic-Tuco A, et al. : Detection of circulating tumour DNA is associated with inferior outcomes in Ewing sarcoma and osteosarcoma: A report from the Children's Oncology Group. Br J Cancer 119:615-621, 2018
    1. Treger TD, Chagtai T, Butcher R, et al. : Somatic TP53 mutations are detectable in circulating tumor DNA from children with anaplastic Wilms tumors. Transl Oncol 11:1301-1306, 2018
    1. Van Roy N, Van Der Linden M, Menten B, et al. : Shallow whole genome sequencing on circulating cell-free DNA allows reliable noninvasive copy-number profiling in neuroblastoma patients. Clin Cancer Res 23:6305-6314, 2017
    1. Dix DB, Seibel NL, Chi YY, et al. : Treatment of stage IV favorable histology Wilms tumor with lung metastases: A report from the Children's Oncology Group AREN0533 study. J Clin Oncol 36:1564-1570, 2018
    1. Adalsteinsson VA, Ha G, Freeman SS, et al. : Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun 8:1324, 2017
    1. Garcia EP, Minkovsky A, Jia Y, et al. : Validation of OncoPanel: A targeted next-generation sequencing assay for the detection of somatic variants in cancer. Arch Pathol Lab Med 141:751-758, 2017
    1. Sholl LM, Do K, Shivdasani P, et al. : Institutional implementation of clinical tumor profiling on an unselected cancer population. JCI Insight 1:e87062, 2016
    1. Landis JR, Koch GG: The measurement of observer agreement for categorical data. Biometrics 33:159-174, 1977
    1. Cajaiba MM, Dyer LM, Geller JI, et al. : The classification of pediatric and young adult renal cell carcinomas registered on the Children's Oncology Group (COG) protocol AREN03B2 after focused genetic testing. Cancer 124:3381-3389, 2018
    1. Gadd S, Huff V, Walz AL, et al. : A Children's Oncology Group and TARGET initiative exploring the genetic landscape of Wilms tumor. Nat Genet 49:1487-1494, 2017
    1. Abbosh C, Birkbak NJ, Wilson GA, et al. : Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 545:446-451, 2017
    1. Spina V, Bruscaggin A, Cuccaro A, et al. : Circulating tumor DNA reveals genetics, clonal evolution, and residual disease in classical Hodgkin lymphoma. Blood 131:2413-2425, 2018
    1. Alix-Panabieres C, Pantel K: Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discov 6:479-491, 2016
    1. Bardelli A, Pantel K: Liquid biopsies, what we do not know (yet). Cancer Cell 31:172-179, 2017
    1. Casbon JA, Osborne RJ, Brenner S, et al. : A method for counting PCR template molecules with application to next-generation sequencing. Nucleic Acids Res 39:e81, 2011
    1. Kang Q, Henry NL, Paoletti C, et al. : Comparative analysis of circulating tumor DNA stability in K3EDTA, Streck, and CellSave blood collection tubes. Clin Biochem 49:1354-1360, 2016
    1. van Dessel LF, Beije N, Helmijr JC, et al. : Application of circulating tumor DNA in prospective clinical oncology trials—Standardization of preanalytical conditions. Mol Oncol 11:295-304, 2017
    1. Merker JD, Oxnard GR, Compton C, et al. : Circulating tumor DNA analysis in patients with cancer: American Society of Clinical Oncology and College of American Pathologists joint review. Arch Pathol Lab Med 142:1242-1253, 2018
    1. Garcia-Murillas I, Schiavon G, Weigelt B, et al. : Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med 7:302ra133, 2015
    1. Krumbholz M, Eiblwieser J, Ranft A, et al. : Quantification of translocation-specific ctDNA provides an integrating parameter for early assessment of treatment response and risk stratification in Ewing sarcoma. Clin Cancer Res 27:5922-5930, 2021
    1. Stankunaite R, George SL, Gallagher L, et al. : Circulating tumour DNA sequencing to determine therapeutic response and identify tumour heterogeneity in patients with paediatric solid tumours. Eur J Cancer 162:209-220, 2022

Source: PubMed

3
Sottoscrivi