Current Role and Future Potential of CSF ctDNA for the Diagnosis and Clinical Management of Pediatric Central Nervous System Tumors

Authors:
Alexandra M. Miller Department of Neurology, and
Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.

Search for other papers by Alexandra M. Miller in
Current site
Google Scholar
PubMed
Close
 MD, PhD
and
Matthias A. Karajannis Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.

Search for other papers by Matthias A. Karajannis in
Current site
Google Scholar
PubMed
Close
 MD, MS
Restricted access

Most pediatric central nervous system (CNS) tumors are located in eloquent anatomic areas, making surgical resection and, in some cases, even biopsy risky or impossible. This diagnostic predicament coupled with the move toward molecular classification for diagnosis has exposed an urgent need to develop a minimally invasive means to obtain diagnostic information. In non-CNS solid tumors, the detection of circulating tumor DNA (ctDNA) in plasma and other bodily fluids has been incorporated into routine practice and clinical trial design for selection of molecular targeted therapy and longitudinal monitoring. For primary CNS tumors, however, detection of ctDNA in plasma has been challenging. This is likely related at least in part to anatomic factors such as the blood–brain barrier. Due to the proximity of primary CNS tumors to the cerebrospinal fluid (CSF) space, our group and others have turned to CSF as a rich alternative source of ctDNA. Although multiple studies at this time have demonstrated the feasibility of CSF ctDNA detection across multiple types of pediatric CNS tumors, the optimal role and utility of CSF ctDNA in the clinical setting has not been established. This review discusses the work-to-date on CSF ctDNA liquid biopsy in pediatric CNS tumors and the associated technical challenges, and reviews the promising opportunities that lie ahead for integration of CSF ctDNA liquid biopsy into clinical care and clinical trial design.

Submitted August 11, 2022; final revision received October 26, 2022; accepted for publication October 28, 2022.

Disclosures: Dr. Karajannis has disclosed receiving consulting and personal fees from AstraZeneca, Bayer, CereXis, QED Therapeutics, and Recursion Pharma. Dr. Miller has disclosed having no financial interests, arrangements, affiliations, or commercial interests with the manufacturers of any products discussed in this article or their competitors.

Funding: Research reported in this publication was supported in part by the NCI of the NIH under Core Grant number P30 CA008748.

Disclaimer: The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Correspondence: Matthias A. Karajannis, MD, MS, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065. Email: karajanm@mskcc.org
  • Collapse
  • Expand
  • 1.

    Louis DN, Perry A, Wesseling P, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro-oncol 2021;23:12311251.

  • 2.

    Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24.

  • 3.

    Newman AM, Bratman SV, To J, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med 2014;20:548554.

  • 4.

    Wang Y, Springer S, Zhang M, et al. Detection of tumor-derived DNA in cerebrospinal fluid of patients with primary tumors of the brain and spinal cord. Proc Natl Acad Sci USA 2015;112:97049709.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5.

    Pentsova EI, Shah RH, Tang J, et al. Evaluating cancer of the central nervous system through next-generation sequencing of cerebrospinal fluid. J Clin Oncol 2016;34:24042415.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6.

    Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446451.

  • 7.

    Mathios D, Johansen JS, Cristiano S, et al. Detection and characterization of lung cancer using cell-free DNA fragmentomes. Nat Commun 2021;12:5060.

  • 8.

    Hellmann MD, Nabet BY, Rizvi H, et al. Circulating tumor DNA analysis to assess risk of progression after long-term response to PD-(L)1 blockade in NSCLC. Clin Cancer Res 2020;26:28492858.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Smyth LM, Reichel JB, Tang J, et al. Utility of serial cfDNA NGS for prospective genomic analysis of patients on a phase I basket study. JCO Precis Oncol 2021;5:PO.20.00184.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Devarakonda S, Sankararaman S, Herzog BH, et al. Circulating tumor DNA profiling in small-cell lung cancer identifies potentially targetable alterations. Clin Cancer Res 2019;25:61196126.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11.

    Heitzer E, van den Broek D, Denis MG, et al. Recommendations for a practical implementation of circulating tumor DNA mutation testing in metastatic non-small-cell lung cancer. ESMO Open 2022;7:100399.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Miller AM, Shah RH, Pentsova EI, et al. Tracking tumour evolution in glioma through liquid biopsies of cerebrospinal fluid. Nature 2019;565:654658.

  • 13.

    De Mattos-Arruda L, Mayor R, Ng CK, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun 2015;6:8839.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Cantor E, Wierzbicki K, Tarapore RS, et al. Serial H3K27M cell-free tumor DNA (cf-tDNA) tracking predicts ONC201 treatment response and progression in diffuse midline glioma. Neuro Oncol 2022;24:13661374.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15.

    Lui YY, Chik KW, Chiu RW, et al. Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation. Clin Chem 2002;48:421427.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Markus H, Chandrananda D, Moore E, et al. Refined characterization of circulating tumor DNA through biological feature integration. Sci Rep 2022;12:1928.

  • 17.

    Leon SA, Shapiro B, Sklaroff DM, et al. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 1977;37:646650.

  • 18.

    Mouliere F, Robert B, Arnau Peyrotte E, et al. High fragmentation characterizes tumour-derived circulating DNA. PLoS One 2011;6:e23418.

  • 19.

    Snyder MW, Kircher M, Hill AJ, et al. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 2016;164:5768.

  • 20.

    Cristiano S, Leal A, Phallen J, et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 2019;570:385389.

  • 21.

    Song Z, Wang W, Li M, et al. Cytological-negative pleural effusion can be an alternative liquid biopsy media for detection of EGFR mutation in NSCLC patients. Lung Cancer 2019;136:2329.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    de Almeida EF, Abdalla TE, Arrym TP, et al. Plasma and urine DNA levels are related to microscopic hematuria in patients with bladder urothelial carcinoma. Clin Biochem 2016;49:12741277.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Diehl F, Schmidt K, Durkee KH, et al. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology 2008;135:489498.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Kimura H, Fujiwara Y, Sone T, et al. EGFR mutation status in tumour-derived DNA from pleural effusion fluid is a practical basis for predicting the response to gefitinib. Br J Cancer 2006;95:13901395.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Panditharatna E, Kilburn LB, Aboian MS, et al. Clinically relevant and minimally invasive tumor surveillance of pediatric diffuse midline gliomas using patient-derived liquid biopsy. Clin Cancer Res 2018;24:58505859.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Liu APY, Smith KS, Kumar R, et al. Serial assessment of measurable residual disease in medulloblastoma liquid biopsies. Cancer Cell 2021;39:15191530.e4.

  • 27.

    Wang Y, Springer S, Mulvey CL, et al. Detection of somatic mutations and HPV in the saliva and plasma of patients with head and neck squamous cell carcinomas. Sci Transl Med 2015;7:293ra104.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Pan W, Gu W, Nagpal S, et al. Brain tumor mutations detected in cerebral spinal fluid. Clin Chem 2015;61:514522.

  • 29.

    Izquierdo E, Proszek P, Pericoli G, et al. Droplet digital PCR-based detection of circulating tumor DNA from pediatric high grade and diffuse midline glioma patients. Neurooncol Adv 2021;3:vdab013.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Fujioka Y, Hata N, Akagi Y, et al. Molecular diagnosis of diffuse glioma using a chip-based digital PCR system to analyze IDH, TERT, and H3 mutations in the cerebrospinal fluid. J Neurooncol 2021;152:4754.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Mouliere F, Mair R, Chandrananda D, et al. Detection of cell-free DNA fragmentation and copy number alterations in cerebrospinal fluid from glioma patients. EMBO Mol Med 2018;10:e9323.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 32.

    Miller AM, Szalontay L, Bouvier N, et al. Next-generation sequencing of cerebrospinal fluid for clinical molecular diagnostics in pediatric, adolescent and young adult (AYA) brain tumor patients. J Neurooncol 2022;24:17631772.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Huang TY, Piunti A, Lulla RR, et al. Detection of histone H3 mutations in cerebrospinal fluid-derived tumor DNA from children with diffuse midline glioma. Acta Neuropathol Commun 2017;5:28.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34.

    Martínez-Ricarte F, Mayor R, Martínez-Sáez E, et al. Molecular diagnosis of diffuse gliomas through sequencing of cell-free circulating tumor DNA from cerebrospinal fluid. Clin Cancer Res 2018;24:28122819.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35.

    Stallard S, Savelieff MG, Wierzbicki K, et al. CSF H3F3A K27M circulating tumor DNA copy number quantifies tumor growth and in vitro treatment response. Acta Neuropathol Commun 2018;6:80.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Li D, Bonner ER, Wierzbicki K, et al. Standardization of the liquid biopsy for pediatric diffuse midline glioma using ddPCR. Sci Rep 2021;11:5098.

  • 37.

    Pan C, Diplas BH, Chen X, et al. Molecular profiling of tumors of the brainstem by sequencing of CSF-derived circulating tumor DNA. Acta Neuropathol 2019;137:297306.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Hayward RM, Patronas N, Baker EH, et al. Inter-observer variability in the measurement of diffuse intrinsic pontine gliomas. J Neurooncol 2008;90:5761.

  • 39.

    Escudero L, Llort A, Arias A, et al. Circulating tumour DNA from the cerebrospinal fluid allows the characterisation and monitoring of medulloblastoma. Nat Commun 2020;11:5376.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40.

    Wu X, Northcott PA, Dubuc A, et al. Clonal selection drives genetic divergence of metastatic medulloblastoma. Nature 2012;482:529533.

  • 41.

    Bruzek AK, Ravi K, Muruganand A, et al. Electronic DNA analysis of CSF cell-free tumor DNA to quantify multi-gene molecular response in pediatric high-grade glioma. Clin Cancer Res 2020;26:62666276.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Li J, Zhao S, Lee M, et al. Reliable tumor detection by whole-genome methylation sequencing of cell-free DNA in cerebrospinal fluid of pediatric medulloblastoma. Sci Adv 2020;6:eabb5427.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43.

    Peneder P, Stütz AM, Surdez D, et al. Multimodal analysis of cell-free DNA whole-genome sequencing for pediatric cancers with low mutational burden. Nat Commun 2021;12:3230.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Zviran A, Schulman RC, Shah M, et al. Genome-wide cell-free DNA mutational integration enables ultra-sensitive cancer monitoring. Nat Med 2020;26:11141124.

  • 45.

    Isaksson S, George AM, Jönsson M, et al. Pre-operative plasma cell-free circulating tumor DNA and serum protein tumor markers as predictors of lung adenocarcinoma recurrence. Acta Oncol 2019;58:10791086.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation

Metrics

All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 1981 738 8
PDF Downloads 1104 244 9
EPUB Downloads 0 0 0