Article

Brief Communication

Ann Lab Med 2025; 45(1): 90-95

Published online September 30, 2024 https://doi.org/10.3343/alm.2024.0257

Copyright © Korean Society for Laboratory Medicine.

Feasibility of Circulating Tumor DNA Detection in the Cerebrospinal Fluid of Patients With Central Nervous System Involvement in Large B-Cell Lymphoma

Seok Jin Kim , M.D., Ph.D.1,2*, Jin Ju Kim , M.D., Ph.D.3*, Mi Ri Park , B.S.4, Bon Park , B.S.2, Kyung Ju Ryu , Ph.D.2, Sang Eun Yoon , M.D., Ph.D.1, Won Seog Kim , M.D., Ph.D.1, Saeam Shin , M.D., Ph.D.4, and Seung-Tae Lee, M.D., Ph.D.4,5

1Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; 2Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Korea; 3Department of Laboratory Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea; 4Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea; 5Dxome Co., Ltd., Seongnam, Korea

Correspondence to: Saeam Shin, M.D., Ph.D.
Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
E-mail: saeam0304@yuhs.ac

* These authors contributed equally to this study as co-first authors.

Received: May 18, 2024; Revised: June 30, 2024; Accepted: August 21, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

We explored the utility of cerebrospinal fluid (CSF) circulating tumor DNA (ctDNA) sequencing as a noninvasive diagnostic tool for detecting central nervous system (CNS) involvement in patients with diffuse large B-cell lymphoma (DLBCL). Secondary CNS involvement in DLBCL, although rare (~5% of cases), presents diagnostic and prognostic challenges during systemic disease progression or relapse. Effective treatment is impeded by the blood–brain barrier. This was a prospective cohort study (Samsung Lymphoma Cohort Study III) involving 17 patients with confirmed CNS involvement. High-throughput sequencing was conducted using targeted gene panels designed to detect low-frequency variants and copy number alterations pertinent to lymphomas in ctDNA extracted from archived CSF samples. Despite challenges such as low DNA concentrations affecting library construction, the overall variant detection rate was 76%. Detected variants included those in genes commonly implicated in CNS lymphoma, such as MYD88. The study highlights the potential of CSF ctDNA sequencing to identify CNS involvement in DLBCL, providing a promising alternative to more invasive diagnostic methods such as brain biopsy, which are not always feasible. Further validation is necessary to establish the clinical utility of this method, which could significantly enhance the management and outcomes of DLBCL patients with suspected CNS involvement.

Keywords: Central nervous system, Cerebrospinal fluid, Circulating tumor DNA, Lymphoma, Relapse

Secondary central nervous system (CNS) involvement in diffuse large B-cell lymphoma (DLBCL), although relatively rare (occurring in ~5% of cases), is associated with a grim prognosis [1, 2]. It can manifest during the treatment of systemic DLBCL as a sign of disease progression, or as a relapse, with or without systemic involvement. Treatment strategies for secondary CNS involvement typically differ from those used for systemic DLBCL, primarily because of the need to penetrate the blood–brain barrier. High-dose methotrexate chemotherapy regimens are the mainstay of treatment because of their ability to penetrate the CNS [3]. However, most patients do not achieve long-lasting remission, leading to disappointing outcomes [4, 5]. Therefore, early detection and management of CNS involvement are crucial for improving patient outcomes [6]. However, brain biopsy is not always feasible, and conventional cerebrospinal fluid (CSF) analyses via cytopathology or flow cytometry and diagnostic magnetic resonance imaging (MRI) have shown suboptimal sensitivity and discriminative capacity to enable the diagnosis of CNS involvement without tissue confirmation. The development of methods that overcome these limitations and allow reliable noninvasive identification of CNS involvement would be transformative for the clinical care of patients with suspected secondary CNS involvement by DLBCL.

Liquid biopsy, which involves analyzing circulating tumor DNA (ctDNA) in the blood or body fluid, is revolutionizing cancer diagnosis and surveillance. ctDNA originating from tumor tissue or lysed circulating tumor cells offers a noninvasive method for monitoring cancer progression and treatment responses [7]. Advances in next-generation sequencing (NGS) have enabled the analysis of variants in ctDNA, allowing tumor genotyping using blood samples [8]. High-throughput ctDNA sequencing can provide comprehensive genetic information regarding the tumor, serving as a surrogate for sequencing the entire tumor genome. This approach holds promise for the identification of therapeutic targets and early detection of relapse or residual disease. We have shown that ctDNA can be detected in plasma from patients with various subtypes of B-, T- or NK-cell lymphomas, correlating with tumor volume and patient outcomes [9-12]. However, only a minority of patients with primary CNS B-cell lymphomas have detectable ctDNA in their plasma, possibly because of the blood–brain barrier [13]. Consequently, ctDNA from CSF has emerged as a promising biomarker for CNS involvement of B-cell lymphomas. In this study, we optimized a customized targeted sequencing approach to achieve ultrasensitive ctDNA profiling and investigated its potential for identifying CNS involvement without the need for biopsy.

We analyzed archived CSF samples collected between July 2020 and October 2022 from patients diagnosed as having large B-cell lymphoma who participated in a prospective cohort study (Samsung Lymphoma Cohort Study III, Institutional Review Board of Samsung Medical Center, File No. SMC 2017-12-068; ClinicalTrials.gov Identifier: NCT03117036). All patients were pathologically confirmed as having large B-cell lymphoma using immunohistochemistry [14]. As we monitored the patients enrolled, we identified those suspected of CNS involvement either based on clinical neurological symptoms and/or manifestations or radiologically by detecting parenchymal or leptomeningeal abnormalities in the brain or spine MRI scans. From these patients, we collected CSF samples in EDTA tubes via lumbar puncture or using an Ommaya reservoir (Integra LifeSciences, Inc., Princeton, NJ, USA). The samples were aliquoted into cryotubes and stored at −80°C until analysis. We selected a cohort of 17 patients with confirmed CNS involvement for a detailed study. CNS involvement was confirmed via brain biopsy in cases of parenchymal abnormalities and via cytological CSF examination for leptomeningeal cases. Patients with leptomeningeal involvement who were cytology-negative were identified based on their response to CNS-directed therapy and imaging findings. We monitored all patients’ clinical outcomes, including survival status, via follow-up MRI.

We extracted cell-free DNA (cfDNA) from 1.0–1.5 mL of CSF using a MagMAX Cell-Free Total Nucleic Acid Isolation Kit (Thermo Fisher Scientific, Waltham, MA, USA). DNA yield and size distribution were assessed using a TapeStation 4150 instrument (Agilent Technologies, Santa Clara, CA, USA) and Qubit 4.0 fluorometer (Thermo Fisher Scientific). We utilized 0.04–40 ng of DNA for library construction. The DNA was ligated using a Twist MF Library prep Kit (Twist Bioscience, San Francisco, USA) with Illumina adapters and indexed with unique dual indices for duplex sequencing (Illumina, San Diego, CA, USA). The sequencing libraries were hybridized with custom probes targeting 112 genes known to be mutated in lymphomas (Supplemental Data Table S1). Pooled libraries were paired-end sequenced (2×150 bp) on the NovaSeq 6000 System (Illumina, San Diego, CA, USA). Single-nucleotide variants (SNVs) and insertions/deletions were called using PiSeq (Dxome, Sungnam, Korea) to differentiate low-frequency variants from amplification artifacts and sequencing errors. The analytical sensitivity for SNVs was assumed to be 0.24% [15]. Copy number alterations were identified using ExomeDepth and a custom tool. Variants were visually confirmed using Integrative Genome Viewer (Broad Institute, Cambridge, MA, USA). The variant allele frequency (%) was calculated as the number of sequencing reads of a specific DNA variant divided by the overall coverage at that locus.

We analyzed 17 patients with CNS involvement who had CSF samples available for targeted sequencing (Fig. 1). Fourteen patients had systemic DLBCL with secondary CNS involvement, such as isolated CNS relapse or disease progression including the CNS, whereas three had primary CNS lymphoma (Table 1). However, their CSF cytology showed negative results although they were suspicious of CNS involvement, such as radiological leptomeningeal involvement in more than half of the cases. The average sequencing coverage depth for CSF samples was 53,733× (range, 12,563–90,023×). The median cfDNA concentration was 45.85 ng/mL (2.15–12,000 ng/mL). Library construction failed for one sample with the lowest cfDNA concentration (2.15 ng/mL, case No. 6), and variants were not detected in three other patients (case Nos. 1, 8, and 11). The overall variant detection rate was 76% (13/17, Fig. 1). In total, 182 variants were detected in 13 patients, with the most common variants occurring in KMT2D, HIST1H1, PIM1, and MYD88 (Fig. 2A). CSF samples from two patients (case Nos. 9 and 16) were collected serially, with consistent ctDNA variant profiles (Fig. 2B). The inability to detect variants or successfully prepare libraries was significantly associated with the DNA concentration in the CSF (Fig. 2C).

Figure 1. Study flow.
Abbreviations: CNS, central nervous system; CSF, cerebrospinal fluid.

Figure 2. Mutation profiles and DNA concentration distribution in CSF from patients with CNS involvement. (A) Mutation profiles detected in ctDNA from 14 patients. (B) Consistency of mutation profiles in serial samples of two patients (No. 9 and 16). (C) DNA concentration distribution according to the status of ctDNA detection.
Abbreviations: ctDNA, circulating tumor DNA; CSF, cerebrospinal fluid; CNS, central nervous system.

Patient clinical characteristics at CSF sampling time and CSF ctDNA detection
Case No.Age (yrs)SexDiagnosisClinical history of CNS involvementRadiologic CNS
involvement pattern
CSF cytologyCSF ctDNASurvival
172MDLBCL, ABC typeRCHOP 6 cycles for stage I → Isolated CNS relapseLeptomeningesNegativeNot detected-Dead
254MDLBCL, ABC typeRCHOP 6 cycles for stage 4 → Isolated CNS relapseParenchyma+LeptomeningesPositiveDetectedDead
341MDLBCL, ABC typeRCHOP 6 cycles for stage 1 → Isolated CNS relapseLeptomeningesPositiveDetectedAlive
434MPrimary CNS DLBCLPrimary CNS involvementParenchymaNegativeDetectedAlive
557FDLBCL, ABC typeRCHOP 6 cycles for stage 1 → Isolated CNS relapseParenchymaPositiveDetectedDead
639FDLBCL, GC typeRCHOP 6 cycles for stage 4 → Systemic relapse → ICED followed by ASCT → Systemic relapse with CNS involvementLeptomeningesNegativeLibrary construction failedDead
773MDLBCL, GC typeRCHOP 6 cycles for stage 3 → Isolated CNS relapseLeptomeningesPositiveDetectedDead
861MDLBCL, ABC typeRCHOP 6 cycles for stage 2 → Isolated CNS relapseParenchymaPositiveNot detectedAlive
940MDLBCL, GC typeRCHOP 3 cycles for stage 4 → Progression with CNS involvementLeptomeningesPositiveDetectedDead
1072MDLBCL, ABC typeRCHOP 6 cycles for stage 4 → Isolated CNS relapseLeptomeningesNegativeDetectedDead
1152MDLBCL, GC typeRCHOP 6 cycles for stage 4 → Systemic relapse with CNS involvementParenchyma+LeptomeningesNegativeNot detectedDead
1272MPrimary CNS DLBCLPrimary CNS involvementParenchymaNegativeDetectedAlive
1353MDLBCL, ABC typeRCHOP 2 cycles for stage 4 → Progression with CNS involvementParenchymaNegativeDetectedDead
1473FDLBCL, GC typeRCHOP 3 cycles for stage 4 → Isolated CNS relapseParenchyma+LeptomeningesNegativeDetectedDead
1571MPrimary CNS DLBCLPrimary CNS involvementParenchyma+LeptomeningesNegativeDetectedDead
1641FDLBCL, ABC typeRCHOP 6 cycles for stage 4 → Systemic relapse with CNS involvementParenchyma+LeptomeningesPositiveDetectedDead
1768FDLBCL, ABC typeRCHOP 6 cycles for stage 4 → Isolated CNS relapseParenchymaNegativeDetectedDead

Abbreviations: CSF, cerebrospinal fluid; ctDNA; circulating tumor DNA; DLBCL, diffuse large B-cell lymphoma; ABC, activated B-cell like; GC, germinal center; CNS, central nervous system; RCHOP, rituximab, cyclophosphamide, doxorubicin, vincristine, prednisolone.



The feasibility of detecting ctDNA in the CSF of patients with large B-cell lymphoma involving the CNS is an area of active research. CNS involvement in large B-cell lymphoma presents diagnostic and therapeutic challenges, and the ability to detect tumor-specific DNA in the CSF may offer valuable insights for diagnosis, monitoring, and treatment decisions. Droplet digital polymerase chain reaction demonstrated a 71% detection rate (10 of 14 cell-free CSF samples) for MYD88 variants in patients with primary CNS lymphoma [16]. A later study reported a slightly higher detection rate of 76.9% (20 of 26 cases) in a similar cohort [17]. However, these studies were limited to specific variants, such as the MYD88 variant. Given the variety of variants present in systemic DLBCL, a broader genetic analysis is recommended. For instance, a Spanish study employing a 300-gene panel assessed the feasibility and utility of ctDNA detection in CSF for both primary CNS lymphoma and systemic lymphoma with secondary CNS involvement [18]. Variants were successfully detected in all six patients with CNS involvement [18]. A recent study using CSF samples from 92 patients reported a 100% variant detection rate using cancer personalized profiling by deep sequencing (CAPP-Seq) for primary CNS lymphoma or secondary CNS involvement [19]. Despite its effectiveness, CAPP-Seq may not be universally available, making targeted sequencing of a large number of genes a more practical option in some settings. Our variant detection rate aligns with these findings, and our study provides a feasible approach for routine clinical application.

Despite its promising performance in variant detection, CSF ctDNA analysis still has some challenges. One challenge is the low concentration of ctDNA in CSF compared to that in peripheral blood, hampering detection [20]. Additionally, distinguishing tumor-derived DNA from background DNA released from normal cells in the CNS presents a challenge, especially considering the potential for contamination from blood or other sources during CSF collection. Despite these challenges, advancements in NGS techniques have improved the ctDNA detection sensitivity, enabling researchers to detect tumor-specific variants in CSF samples.

Our study demonstrated the feasibility of CSF ctDNA sequencing as a diagnostic tool for detecting CNS involvement in DLBCL despite most patients showing negative CSF cytological results. The development of early detection methods for identifying tumor cells in CSF may significantly impact treatment strategies for patients with systemic DLBCL. This would enable the initiation of targeted therapies for patients who are CSF cytology-negative yet suspected of CNS involvement, potentially altering their disease course. Advancements in CSF-based targeted sequencing may help in predicting the risk of CNS relapse or progression in patients with systemic DLBCL, facilitating the development of preemptive strategies for those at high risk. Given these potential benefits, there is a pressing need for further research to enhance the feasibility and accuracy of CSF targeted sequencing. This may ultimately lead to more precise and effective management of patients with DLBCL with or at risk of CNS involvement.

We are grateful to all our colleagues who participated in the research project.

Conception and design: Kim SJ; Resources: Yoon SE and Kim WS; Acquisition of data and experiments: Park MR, Park B, Ryu KJ, and Shin S; Analysis and interpretation of data: Kim SJ, Kim JJ, Shin S, and Lee ST; Drafting of manuscript: Kim SJ, Kim JJ, and Shin S. All authors read and approved the final manuscript.

This study was supported by a grant from the National Research Foundation of Korea (NRF-2021R1I1A1A01045980).

  1. Ghose A, Elias HK, Guha G, Yellu M, Kundu R, Latif T, assignee. Influence of rituximab on central nervous system relapse in diffuse large B-cell lymphoma and role of prophylaxis-a systematic review of prospective studies. Clin Lymphoma Myeloma Leuk 2015;15:451-7.
    Pubmed CrossRef
  2. Jeong SY, Yoon SE, Cho D, Kang ES, Cho J, Kim WS, et al, assignee. Real-world experiences of CNS-directed chemotherapy followed by autologous stem cell transplantation for secondary CNS involvement in relapsed or refractory diffuse large B-cell lymphoma. Front Oncol 2022;12:1071281.
    Pubmed KoreaMed CrossRef
  3. Thiel E, Korfel A, Martus P, Kanz L, Griesinger F, Rauch M, et al, assignee. High-dose methotrexate with or without whole brain radiotherapy for primary CNS lymphoma (G-PCNSL-SG-1): a phase 3, randomised, non-inferiority trial. Lancet Oncol 2010;11:1036-47.
    Pubmed CrossRef
  4. Tomita N, Kodama F, Kanamori H, Motomura S, Ishigatsubo Y, assignee. Secondary central nervous system lymphoma. Int J Hematol 2006;84:128-35.
    Pubmed CrossRef
  5. Kim SJ, Oh SY, Kim JS, Kim H, Lee GW, Won JH, et al, assignee. Secondary central nervous system (CNS) involvement in patients with diffuse large B-cell lymphoma: a therapeutic dilemma. Ann Hematol 2011;90:539-46.
    Pubmed CrossRef
  6. Demirci U, Yüksel MK, Kırkızlar HO, Ateşoğlu EB, Mehtap Ö, Salim O, et al, assignee. A survey evaluating hematology physicians' perspectives on central nervous system prophylaxis. Blood Res 2023;58:99-104.
    Pubmed KoreaMed CrossRef
  7. Jen J, Wu L, Sidransky D, assignee. An overview on the isolation and analysis of circulating tumor DNA in plasma and serum. Ann N Y Acad Sci 2000;906:8-12.
    Pubmed CrossRef
  8. Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, et al, assignee. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012;366:883-92.
    Pubmed KoreaMed CrossRef
  9. Shin SH, Kim YJ, Lee D, Cho D, Ko YH, Cho J, et al, assignee. Analysis of circulating tumor DNA by targeted ultra-deep sequencing across various non-Hodgkin lymphoma subtypes. Leuk Lymphoma 2019;60:2237-46.
    Pubmed CrossRef
  10. Kim JJ, Kim HY, Choi Z, Hwang SY, Jeong H, Choi JR, et al, assignee. In-depth circulating tumor DNA sequencing for prognostication and monitoring in natural killer/T-cell lymphomas. Front Oncol 2023;13:1109715.
    Pubmed KoreaMed CrossRef
  11. Kim SJ, Kim YJ, Yoon SE, Ryu KJ, Park B, Park D, et al, assignee. Circulating tumor DNA-based genotyping and monitoring for predicting disease relapses of patients with peripheral T-cell lymphomas. Cancer Res Treat 2023;55:291-303.
    Pubmed KoreaMed CrossRef
  12. Yoon SE, Shin SH, Nam DK, Cho J, Kim WS, Kim SJ, assignee. Feasibility of circulating tumor DNA analysis in patients with follicular lymphoma. Cancer Res Treat 2024;56:920-35.
    Pubmed KoreaMed CrossRef
  13. Yoon SE, Kim YJ, Shim JH, Park D, Cho J, Ko YH, et al, assignee. Plasma circulating tumor DNA in patients with primary central nervous system lymphoma. Cancer Res Treat 2022;54:597-612.
    Pubmed KoreaMed CrossRef
  14. Cho J, assignee. Basic immunohistochemistry for lymphoma diagnosis. Blood Res 2022;57:55-61.
    Pubmed KoreaMed CrossRef
  15. Lee KS, Seo J, Lee CK, Shin S, Choi Z, Min S, et al, assignee. Analytical and clinical validation of cell-free circulating tumor DNA assay for the estimation of tumor mutational burden. Clin Chem 2022;68:1519-28.
    Pubmed CrossRef
  16. Rimelen V, Ahle G, Pencreach E, Zinniger N, Debliquis A, Zalmai L, et al, assignee. Tumor cell-free DNA detection in CSF for primary CNS lymphoma diagnosis. Acta Neuropathol Commun 2019;7:43.
    Pubmed KoreaMed CrossRef
  17. Watanabe J, Natsumeda M, Okada M, Kobayashi D, Kanemaru Y, Tsukamoto Y, et al, assignee. High detection rate of MYD88 mutations in cerebrospinal fluid from patients with CNS lymphomas. JCO Precis Oncol 2019;3:1-13.
    Pubmed CrossRef
  18. Bobillo S, Crespo M, Escudero L, Mayor R, Raheja P, Carpio C, et al, assignee. Cell free circulating tumor DNA in cerebrospinal fluid detects and monitors central nervous system involvement of B-cell lymphomas. Haematologica 2021;106:513-21.
    Pubmed KoreaMed CrossRef
  19. Mutter JA, Alig SK, Esfahani MS, Lauer EM, Mitschke J, Kurtz DM, et al, assignee. Circulating tumor DNA profiling for detection, risk stratification, and classification of brain lymphomas. J Clin Oncol 2023;41:1684-94.
    Pubmed KoreaMed CrossRef
  20. Song HH, Park H, Cho D, Bang HI, Oh HJ, Kim J, assignee. Optimization of a Protocol for Isolating Cell-free DNA From Cerebrospinal Fluid. Ann Lab Med 2024;44:294-8.
    Pubmed KoreaMed CrossRef