- Review
- Open access
- Published:
Circulating tumor DNA in lymphoma: technologies and applications
Journal of Hematology & Oncology volume 18, Article number: 29 (2025)
Abstract
Lymphoma, a malignant tumor derived from lymphocytes and lymphoid tissues, presents with complex and heterogeneous clinical manifestations, requiring accurate patient classification for appropriate treatment. While invasive pathological examination of lymph nodes or lymphoid tissue remains the gold standard for lymphoma diagnosis, its utility is limited in cases of deep-seated tumors such as intraperitoneal and central nervous system lymphomas. In addition, biopsy procedures carry an inherent risk of complications. Computed tomography (CT) and positron emission tomography/computed tomography (PET/CT) imaging are essential for treatment assessment and monitoring, but lack the ability to detect early clonal evolution and minimal residual disease (MRD). Liquid biopsy-based analysis of circulating tumor DNA (ctDNA) offers a non-invasive alternative that allows for repeated sampling and overcomes the limitations of spatial heterogeneity and invasive biopsies. ctDNA provides genetic and epigenetic insights into lymphoma and serves as a dynamic, quantifiable biomarker for diagnosis, risk stratification, and treatment response. This review comprehensively summarizes common genetic variations in lymphoma and systematically evaluates ctDNA detection technologies, including PCR-based assays and next-generation sequencing (NGS). Applications of ctDNA detection in noninvasive genotyping, risk stratification, therapeutic response monitoring, and MRD detection are discussed across various lymphoma subtypes, including diffuse large B-cell lymphoma, Hodgkin lymphoma, follicular lymphoma, and T-cell lymphoma. By integrating recent research findings, the review highlights the role of ctDNA profiling in advancing precision medicine, enabling personalized therapeutic strategies, and improving clinical outcomes in lymphoma.
Background
Lymphoma, a malignant neoplasm that originates from lymphocytes and lymphoid tissues, has experienced a significant increase in incidence over recent years [1]. The complexity of its pathological types necessitates a comprehensive classification system. The latest World Health Organization (WHO) classification, authored by a diverse group of experts in hematopathology, hematology, oncology, genetics, and molecular biology, categorizes lymphomas based on cell lineage, such as B-, T-, or natural killer (NK) cells, and further stratifies subtypes within each lineage according to morphology, immunophenotype, genetic features, and clinical presentation [2]. The clinical manifestations of different lymphoma subtypes exhibit marked heterogeneity, resulting in different treatments [3,4,5].
Recent advances in biomedical technologies, such as next-generation sequencing (NGS) and high-throughput drug screening, have enhanced our understanding of lymphoma and facilitated the development of targeted therapies and immunotherapy, thereby improving treatment outcomes and survival rates [5,6,7]. Although aggressive B-cell lymphomas are often curable with combination chemotherapy and immunotherapy, indolent lymphomas typically achieve durable remissions but require lifelong monitoring due to their incurable nature. Among aggressive subtypes, diffuse large B-cell lymphoma (DLBCL) poses a significant challenge, as resistance or relapse after first-line therapy is common for a subset of patients [8]. Therefore, accurately identifying patients at risk of refractory disease or relapse, along with early prognostic predictions at diagnosis, has become increasingly important. Such prognostic assessments are crucial for guiding personalized treatment strategies [5, 9, 10].
Circulating cell-free DNA (cfDNA) refers to small fragments of DNA, typically ∼ 70–200 base pairs in length, that are released into the bloodstream or other body fluids as a result of cellular apoptosis (programmed cell death) or necrosis (cell death due to injury or disease) [11,12,13,14]. Originating from various organs and tissues, cfDNA can be detected in blood, urine, saliva, and additional body fluids [15]. Notably, as a dynamic biomarker, its concentration fluctuates under different physiological and pathological conditions, such as pregnancy, organ transplantation, and cancer. In recent years, cfDNA has garnered attention in clinical and research settings due to its non-invasive nature and utility as a biomarker for conditions such as cancer, prenatal testing, organ transplant monitoring, and infectious diseases [16,17,18,19,20]. Analysis of cfDNA can reveal genetic and epigenetic changes, offering insights into underlying physiological or pathological processes [21,22,23].
Circulating tumor DNA (ctDNA), a subset of cfDNA that originates from tumor cells, carries distinct genetic and epigenetic signatures specific to cancer (Fig. 1). ctDNA constitutes a variable fraction of cfDNA and has been extensively studied across a range of cancers, including lung, breast, colorectal, kidney, and hematological malignancies [23]. As ctDNA reflects the molecular characteristics of the tumor, it is increasingly being used for cancer prognosis, diagnosis, and monitoring of therapeutic responses. Recent research highlights the potential of ctDNA in lymphoma for risk stratification, therapeutic response assessment, and disease progression monitoring [24,25,26,27].
In this review, we provide a comprehensive overview of common genetic alterations in lymphoma and evaluate the current ctDNA detection technologies. We also examine recent advancements in the application of ctDNA in malignant lymphoma, emphasizing its pivotal role in advancing personalized medicine through non-invasive approaches.
Molecular genetics of lymphoma
Lymphomas are broadly classified into two main types: non-Hodgkin lymphoma (NHL), accounting for about 85–90% of cases, and Hodgkin lymphoma (HL), comprising the remaining 10–15% [3]. NHL itself represents a diverse group of malignancies with distinct genetic profiles [28, 29]. Approximately 85–90% of NHL cases originate from B cells, including DLBCL, follicular lymphoma (FL), marginal zone lymphoma (MZL), and mantle cell lymphoma (MCL), while the remainder derive from T cells or natural killer (NK) cells. Recent advances in NGS have provided comprehensive insights into the genomic landscape of these lymphoma subtypes, as illustrated in Fig. 2.
Overview of lymphoma types, subtypes, genetic changes, and impacted signaling pathways in this review. Orange: lymphoma subtypes; light blue: the cell-of-origin classification of DLBCL; green: genetic subtypes of lymphoma; dark gray: genetic hallmarks of lymphoma; light gray: deregulated biological pathways. Abbreviations: DLBCL, diffuse large B-cell lymphoma; MCL, mantle cell lymphoma; PCNSL, primary central nervous system lymphoma; BL, Burkitt lymphoma; PTCL, peripheral T-cell lymphoma; FL, follicular lymphoma; cHL, classical Hodgkin lymphoma. N/A, not available
Diffuse large B-cell lymphoma
DLBCL, accounting for 30–40% of B-cell NHL, exhibits significant genetic heterogeneity that complicates a complete definition of its landscape (Fig. 2) [30, 31]. Integrative analyses of large biopsy cohorts have clarified its genetic variation, while ctDNA analysis detects alterations missed in tissue biopsies [32,33,34]. The pathogenic alterations affect key processes: B-cell differentiation (BCL-6 translocations disrupt germinal center responses and PRDM1 alterations enhance NF-κB activation and impair plasma cell differentiation); B-cell receptor signaling (mutations in CD79B and CARD11, BCL10 amplifications, the MYD88L265P mutation in Toll-like receptor signaling, TNFAIP3 mutations, increased REL expression, PTEN deletions, and PIK3CA amplifications or activating mutations in the PI3K–AKT–mTOR pathway); apoptosis (alterations in BCL2 and FAS); epigenetic regulation (mutations in KMT2D, CREBBP, EZH2, and EP300); and immune evasion (PD-L1 overexpression) [30, 35].
Molecular subtypes based on these features enable risk stratification through RNA-based cell-of-origin classifications (germinal center B-cell-like and activated B-cell-like) and genomic aberration-based systems defining five clusters (C1- C5) and seven subtypes (EZB MYC+, EZB MYC-, ST2, BN2, A53, N1, and MCD) (Fig. 2) [36,37,38,39,40], with plasma ctDNA genotyping further complementing RNA-based classification [41, 42]. Collectively, these genetic insights underscore the complexity of DLBCL and suggest avenues for targeted therapies and prognostic markers.
Mantle cell lymphoma and Burkitt lymphoma(BL)
MCL and BL have distinctive genetic hallmarks (Fig. 2). MCL is defined by the t(11;14)(p13;q32) translocation leading to CCND1 overexpression and recurrent variations in DNA damage repair (ATM, TP53), epigenetic regulation (NSD2, KMT2D, MEF2B, KMT2C, SMARCA4), and cellular homeostasis and growth (CCND1, CDKN2A, BIRC3, CARD11, TRAF2, RB1, POT1, NOTCH1/2) [43,44,45,46]. BL is characterized by the t(8;14) translocation that activates MYC, along with mutations in TCF3, ID3, CCND3, TP53, and CDKN2A [47,48,49,50,51]. Both MCL and BL can be divided into outcome-related subtypes based on comprehensive genomic and transcriptomic profiles [50, 52].
Follicular lymphoma and marginal zone lymphoma
FL and MZL are common indolent B-NHLs, with approximately 15% of cases transforming to aggressive B-cell lymphomas. FL is characterized by the t(14;18)(q32;q21) translocation, which is necessary but not sufficient for its development [53, 54], and frequently shows alterations in epigenetic regulators such as CREBBP, KMT2D, EZH2, and EP300 [55, 56]. MZL comprises three subtypes: extranodal, splenic, and nodal. Extranodal MZL is frequently associated with the t(11;18)(q21;q21) translocation that produces the API2-MALT1 fusion and is less often linked to the t(14;18)(q32;q21) translocation (IGH-MALT1) or the t(1;14)(p22;q32) translocation (BCL10-IGH) [57, 58]. Both splenic and nodal MZL share genomic alterations including mutations in KMT2D, NOTCH2, PTPRD, TNFAIP3, and KLF2 [59]. Predictive models based on these genetic features have been developed to assess the risk of histological transformation of FL and MZL to aggressive lymphoma [60,61,62].
Central nervous system lymphoma (CNSL)
CNSL is a rare, aggressive NHL subtype comprising primary CNS lymphoma (PCNSL) and secondary CNS lymphoma (SCNSL), the latter indicating lymphoma metastasized to the CNS from systemic disease. PCNSL, confined to the CNS at diagnosis, is marked by recurrent somatic mutations in key genes (PIM1, MYD88, CD79B, KMT2D, and BTG2) that affect critical signaling pathways (JAK-STAT, NF-κB, and B-cell receptor) and drive lymphomagenesis and progression [63, 64]. Additional alterations include recurrent amplifications and deletions at 18q21.23 and 6p21, along with a notable loss of MHC class I expression that may aid immune evasion. An integrative analysis of 240 PCNSL cases identified four distinct molecular clusters (C1, C2, C3, and C4) (Fig. 2), each with unique genetic and epigenetic profiles and prognostic outcomes [65]. These classifications offer valuable insights into PCNSL heterogeneity and have significant implications for personalized therapeutic strategies and prognostication.
Peripheral T-cell lymphoma (PTCL)
PTCL refers to an uncommon and diverse collection of aggressive NHL originating from mature T cells and NK cells [66]. Its major subtypes include PTCL-NOS, angioimmunoblastic T-cell lymphoma (AITL), ALK-positive/negative anaplastic large cell lymphoma (ALCL), and extranodal NK/T-cell lymphoma (ENKTL) (Fig. 2). PTCL-NOS often harbors mutations in histone-modifying genes (EZH2, KDM6A, and KMT2) [67], while AITL is marked by recurrent inactivating RHOAG17V mutations and alterations in epigenetic regulators (TET2, DNMT3A, and IDH2), alongside activation of TCR and PI3K-AKT pathways [68, 69]. ENKTL exhibits mutations in RNA helicase genes (e.g., DDX3X), aberrations in the JAK-STAT and RAS-MAPK pathways, and alterations in epigenetic modulators (KMT2C, KMT2D) [70]. ALK-positive ALCL is defined by the NPM-ALK fusion that activates STAT3, whereas ALK-negative ALCL involves NFKB2-ROS2 and NFKB2-TYK2 fusions driving STAT3 activation [71]. Comprehensive genomic profiling has delineated four distinct molecular and microenvironmental PTCL subtypes with unique features [72].
Hodgkin lymphoma (HL)
Over 90% of HL cases are classified as classical HL (cHL), while nodular lymphocyte-predominant HL (NLPHL) accounts for about 5–10%. Tissue-based genomic profiling of cHL is limited by the low abundance of Hodgkin and Reed-Sternberg (HRS) cells, which represent only 0.1–10% of tumor cellularity [73, 74]. In contrast, noninvasive cfDNA profiling has demonstrated superior performance [34, 75,76,77]. cfDNA and flow-sorted HRS cell sequencing have revealed recurrent mutations in SOCS1, TNFAIP3, B2M, STAT6, CSF2RB, GNA13, PTPN1, ARID1A, ZNF217, IL4R, NFKBIA, ACTB, PCBP1, CISH, NFKB2, and linker histone H1-5, along with recurrent copy number variants (CNVs), including 2p15 (REL), 9p24.1-9p24.2 (PDL1), 5p15.33 (TERT), 17q21.31 (MAP3K14), 6q27 (TNFAIP3), 17p13.1 (TP53), 9p21.3 (CDKN2A/B), 11q22.3 (BIRC3) and 6p21-22 (H1-5, HLA-A and HLA-C) [75,76,77,78,79,80,81,82]. Targeted ctDNA sequencing of 366 patients has defined two cHL subtypes: cluster H1 (68% of cases), characterized by mutations in the NF-κB, JAK/STAT, and PI3K-AKT pathways, and cluster H2 (32%), which exhibits broader structural abnormalities and harbors mutations in TP53 and KMT2D [34].
NLPHL displays a distinct genetic profile. It is defined by the presence of lymphocyte-predominant cells and lacks many of the mutations common in cHL [2, 73] In NLPHL, targeted sequencing reveals mutations in SGK1, DUSP2, and JUNB, as well as frequent BCL6 translocations - a finding that is rare in cHL. Moreover, mutations in STAT6, JAK2, TNFAIP3, and NFKBIA, which are prevalent in cHL, are uncommon in NLPHL [83, 84].
Methods for ctDNA detection and analysis
The clinical application of conventional genomic profiling in lymphoma management faces challenges due to spatial heterogeneity and the limited tumor material typically obtained from fine-needle aspirations and core needle biopsies. ctDNA profiling presents a noninvasive approach to capture comprehensive molecular characteristics without these sampling limitations, showing potential for genotyping, response assessment, and MRD monitoring in lymphomas.
Liquid biopsy technologies, particularly through analysis of ctDNA in body fluids like peripheral blood, have emerged as valuable tools for lymphoma detection and monitoring. Optimal ctDNA collection and processing depend on key preanalytical factors, including blood volume, timing of plasma isolation, and use of cell-stabilizing tubes to prevent cellular DNA contamination [25, 85,86,87,88]. Among ctDNA profiling technologies, PCR-based methods and NGS-based approaches are most common and have been extensively studied in recent years (Table 1; Fig. 3).
Summary of key ctDNA detection technologies in lymphoma. Abbreviations: PCR, polymerase chain reaction; BEAMing, beads, emulsion, amplification and magnetics; ASO-PCR, allele specific oligonucleotide polymerase chain reaction; CNVs, copy number variants; ddPCR, droplet digital polymerase chain reaction; NGS, next generation sequencing; IgHTS, immunoglobulin high-throughput sequencing; VDJ, variable, diversity, joining; WGS, whole genome sequencing; WES, whole exome sequencing; CAPP-seq, cancer personalized profiling by deep sequencing; SNVs, single nucleotide variants; PhasED-seq, phased variant enrichment and detection sequencing; TBS, Targeted bisulfite sequencing
PCR-based methods
PCR assays, including BEAMing (beads, emulsion, amplification, and magnetics) [89], allele-specific oligonucleotide PCR (ASO-PCR) [90] and digital droplet PCR (ddPCR) [91], are cost-effective and provide rapid turnaround times, making them well-suited for ctDNA-based assays in lymphomas (Fig. 3). BEAMing leverages magnetic bead-based PCR amplification within microemulsions and flow cytometry, employing streptavidin-coated beads and biotinylated oligonucleotides for the highly sensitive detection and quantification of nucleotide variations through fluorescent labeling and analysis of PCR products (Fig. 3a) [89]. With a sensitivity as low as 0.01%, this technique reliably detects genetic alterations in ctDNA and shows strong concordance with alterations identified in patient tissue samples (Table 1) [92].
ASO-PCR, or amplification refractory mutation system (ARMS), utilizes uniquely designed primers to amplify DNA when there is a perfect match at single-nucleotide variant (SNV) or wild-type sequences, enabling precise SNV detection through specific PCR product patterns (Fig. 3b) [90]. For example, Jimenez et al. used ASO-PCR to reliably detect the MYD88L265P mutation in lymphoproliferative disorders, highlighting its utility as a sensitive, cost-effective diagnostic tool [87].
ddPCR is considered the gold standard for quantifying DNA mutations due to its ability to partition DNA molecules into thousands of droplets for individual PCR amplification, enabling high sensitivity and absolute quantification without needing standard curves (Fig. 3c) [93]. The method is effective in identifying genetic alternations in lymphomas, such as detecting t(14;18) translocation in FL [54], t(11;14) translocation in MCL [94], and MYD88L265P in PCNSL [95].
Despite their advantages, PCR-based methods are typically limited to detecting a single or few known mutations, with a sensitivity threshold of approximately 0.01% allele frequency (AF) (Table 1) [96].
NGS-based methods
NGS-based technologies allow massive parallel sequencing of DNA molecules, enabling comprehensive assessment of mutational landscapes, including SNVs, insertions and deletions and CNVs [97,98,99,100]. Targeted amplicon-based and hybrid-capture NGS approaches offer advantages over single-gene assays by identifying a broad range of genetic alterations in lymphoma without the need for patient-specific optimization [101]. For example, Dubois et al. developed Lymphopanel-a 34-gene panel applied to samples from 215 patients-that revealed the molecular heterogeneity among DLBCL subtypes and identified mutations with potential therapeutic and prognostic significance [102].
Immunoglobulin high-throughput sequencing (IgHTS), marketed as clonoSEQ by Adaptive Biotechnology, is FDA approved for the detection of MRD in chronic lymphocytic leukemia (CLL), multiple myeloma (MM), and B-cell acute lymphoblastic leukemia (B-ALL) [85]. This technique uses universal primers for PCR amplification of immunoglobulin genomic regions, followed by sequencing to identify the amplified IgV(D)J clonotypes (Fig. 3d) [103]. IgHTS achieves a sensitivity of ∼ 0.005%, although its effectiveness may be limited by somatic hypermutation and the amount of cfDNA analyzed (Table 1) [104]. Because it doesn’t require patient-specific primers, it is widely applicable, although there are some limitations for highly mutated lymphoma subtypes [105].
Cancer personalized profiling by deep sequencing (CAPP-seq) [99], initially developed for non-small cell lung cancer with approximately 125 kb coverage, combines unique barcoding strategies with bioinformatics algorithms to improve sensitivity and enable ctDNA detection down to allele frequencies of ∼ 0.002% (Fig. 3e; Table 1) [106]. This ultra-sensitive assay is used in diverse oncology research areas, including early detection, non-invasive genotyping, resistance mutation identification, and disease burden quantification [41, 43, 107].
Phased variant enrichment and detection sequencing (PhasED-Seq) is a hybrid capture method that uses phased variants, detects mutations within 150 base pairs on the same DNA strand for higher ctDNA sensitivity (Fig. 3f) [97]. Phased mutations are commonly found in specific genomic regions of B-cell lymphoma due to both normal and abnormal somatic hypermutation. Analytical sensitivity, tested by diluting lymphoma ctDNA in healthy cfDNA, achieved a detection threshold of 0.00005% (Table 1) [108].
Other techniques, including pyrosequencing [109], whole exome sequencing (WES) [110] and whole genome sequencing (WGS) [82], have been widely used for ctDNA analysis in lymphoma patients (Fig. 3g). Additionally, methods assessing epigenetic modifications as biomarkers for lymphoma diagnosis and monitoring are being explored (Fig. 3h) [111]. Using pyrosequencing, Kristensen and colleagues demonstrated the feasibility of detecting aberrant promoter DNA methylation in cfDNA from the plasma of DLBCL patients. They identified aberrant DAPK1 methylation as an independent prognostic marker associated with treatment response and patient survival [112]. Furthermore, targeted bisulfite sequencing (TBS) of over 100,000 genomic regions using ctDNA has enabled the detection of more than 50 cancer types, including lymphoma, across all stages, achieving an average sensitivity of 54.9% at a specificity exceeding 99% and a single false positive rate below 1% [23].
Although numerous techniques have been reported, significant challenges remain in the analysis of ctDNA in lymphoma, including the detection of low absolute and fractional amounts (often less than 0.5%) of ctDNA, as well as the diversity of mutations present in lymphoma (Table 1) [85, 86, 105].
Application of ctDNA detection in lymphoma
Studies on the application of ctDNA in lymphoma have predominantly focused on DLBCL, with several investigations extending to other lymphoma subtypes (Tables 2 and 3). This section summarizes the clinical applications of ctDNA for noninvasive genotyping, treatment response monitoring, and MRD assessment across various lymphoma subtypes (Fig. 4).
Clinical applications of ctDNA testing in lymphoma. ctDNA profiling provides a noninvasive liquid biopsy method to capture comprehensive molecular characteristics, overcoming the limitations of traditional tissue sampling. Applications include diagnosis, genotyping, outcome prediction, response assessment, and MRD monitoring in lymphoma
Clinical significance of ctDNA testing in diffuse large B-cell lymphoma
ctDNA as a non-invasive genotyping biomarker
Genetic profiling of lymphoma tissue obtained via biopsy or surgery is essential for diagnosis and subtype classification. However, such tissue analysis can be challenging due to spatial heterogeneity and limited material from fine needle aspirations or core needle biopsies. To overcome these limitations, ctDNA has been investigated as a noninvasive biomarker for capturing the complete molecular landscape of DLBCL (Table 2). Studies have demonstrated a concordance rate greater than 70% between ctDNA and tissue-based genotyping in DLBCL [32, 113,114,115]. This high concordance highlights the potential for ctDNA genotyping in individualized therapy selection for DLBCL. For instance, patients with the ABC-DLBCL subtype harboring B-cell receptor mutations, particularly those with MYD88 mutations, have shown high response rates to ibrutinib (80%) [116]. Furthermore, ctDNA-based cell of origin (COO) classification tools have shown strong concordance with tumor biopsies and have been used effectively for individualized risk stratification [41, 42]. Additionally, ctDNA analysis often detects novel mutations not found in tissue biopsies, which could be due to tumor spatial heterogeneity, clonal evolution in recurrence, or selective pressures from targeted therapy [32, 41, 113, 117,118,119]. These findings underscore that ctDNA genotyping is a clinically feasible, noninvasive tool for DLBCL patients.
ctDNA quantification in evaluating tumor burden and treatment efficacy
Beyond genotyping, pretreatment ctDNA levels can provide a reliable assessment of disease burden and predict outcomes in DLBCL. Baseline ctDNA levels correlate with total metabolic tumor volume (TMTV), international prognostic index (IPI), lactate dehydrogenase (LDH) levels, and Ann Arbor stage, with high predictive value for clinical outcomes in patients receiving standard immunochemotherapy [41, 107, 113, 120,121,122,123]. A study by Alig et al. demonstrated that pretreatment ctDNA levels also predicted a short diagnosis-to-treatment interval and served as an independent prognostic marker for event-free survival (EFS) in 267 DLBCL patients [124].
Traditional risk stratification tools like IPI and TMTV are often used only once before treatment and have not achieved the desired precision for personalized treatment [125,126,127,128,129]. Inspired by molecular response models in chronic myelogenous leukemia and CLL, recent studies have investigated the prognostic utility of ctDNA molecular responses in DLBCL treated with anthracycline-based regimens [130, 131]. In an early study, DLBCL patients with undetectable interim ctDNA by lgHTS after two cycles of dose-adjusted EPOCH-R treatment demonstrated favorable 5-year progression-free survival (PFS) [120]. Based on these findings, Kurtz and colleagues proposed thresholds for early molecular response (EMR) using CAPP-seq after a single cycle of front-line R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone) therapy. In their framework, a 2-log ctDNA reduction defined EMR, and a 2.5-log reduction after two cycles was classified as major molecular response (MMR). Both EMR and MMR were significantly correlated with improved EFS at 24 months for patients receiving either front-line or salvage therapy, with multivariate analyses indicating that EMR and MMR independently predicted EFS and overall survival (OS) [107]. It is also demonstrated that the molecular response (EMR or MMR, hazard ratios for EMR and MMR were 6.5–10 and 11–26) showed more strongly prognostic of outcomes than pretreatment ctDNA (hazard ratio of 2.4–2.6). Additionally, ctDNA clearance after four treatment cycles has been identified as an independent prognostic biomarker in multivariate analyses [132]. To address limitations associated with fixed-point ctDNA assessments, the Kurtz group developed the continuous individualized risk index (CIRI), a dynamic model incorporating IPI, pretreatment ctDNA, COO, EMR, MMR, and interim PET/CT to provide individualized risk profiles. The CIRI model outperformed traditional methods in predicting outcomes [133]. Furthermore, combining MMR with interim PET/CT improved PFS stratification, yielding 2-year PFS rates of 84%, 17%, and 0% across different risk groups (p < 0.001) [134].
ctDNA for risk stratification and treatment response in relapsed/refractory (R/R) diffuse large B cell lymphoma
The clinical utility of ctDNA levels in R/R DLBCL has shown promise across various therapies, notably chimeric antigen receptor (CAR) T-cell therapy, where risk stratification and response assessment remain challenging. In a study of 72 R/R DLBCL patients treated with axicabtagene ciloleucel, undetectable ctDNA one-week post-infusion was observed in 70% of patients with durable responses compared to only 13% of those with disease progression (p < 0.0001), while detectable ctDNA at day 28 predicted poorer PFS and OS [121]. An additional study using CAPP-seq in two independent CAR T-cell cohorts revealed that higher ctDNA levels at weeks 1 and 4 were significantly associated with progression (p < 0.05), with pretreatment ctDNA (p = 0.003) and minimal molecular residual at week 4 (p = 0.028) predicting EFS [117].
Baseline ctDNA levels have also been evaluated by CAPP-seq in R/R DLBCL patients receiving polatuzumab vedotin in combination with bendamustine and rituximab (BR) or BR alone, with results showing a correlation between baseline ctDNA and both PFS and OS. ctDNA levels decreased significantly in patients who achieved complete response (CR) compared to non-CR patients [135]. In 40 patients with R/R DLBCL (27 de novo and 13 transformed) treated with panobinostat, with or without rituximab, ctDNA changes at day 15 showed 71.4% sensitivity and 100% specificity in predicting treatment response, compared to baseline ctDNA levels [136]. Dynamic ctDNA monitoring in an Asian cohort of 23 R/R DLBCL patients indicated that undetectable ctDNA at day 28 post-CAR19 T-cell therapy was linked to longer PFS (p = 0.004) and OS (p = 0.004). In addition, shorter ctDNA fragments (< 170 bp) were associated with poorer PFS (P = 0.002) and OS (p = 0.008) [33]. These findings suggest that baseline ctDNA levels, early ctDNA dynamics, and fragment size are promising predictors of treatment response and survival outcomes in R/R DLBCL.
ctDNA for minimal residual disease monitoring
Relapse remains a concern in DLBCL, even among patients achieving remission following first-line anthracycline-based therapy. Conventional surveillance techniques, including clinical examinations and imaging, often lack the sensitivity and specificity to detect early relapse [137, 138]. ctDNA monitoring offers a noninvasive and radiation-free alternative, allowing for personalized MRD detection through tumor-informed or disease-specific assays. Multiple studies have highlighted the utility of ctDNA over imaging for MRD monitoring in DLBCL. Two independent studies demonstrate that IgHTS analysis of plasma ctDNA provided superior specificity and a high negative predictive value, detecting relapse three to 3.5 months before radiographic evidence [104, 120]. Scherer and co-workers applied CAPP-seq to monitor plasma ctDNA in DLBCL patients with CR or recurrence, finding that MRD detection anticipated relapse by an average of 6 months, outperforming IgHTS and imaging techniques [41]. PhasED-Seq further improved sensitivity, detecting MRD in all samples that were undetectable by CAPP-seq prior to biopsy-confirmed recurrence [97]. Moreover, combining ctDNA fragment analysis with mutation-based MRD monitoring may enhance detection accuracy in patients who test negative with mutation-based assays or show positive PET results [122].
Clinical significance of ctDNA testing in other lymphoma types
The application of ctDNA has been also investigated in other NHL types, including MCL, FL, MZL, CNSL, Peripheral T-cell lymphoma and HL as outlined in Table 3.
Mantle cell lymphoma
In untreated MCL, higher pretreatment ctDNA levels have been associated with several prognostic indicators, including the IPI (P = 0.0004) and TMTV (r = 0.73; p = 0.0001). However, no statistically significant survival difference was observed between patients with high pretreatment ctDNA (above the median) and those with low pretreatment ctDNA (below the median) [139]. Noninvasive ctDNA genotyping may also provide insights into treatment resistance mechanisms. For instance, in MCL patients undergoing ibrutinib-venetoclax therapy, ctDNA analysis revealed chromosome 9p21.1–p24.3 loss and specific mutations associated with resistance in all five study participants. Although these findings are promising, further research is needed to determine if such genotypic data can guide personalized treatments in MCL [140].
In a longitudinal study of 53 untreated MCL patients with a median follow-up of 12.7 years, patients achieving undetectable ctDNA after two cycles of induction therapy had significantly longer PFS and OS than those with detectable ctDNA (median PFS: 2.7 vs. 1.8 years, p = 0.005; median OS: 13.8 vs. 7.4 years, p = 0.03). Molecular relapse was detectable before clinical progression in seven patients, suggesting that MRD-guided treatment strategies could improve outcomes [139]. Nevertheless, further investigation is needed to assess MRD-guided treatment by comparing preemptive treatment with conventional therapy for MCL.
Follicular lymphoma and marginal zone lymphoma
EZH2 is an important biomarker for guiding frontline treatment in FL, with tazemetostat being approved for patients harboring EZH2 mutations [141]. In a cohort of 123 FL patients, multiplex ddPCR identified an EZH2 mutation frequency of 41.5% using paired biopsy tissue and ctDNA, which is higher than previous estimates of 20–27% [142,143,144]. This highlights the potential need for routine EZH2 mutation screening in ctDNA sample of FL to refine patient selection for targeted therapies.
In addition to genetic mutation screening, liquid biopsy analyses have provided further insights into the prognostic landscape of FL. High levels of plasma ctDNA mutations in genes such as BCL2, KMT2D, EP300, STAT6, CREBBP, and TP53 have been linked to poor survival outcomes in FL [145,146,147]. The distinct prognostic value of ctDNA, CTCs, and TMTV has been clearly demonstrated. FL patients with elevated TMTV (> 510 cm³, p = 0.0004), high CTC counts (> 0.0018 PB cells, p = 0.03), or increased cfDNA levels (> 2,550 equivalent genomes/ml, p = 0.04) showed a lower 4-year PFS. Both cfDNA and TMTV remained independently predictive of outcomes in multivariate Cox analysis, underscoring their prognostic relevance [148].
Pretreatment ctDNA levels are particularly prognostic in FL, with high levels emerging as the only independent factor associated with PFS in multivariate analysis (HR 4, 95% CI: 1.1–37, p = 0.039). Patients with elevated ctDNA before treatment had a notably poorer prognosis, with higher levels observed in those who failed to achieve CR or who experienced disease progression within 24 months (POD24), compared to patients who achieved CR or were POD24-negative (p = 0.02 and p < 0.001, respectively) [149, 150]. In a longitudinal study of 13 FL patients who achieved CR after frontline chemotherapy, persistent or re-emergent ctDNA mutations were closely linked to disease relapse, emphasizing the utility of ctDNA in monitoring for early signs of recurrence [147].
ctDNA levels have also shown promise in assessing treatment responses across different FL therapies. Among FL patients treated with anti-CD19 CAR T-cell therapy, two of four patients who were PET/CT-positive post-treatment were found to be MRD-negative by ctDNA analysis and experienced no relapse after a median follow-up of 34 months [151]. This suggests that ctDNA monitoring may complement PET/CT in confirming treatment success and assessing MRD status in FL.
Transformation from indolent FL to aggressive DLBCL often indicates a worsening prognosis [152]. Scherer et al. developed a noninvasive ctDNA-based prediction model with 83% sensitivity and 89% specificity to identify FL transformation, allowing for earlier detection. In one case, the ctDNA model captured both indolent and aggressive clones prior to clinical transformation, highlighting the ability of ctDNA to reflect tumor heterogeneity [41]. Similarly, a study by Schroers-Martin et al. using CAPP-seq on pre-diagnostic samples found that CREBBP mutations in blood could help identify patients at risk of developing FL, suggesting a potential role for ctDNA in early detection and risk stratification [153].
In MZL, ctDNA profiling through targeted NGS assays showed increases in primary ctDNA levels at disease progression, with ctDNA mutation burden decreasing in patients achieving partial remission, indicating the potential for ctDNA to serve as a marker for treatment response [154].
Central nervous system lymphoma
Minimally invasive ctDNA analysis of cerebrospinal fluid (CSF) or plasma offers significant potential for enhancing the diagnosis, surveillance, and prognosis of CNSL. Studies employing targeted NGS of ctDNA in PCNSL patients have demonstrated a 45% sensitivity for detecting mutations found in primary tumor tissue [155]. However, assessment of IgH gene rearrangements in plasma for residual disease tracking has shown limited effectiveness, with only one tracking clone detected out of four patients, highlighting the challenges of ctDNA monitoring in PCNSL [156]. Interestingly, the genetic profiles of CSF ctDNA show greater concordance with tissue findings compared to plasma ctDNA, suggesting CSF as a more reliable medium for analysis in CNSL [157, 158].
Building upon these findings, ultrasensitive ctDNA profiling detected ctDNA in 78% of plasma and all CSF samples from CNSL patients before treatment. Patients with detectable plasma ctDNA prior to treatment had significantly shorter PFS (P < 0.0001) and OS (P = 0.0001), with plasma ctDNA-based MRD monitoring effectively identifying high-risk patients (PFS, P = 0.0002; OS, P = 0.004) [159]. Additionally, a biopsy-free CNSL identification model based on ctDNA mutation patterns and burden demonstrated sensitivities of 59% in CSF and 25% in plasma, maintaining 100% specificity and positive predictive value. Moreover, for vitreoretinal lymphomas, ctDNA sequencing from aqueous humor has shown high concordance with vitreous fluid, suggesting that aqueous humor ctDNA may be a viable, noninvasive alternative to vitreous fluid for diagnosis and monitoring therapeutic response [160,161,162].
In parallel to these genetic analyses, epigenetic biomarkers in ctDNA, established for various solid tumors, are also being evaluated for CNSL. In a pilot study, two methylated markers (cg054 and SCG3) showed a sensitivity of 20% for distinguishing PCNSL from other CNS tumors [163]. Moreover, a peripheral residual disease biomarker has demonstrated high predictive value for relapse, and when integrated with clinical risk factors and radiographic response into a molecular prognostic index, it provided strong predictive power for CNSL outcomes [164].
Beyond primary CNSL, predicting CNS relapse in DLBCL remained challenging. Mutations in MYD88L265P and CD79BY196 were detectable in CSF ctDNA approximately one month before clinical diagnosis, suggesting their utility in early detection of CNS relapse in lymphoma patients [165,166,167,168]. However, as 15-20% of CNSL cases lack these mutations, negative results should be interpreted with caution. In a cohort study of 126 newly diagnosed DLBCL and 24 PCNSL cases, pretreatment CSF ctDNA demonstrated 100% sensitivity and 77.3% specificity for predicting CNS relapse when analyzed with a panel of 475 leukemia- and lymphoma-related genes [169]. Furthermore, clonotypic DNA was identified in all CSF ctDNA samples from patients with parenchymal CNS involvement and in 36% of aggressive lymphomas, indicating a 29% risk of CNS recurrence [170].
Peripheral T-cell lymphoma
ctDNA analysis has emerged as a noninvasive diagnostic and genetic profiling tool with broad clinical applications in PTCL. Detection of mutations in genes such as TET2, RHOA, DNMT3A, and IDH2 in plasma ctDNA has provided a noninvasive method for diagnosing AITL (a subtype of PTCL) [171, 172]. In ENKTL patients, plasma ctDNA testing demonstrated a sensitivity of 72.4% for detecting tumor biopsy variants [173]. Furthermore, CAPP-seq analysis of cfDNA from PTCL patients identified novel RHOA mutations, including c.73 A > G (p.Phe25Leu) and c.48 A > T (p.Cys16*), which were validated in additional tissue cohorts. This finding suggests that ctDNA sequencing can identify somatic mutations not detected in tumor genomic DNA, overcoming tumor spatial heterogeneity and providing comprehensive genotypic information [174]. The diagnostic capabilities, combined with the ability to identify novel mutations, highlight the potential of ctDNA analysis not only for initial diagnosis but also for guiding subsequent treatment strategies.
Despite chemotherapy being the standard first-line treatment for most PTCL subtypes, treatment resistance limits its efficacy. Noninvasive ctDNA monitoring offers dynamic assessment of molecular burden, treatment response, prognostic risk, and MRD. Mutations in DDX3X and KMT2D detected in ctDNA from ENKTL patients have been associated with poor PFS [175, 176]. In cases where Epstein-Barr virus (EBV) DNA was undetectable in whole blood, ctDNA mutations were identified in 7 of 14 patients, suggesting that ctDNA profiling can complement EBV DNA quantification in ENKTL monitoring [175]. Analyzing plasma ctDNA mutation profiles in 94 PTCL patients using targeted NGS revealed a significant association between post-treatment ctDNA levels and survival outcomes [177]. Moreover, tumor-specific clones were identified in 76% of patients using NGS-based TCR sequencing of ctDNA; detectable ctDNA after treatment predicted worse survival, although the prognostic significance throughout treatment was not statistically significant [178]. Another study involving 64 Chinese PTCL patients found that high pretreatment ctDNA levels were significantly associated with adverse clinical markers, and MRD negativity correlated with higher remission rates [179]. Collectively, these data indicate that ctDNA may have potential for noninvasive monitoring of treatment response and predicting outcomes in PTCL patients, with emerging evidence highlighting its role in high-risk subgroups.
In high-risk ENKTL patients, targeted NGS of tumor tissue and longitudinal plasma ctDNA showed that low pretreatment cfDNA concentrations were associated with favorable survival outcomes (1-year PFS: 90.0% vs. 36.4%; p = 0.012). Patients with rapid clearance of ctDNA mutations achieved significantly higher complete remission rates (80.0% vs. 0%; p = 0.004) and more favorable PFS (79.0% vs. 20.0%; p = 0.002) compared to those with persistent detectable mutations [173]. A phase 1b/2 study assessing ctDNA biomarkers in 38 R/R ENKTL patients treated with anti-PD-1 antibodies found that integrating plasma ctDNA with EBV DNA provided better prognostic value than either biomarker alone; notably, the presence of STAT3 mutations predicted an inferior prognosis [180]. These findings reinforce the clinical utility of ctDNA dynamics in risk stratification and treatment response assessment. However, due to the limited sample size, larger cohorts are needed to validate the predictive value of ctDNA monitoring for treatment outcomes.
Apart from tumor-specific genetic variations, epigenetic alterations in ctDNA have been investigated for diagnostic and prognostic purposes in ENKTL. A diagnostic prediction model incorporating seven ctDNA methylation markers achieved an area under the curve (AUC) of 0.988 in an independent validation cohort. By combining the seven-marker ctDNA methylation prognostic score with the prognostic index of natural killer (PINK) risk system, the PINK-C risk stratification model was developed, achieving an AUC of 0.773 in predicting prognosis [181]. The PINK-C model demonstrated distinct prognostic stratification levels. However, as these models are based on retrospective data, their specificity and sensitivity require validation in future prospective studies.
Hodgkin’s lymphoma
For cHL, ctDNA profiling with CAPP-seq has shown great promise. Specifically, ctDNA analysis identified approximately 87.5% of tumor variants present in biopsy samples from 80 newly diagnosed and 32 refractory patients, supporting ctDNA as a potential noninvasive profiling tool [76]. Notably, plasma ctDNA exhibited a higher median variant allele fraction than biopsy samples, likely reflecting the low tumor cell content typically present in cHL biopsies. This observation further underscores the utility of ctDNA in molecular profiling [34]. Certain mutations detected by ctDNA, including XPO1E571K, STAT6 and SOCS1, help distinguish cHL from other lymphoma types such as DLBCL, PMBL, ALCL and MGZL [76, 81, 182,183,184]. In addition to diagnostic insights, ctDNA profiling offers valuable information about the clonal structure and evolution in cHL. Some mutations in oncogenes and tumor suppressors, such as GNA13, XPO1, NFKBIE, IKBKB, CSF2RB, and B2M, are clonal, present across most cells, while others, like PRBM1, NOTCH2, CHD2, and BCR, appear as subclonal mutations [75]. Longitudinal ctDNA monitoring (41 samples from 13 patients) revealed that chemotherapy partially reshapes subclonal diversity, while salvage therapy with nivolumab suppresses dominant clones and promotes the emergence of new ones, thereby highlighting the utility of ctDNA in tracking clonal shifts over the course of treatment [76].
ctDNA profiling in cHL also carries prognostic significance. For example, detecting the XPO1E571K mutation via ddPCR is associated with shorter PFS, with a 2-year PFS of 57.1% in mutation-positive patients compared to 90.5% in mutation-negative patients [182]. Similarly, TP53 mutations in ctDNA correlate with inferior PFS (p = 0.0038) [75]. Baseline ctDNA levels before treatment initiation have been linked to clinical features such as elevated TMTV, higher Hasenclever international prognostic scores (≥ 3), increased LDH levels, and advanced disease stages, suggesting that baseline ctDNA may serve as a valuable supplement to traditional prognostic markers [77, 81, 185].
Moreover, the role of ctDNA in monitoring treatment response and predicting relapse has also been demonstrated in HL. Longitudinal ctDNA monitoring combined with PET/CT imaging identified disease progression in 38% of patients, with a negative predictive value of 99% when both ctDNA and PET/CT results were negative, suggesting that ctDNA may improve the predictive accuracy of PET/CT in clinical management [76]. Furthermore, in patients with advanced cHL, a 2-log reduction in ctDNA levels after two cycles of ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine) chemotherapy was predictive of CR, supporting a threshold previously validated in DLBCL [76].
The timing of ctDNA assessment during treatment has been shown to impact prognostic accuracy. Patients with high pretreatment ctDNA levels and detectable ctDNA throughout treatment (e.g., at C1D15, C3D1, and post-four cycles) experienced significantly worse PFS (p < 0.05) [34]. Molecular remission rates improved at sequential time points (C1D15, C2D1, and C3D1), with MRD negativity rates reaching 38%, 85% and 90%, respectively. Additionally, ctDNA monitoring as early as one week into treatment correlated with PET response and predicted PFS [75]. In pediatric Hodgkin lymphoma, targeted NGS showed that ctDNA was undetectable in patients who achieved an early PET response (qPET < 3), suggesting favorable outcomes [77]. Although ctDNA has shown considerable potential for clinical applications in HL, further prospective clinical trials are necessary to determine if MRD status, whether detectable or undetectable, can reliably inform decisions on treatment intensification or de-escalation.
Conclusions and perspectives
The emergence of ctDNA as a biomarker represents a major advance in lymphoma management, with applications spanning diagnosis, risk stratification, treatment monitoring, and MRD assessment. By providing comprehensive molecular insights through non-invasive sampling, ctDNA enables real-time tracking of disease burden, response to therapy, and clonal evolution, paving the way for personalized approaches to lymphoma care. Recent innovations in ctDNA analysis, including ctDNA fragmentation and methylation profiling, further expand its diagnostic and prognostic capabilities, potentially improving precision medicine in lymphoma.
The opportunities presented by ctDNA are substantial. It facilitates non-invasive genotyping, overcoming the limitations of tissue biopsies, such as spatial heterogeneity and insufficient sample material. ctDNA quantification provides a dynamic assessment of tumor burden, correlating with established prognostic factors like the IPI and TMTV. Moreover, its role in MRD detection surpasses traditional imaging techniques in sensitivity and specificity, allowing for earlier intervention upon relapse. When combined with emerging technologies such as NGS and epigenetic profiling, ctDNA can refine prognostic models and identify therapeutic targets. However, several challenges must be overcome before ctDNA can be fully integrated into routine clinical practice (Fig. 5). Standardizing preanalytical workflows remains critical, as variability in sample handling and processing can impact test accuracy. Furthermore, while ctDNA provides valuable insights, establishing optimal thresholds for clinical decision making, such as MRD or treatment response, requires further validation in large, prospective studies. In addition, the complexity of ctDNA data requires robust computational methods to integrate multiple molecular and clinical parameters. Advances in machine learning may help overcome these challenges and ultimately advance the clinical application of ctDNA as a reliable tool in lymphoma management.
Despite challenges in clinical implementation, emerging evidence supports ctDNA as a dynamic biomarker to guide lymphoma therapy [25,26,27]. Integrating ctDNA detection into clinical trial designs may improve therapeutic precision by enabling early and accurate monitoring of treatment response, MRD, and clonal evolution - potentially surpassing conventional imaging and biopsy methods. For instance, future trials in aggressive lymphomas such as DLBCL could incorporate early ctDNA monitoring to stratify patients based on their molecular response to frontline chemoimmunotherapy (e.g., R-CHOP). Patients who demonstrate rapid ctDNA clearance after one or two cycles of treatment could be candidates for de-escalated therapy, thereby reducing exposure to potentially toxic regimens without compromising efficacy. Conversely, patients with persistent ctDNA positivity could be assigned to intensified or alternative therapeutic arms incorporating novel agents (e.g., targeted therapies or immunomodulators) to overcome early resistance. This adaptive approach leverages ctDNA dynamics to inform real-time, personalized treatment decisions that may ultimately improve progression-free and overall survival.
In indolent lymphomas such as FL, which exhibit a slow yet variable course and significant clonal heterogeneity, serial ctDNA-based testing could help track clonal evolution and emerging resistance mutations during targeted therapies (e.g., PI3K inhibitors, immunomodulators) [147]. A future trial might regularly assess ctDNA profiles throughout treatment and follow-up, enabling early identification of resistance-associated genetic alterations. This, in turn, could prompt a timely therapeutic switch or the addition of combination strategies aimed at suppressing resistant clones before overt relapse. Such a personalized approach not only refines treatment decisions but also provides deeper insights into the molecular mechanisms driving disease progression and resistance.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- ABC:
-
Activated B-cell-like
- AITL:
-
Angioimmunoblastic T-cell lymphoma
- ALCL:
-
Anaplastic large cell lymphoma
- ASO-PCR:
-
Allele specific oligonucleotide polymerase chain reaction
- B-NHL:
-
B-cell non-Hodgkin lymphoma
- BEAMing:
-
Beads, emulsion, amplification and magnetics
- Bp:
-
Base pairs
- CAPP-seq:
-
Cancer personalized profiling by deep sequencing
- cfDNA:
-
Circulating cell-free DNA
- cHL:
-
Classical Hodgkin lymphoma
- CHOP:
-
Rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone
- CIRI:
-
Continuous individualized risk index
- CLL:
-
Chronic lymphocytic leukaemia
- CNS:
-
Central nervous system
- CNS:
-
Central nervous system
- CNSL:
-
Central nervous system lymphoma
- CNVs:
-
Copy number variants
- COO:
-
Cell of origin
- CR:
-
Complete response
- CSF:
-
Cerebrospinal fluid
- CT:
-
Computed tomography
- CTCL:
-
Cutaneous T-cell lymphoma
- ctDNA:
-
Circulating tumor DNA
- ddPCR:
-
Droplet digital polymerase chain reaction
- DLBCL:
-
Diffuse large B-cell lymphoma
- EFS:
-
Event-free survival
- EMR:
-
Early molecular response
- ENKTL:
-
Extranodal natural killer/T-cell lymphoma
- EPIC-seq:
-
Epigenetic expression inference from cell-free DNA sequencing
- FL3b:
-
Grade 3b follicular lymphoma
- FL:
-
Follicular lymphoma
- GCB:
-
Germinal center B-cell-like
- HGBL:
-
High grade B cell lymphoma
- HL:
-
Hodgkin Lymphoma
- HR:
-
Hazard ratio
- Ig-NGS:
-
Immunoglobulin next generation sequencing
- IgHTS:
-
Immunoglobulin high-throughput sequencing
- IPI:
-
International prognostic index
- LBCL:
-
Large B-cell lymphoma
- LDH:
-
Lactate dehydrogenase
- MCL:
-
Mantle cell lymphoma
- MMR:
-
Major molecular response
- MRD:
-
Molecular residual disease
- MZL:
-
Marginal zone lymphoma
- NGS:
-
Next generation sequencing
- NK:
-
Natural killer
- OS:
-
Overall survival
- PCNSL:
-
Primary central nervous system lymphoma
- PCR:
-
Polymerase chain reaction
- PET/CT:
-
Positron emission tomography/computed tomography
- PFS:
-
Progression-free survival
- PhasED-seq:
-
Phased variant enrichment and detection sequencing
- PHL:
-
Pediatric Hodgkin lymphoma
- PINK:
-
Prognostic index of natural killer
- PMBCL:
-
Primary mediastinal B-cell lymphoma
- PMBL:
-
Primary mediastinal B-cell lymphoma
- POD24:
-
Progression within 24 months.
- PTCL-NOS:
-
PTCL not otherwise specified
- PTCL:
-
Peripheral T-cell lymphoma
- PTLD:
-
Posttransplant lymphoproliferative disorders
- R/R:
-
Relapsed/refractory
- RS:
-
Richter’s syndrome
- RT-PCR:
-
Real time polymerase chain reaction
- SNVs:
-
Single nucleotide variants
- T-NHL:
-
T-cell non-Hodgkin lymphoma
- TCR:
-
T-cell receptor
- TFHL:
-
T follicular helper lymphoma
- tFL:
-
Transformed follicular lymphoma
- THRLBCL:
-
T-cell/histiocyte-rich large B-cell lymphoma
- TMTV:
-
Total metabolic tumor volume
- VDJ:
-
Variable, diversity, joining
- WES:
-
Whole exome sequencing
- WGBS:
-
Whole genome bisulfite sequencing
- WGS:
-
Whole genome sequencing
- WHO:
-
World health organization
References
Global Burden of Disease, Cancer C, Fitzmaurice C, Akinyemiju TF, Al Lami FH, Alam T, Alizadeh-Navaei R, Allen C, Alsharif U, Alvis-Guzman N, Amini E, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 1990 to 2016: a systematic analysis for the global burden of disease study. JAMA Oncol. 2018;4(11):1553–68.
Alaggio R, Amador C, Anagnostopoulos I, Attygalle AD, Araujo IBO, Berti E, Bhagat G, Borges AM, Boyer D, Calaminici M, et al. The 5th edition of the World Health Organization classification of haematolymphoid tumours: lymphoid neoplasms. Leukemia. 2022;36(7):1720–48.
Wang L, Qin W, Huo YJ, Li X, Shi Q, Rasko JEJ, Janin A, Zhao WL. Advances in targeted therapy for malignant lymphoma. Signal Transduct Target Ther. 2020;5(1):15.
Chaudhari K, Rizvi S, Syed BA. Non-hodgkin lymphoma therapy landscape. Nat Rev Drug Discov. 2019;18(9):663–4.
Younes A, Ansell S, Fowler N, Wilson W, de Vos S, Seymour J, Advani R, Forero A, Morschhauser F, Kersten MJ, et al. The landscape of new drugs in lymphoma. Nat Rev Clin Oncol. 2017;14(6):335–46.
de Leval L, Alizadeh AA, Bergsagel PL, Campo E, Davies A, Dogan A, Fitzgibbon J, Horwitz SM, Melnick AM, Morice WG, et al. Genomic profiling for clinical decision making in lymphoid neoplasms. Blood. 2022;140(21):2193–227.
Tun AM, Ansell SM. Immunotherapy in Hodgkin and non-hodgkin lymphoma: innate, adaptive and targeted immunological strategies. Cancer Treat Rev. 2020;88:102042.
Susanibar-Adaniya S, Barta SK. 2021 update on diffuse large B cell lymphoma: a review of current data and potential applications on risk stratification and management. Am J Hematol. 2021;96(5):617–29.
Mohty R, Dulery R, Bazarbachi AH, Savani M, Hamed RA, Bazarbachi A, Mohty M. Latest advances in the management of classical Hodgkin lymphoma: the era of novel therapies. Blood Cancer J. 2021;11(7):126.
Cheson BD, Nowakowski G, Salles G. Diffuse large B-cell lymphoma: new targets and novel therapies. Blood Cancer J. 2021;11(4):68.
Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977;37(3):646–50.
Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61(4):1659–65.
El Messaoudi S, Rolet F, Mouliere F, Thierry AR. Circulating cell free DNA: preanalytical considerations. Clin Chim Acta. 2013;424:222–30.
Mouliere F, Chandrananda D, Piskorz AM, Moore EK, Morris J, Ahlborn LB, Mair R, Goranova T, Marass F, Heider K et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med. 2018;10(466).
Kustanovich A, Schwartz R, Peretz T, Grinshpun A. Life and death of circulating cell-free DNA. Cancer Biol Ther. 2019;20(8):1057–67.
Sun K, Jiang P, Chan KC, Wong J, Cheng YK, Liang RH, Chan WK, Ma ES, Chan SL, Cheng SH, et al. Plasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessments. Proc Natl Acad Sci U S A. 2015;112(40):E5503–5512.
Han D, Li R, Shi J, Tan P, Zhang R, Li J. Liquid biopsy for infectious diseases: a focus on microbial cell-free DNA sequencing. Theranostics. 2020;10(12):5501–13.
Park SY, Chang EJ, Ledeboer N, Messacar K, Lindner MS, Venkatasubrahmanyam S, Wilber JC, Vaughn ML, Bercovici S, Perkins BA, et al. Plasma microbial cell-free DNA sequencing from over 15,000 patients identified a broad spectrum of pathogens. J Clin Microbiol. 2023;61(8):e0185522.
Gao Q, Zeng Q, Wang Z, Li C, Xu Y, Cui P, Zhu X, Lu H, Wang G, Cai S, et al. Circulating cell-free DNA for cancer early detection. Innov (Camb). 2022;3(4):100259.
Taglauer ES, Wilkins-Haug L, Bianchi DW. Review: cell-free fetal DNA in the maternal circulation as an indication of placental health and disease. Placenta. 2014;35(SupplSuppl):S64–68.
Kananen L, Hurme M, Burkle A, Moreno-Villanueva M, Bernhardt J, Debacq-Chainiaux F, Grubeck-Loebenstein B, Malavolta M, Basso A, Piacenza F, et al. Circulating cell-free DNA in health and disease - the relationship to health behaviours, ageing phenotypes and metabolomics. Geroscience. 2023;45(1):85–103.
Moss J, Magenheim J, Neiman D, Zemmour H, Loyfer N, Korach A, Samet Y, Maoz M, Druid H, Arner P, et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun. 2018;9(1):5068.
Liu MC, Oxnard GR, Klein EA, Swanton C, Seiden MV, Consortium C. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann Oncol. 2020;31(6):745–59.
Borchmann S. Liquid biopsy in Hodgkin lymphoma - moving beyond the proof of principle. Br J Haematol. 2021;195(4):493–4.
Huang Z, Fu Y, Yang H, Zhou Y, Shi M, Li Q, Liu W, Liang J, Zhu L, Qin S, et al. Liquid biopsy in T-cell lymphoma: biomarker detection techniques and clinical application. Mol Cancer. 2024;23(1):36.
Rossi D, Spina V, Bruscaggin A, Gaidano G. Liquid biopsy in lymphoma. Haematologica. 2019;104(4):648–52.
Roschewski M, Rossi D, Kurtz DM, Alizadeh AA, Wilson WH. Circulating tumor DNA in lymphoma: principles and future directions. Blood Cancer Discov. 2022;3(1):5–15.
Armitage JO, Gascoyne RD, Lunning MA, Cavalli F. Non-hodgkin lymphoma. Lancet. 2017;390(10091):298–310.
Silkenstedt E, Salles G, Campo E, Dreyling M. B-cell non-hodgkin lymphomas. Lancet. 2024.
Miao Y, Medeiros LJ, Li Y, Li J, Young KH. Genetic alterations and their clinical implications in DLBCL. Nat Rev Clin Oncol. 2019;16(10):634–52.
A clinical evaluation of the International Lymphoma Study Group. Classification of non-hodgkin’s lymphoma. The Non-hodgkin’s lymphoma classification project. Blood. 1997;89(11):3909–18.
Rossi D, Diop F, Spaccarotella E, Monti S, Zanni M, Rasi S, Deambrogi C, Spina V, Bruscaggin A, Favini C, et al. Diffuse large B-cell lymphoma genotyping on the liquid biopsy. Blood. 2017;129(14):1947–57.
Zou H, Liu W, Wang X, Wang Y, Wang C, Qiu C, Liu H, Shan D, Xie T, Huang W et al. Dynamic monitoring of circulating tumor DNA reveals outcomes and genomic alterations in patients with relapsed or refractory large B-cell lymphoma undergoing CAR T-cell therapy. J Immunother Cancer. 2024;12(3).
Alig SK, Shahrokh Esfahani M, Garofalo A, Li MY, Rossi C, Flerlage T, Flerlage JE, Adams R, Binkley MS, Shukla N, et al. Distinct Hodgkin lymphoma subtypes defined by noninvasive genomic profiling. Nature. 2024;625(7996):778–87.
Reddy A, Zhang J, Davis NS, Moffitt AB, Love CL, Waldrop A, Leppa S, Pasanen A, Meriranta L, Karjalainen-Lindsberg ML, et al. Genetic and functional drivers of diffuse large B cell lymphoma. Cell. 2017;171(2):481–e494415.
Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503–11.
Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI, Gascoyne RD, Muller-Hermelink HK, Smeland EB, Giltnane JM, et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med. 2002;346(25):1937–47.
Chapuy B, Stewart C, Dunford AJ, Kim J, Kamburov A, Redd RA, Lawrence MS, Roemer MGM, Li AJ, Ziepert M, et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med. 2018;24(5):679–90.
Schmitz R, Wright GW, Huang DW, Johnson CA, Phelan JD, Wang JQ, Roulland S, Kasbekar M, Young RM, Shaffer AL, et al. Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med. 2018;378(15):1396–407.
Wright GW, Huang DW, Phelan JD, Coulibaly ZA, Roulland S, Young RM, Wang JQ, Schmitz R, Morin RD, Tang J, et al. A probabilistic classification Tool for genetic subtypes of diffuse large B cell lymphoma with therapeutic implications. Cancer Cell. 2020;37(4):551–e568514.
Scherer F, Kurtz DM, Newman AM, Stehr H, Craig AF, Esfahani MS, Lovejoy AF, Chabon JJ, Klass DM, Liu CL, et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016;8(364):364ra155.
Esfahani MS, Hamilton EG, Mehrmohamadi M, Nabet BY, Alig SK, King DA, Steen CB, Macaulay CW, Schultz A, Nesselbush MC, et al. Inferring gene expression from cell-free DNA fragmentation profiles. Nat Biotechnol. 2022;40(4):585–97.
Bratman SV, Newman AM, Alizadeh AA, Diehn M. Potential clinical utility of ultrasensitive circulating tumor DNA detection with CAPP-Seq. Expert Rev Mol Diagn. 2015;15(6):715–9.
Kridel R, Meissner B, Rogic S, Boyle M, Telenius A, Woolcock B, Gunawardana J, Jenkins CE, Cochrane C, Ben-Neriah S, et al. Whole transcriptome sequencing reveals recurrent NOTCH1 mutations in mantle cell lymphoma. Blood. 2012;119(9):1963–71.
Pararajalingam P, Coyle KM, Arthur SE, Thomas N, Alcaide M, Meissner B, Boyle M, Qureshi Q, Grande BM, Rushton C, et al. Coding and noncoding drivers of mantle cell lymphoma identified through exome and genome sequencing. Blood. 2020;136(5):572–84.
Zhang J, Jima D, Moffitt AB, Liu Q, Czader M, Hsi ED, Fedoriw Y, Dunphy CH, Richards KL, Gill JI, et al. The genomic landscape of mantle cell lymphoma is related to the epigenetically determined chromatin state of normal B cells. Blood. 2014;123(19):2988–96.
Love C, Sun Z, Jima D, Li G, Zhang J, Miles R, Richards KL, Dunphy CH, Choi WW, Srivastava G, et al. The genetic landscape of mutations in Burkitt lymphoma. Nat Genet. 2012;44(12):1321–5.
Richter J, Schlesner M, Hoffmann S, Kreuz M, Leich E, Burkhardt B, Rosolowski M, Ammerpohl O, Wagener R, Bernhart SH, et al. Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing. Nat Genet. 2012;44(12):1316–20.
Schmitz R, Young RM, Ceribelli M, Jhavar S, Xiao W, Zhang M, Wright G, Shaffer AL, Hodson DJ, Buras E, et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature. 2012;490(7418):116–20.
Thomas N, Dreval K, Gerhard DS, Hilton LK, Abramson JS, Ambinder RF, Barta S, Bartlett NL, Bethony J, Bhatia K, et al. Genetic subgroups inform on pathobiology in adult and pediatric Burkitt lymphoma. Blood. 2023;141(8):904–16.
Grande BM, Gerhard DS, Jiang A, Griner NB, Abramson JS, Alexander TB, Allen H, Ayers LW, Bethony JM, Bhatia K, et al. Genome-wide discovery of somatic coding and noncoding mutations in pediatric endemic and sporadic Burkitt lymphoma. Blood. 2019;133(12):1313–24.
Yi S, Yan Y, Jin M, Bhattacharya S, Wang Y, Wu Y, Yang L, Gine E, Clot G, Chen L et al. Genomic and transcriptomic profiling reveals distinct molecular subsets associated with outcomes in mantle cell lymphoma. J Clin Invest. 2022;132(3).
Tsujimoto Y, Cossman J, Jaffe E, Croce CM. Involvement of the bcl-2 gene in human follicular lymphoma. Science. 1985;228(4706):1440–3.
Schuler F, Dolken L, Hirt C, Kiefer T, Berg T, Fusch G, Weitmann K, Hoffmann W, Fusch C, Janz S, et al. Prevalence and frequency of circulating t(14;18)-MBR translocation carrying cells in healthy individuals. Int J Cancer. 2009;124(4):958–63.
Green MR. Chromatin modifying gene mutations in follicular lymphoma. Blood. 2018;131(6):595–604.
Morin RD, Mendez-Lago M, Mungall AJ, Goya R, Mungall KL, Corbett RD, Johnson NA, Severson TM, Chiu R, Field M, et al. Frequent mutation of histone-modifying genes in non-hodgkin lymphoma. Nature. 2011;476(7360):298–303.
Isaacson PG, Du MQ. MALT lymphoma: from morphology to molecules. Nat Rev Cancer. 2004;4(8):644–53.
Zucca E, Arcaini L, Buske C, Johnson PW, Ponzoni M, Raderer M, Ricardi U, Salar A, Stamatopoulos K, Thieblemont C, et al. Marginal zone lymphomas: ESMO Clinical Practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020;31(1):17–29.
Bühler MM, Martin-Subero JI, Pan-Hammarström Q, Campo E, Rosenquist R. Towards precision medicine in lymphoid malignancies. J Intern Med. 2022;292(2):221–42.
Dreval K, Hilton LK, Cruz M, Shaalan H, Ben-Neriah S, Boyle M, Collinge B, Coyle KM, Duns G, Farinha P, et al. Genetic subdivisions of follicular lymphoma defined by distinct coding and noncoding mutation patterns. Blood. 2023;142(6):561–73.
Fernández-Miranda I, Pedrosa L, González-Rincón J, Espinet B, de la Cruz Vicente F, Climent F, Gómez S, Royuela A, Camacho FI, Martín-Acosta P, et al. Generation and external validation of a histologic transformation risk model for patients with Follicular lymphoma. Mod Pathol. 2024;37(7):100516.
Enemark MH, Hemmingsen JK, Jensen ML, Kridel R, Ludvigsen M. Molecular biomarkers in prediction of high-grade transformation and outcome in patients with follicular lymphoma: a comprehensive systemic review. Int J Mol Sci. 2024;25(20).
Zhou Y, Liu W, Xu Z, Zhu H, Xiao D, Su W, Zeng R, Feng Y, Duan Y, Zhou J, et al. Analysis of genomic alteration in primary central nervous system lymphoma and the expression of some related genes. Neoplasia. 2018;20(10):1059–69.
Radke J, Ishaque N, Koll R, Gu Z, Schumann E, Sieverling L, Uhrig S, Hübschmann D, Toprak UH, López C, et al. The genomic and transcriptional landscape of primary central nervous system lymphoma. Nat Commun. 2022;13(1):2558.
Hernández-Verdin I, Kirasic E, Wienand K, Mokhtari K, Eimer S, Loiseau H, Rousseau A, Paillassa J, Ahle G, Lerintiu F, et al. Molecular and clinical diversity in primary central nervous system lymphoma. Ann Oncol. 2023;34(2):186–99.
Bellei M, Chiattone CS, Luminari S, Pesce EA, Cabrera ME, de Souza CA, Gabus R, Zoppegno L, Zoppegno L, Milone J, et al. T-cell lymphomas in South America and europe. Rev Bras Hematol Hemoter. 2012;34(1):42–7.
Ji MM, Huang YH, Huang JY, Wang ZF, Fu D, Liu H, Liu F, Leboeuf C, Wang L, Ye J, et al. Histone modifier gene mutations in peripheral T-cell lymphoma not otherwise specified. Haematologica. 2018;103(4):679–87.
Sakata-Yanagimoto M, Enami T, Yoshida K, Shiraishi Y, Ishii R, Miyake Y, Muto H, Tsuyama N, Sato-Otsubo A, Okuno Y, et al. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46(2):171–5.
Odejide O, Weigert O, Lane AA, Toscano D, Lunning MA, Kopp N, Kim S, van Bodegom D, Bolla S, Schatz JH, et al. A targeted mutational landscape of angioimmunoblastic T-cell lymphoma. Blood. 2014;123(9):1293–6.
Jiang L, Gu ZH, Yan ZX, Zhao X, Xie YY, Zhang ZG, Pan CM, Hu Y, Cai CP, Dong Y, et al. Exome sequencing identifies somatic mutations of DDX3X in natural killer/T-cell lymphoma. Nat Genet. 2015;47(9):1061–6.
Crescenzo R, Abate F, Lasorsa E, Tabbo F, Gaudiano M, Chiesa N, Di Giacomo F, Spaccarotella E, Barbarossa L, Ercole E, et al. Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma. Cancer Cell. 2015;27(4):516–32.
Huang YH, Qiu YR, Zhang QL, Cai MC, Yu H, Zhang JM, Jiang L, Ji MM, Xu PP, Wang L, et al. Genomic and transcriptomic profiling of peripheral T cell lymphoma reveals distinct molecular and microenvironment subtypes. Cell Rep Med. 2024;5(2):101416.
Re D, Kuppers R, Diehl V. Molecular pathogenesis of Hodgkin’s lymphoma. J Clin Oncol. 2005;23(26):6379–86.
Maco M, Kupcova K, Herman V, Ondeckova I, Kozak T, Mocikova H, Havranek O. Czech Hodgkin lymphoma study G: circulating tumor DNA in Hodgkin lymphoma. Ann Hematol. 2022;101(11):2393–403.
Sobesky S, Mammadova L, Cirillo M, Drees EEE, Mattlener J, Dörr H, Altmüller J, Shi Z, Bröckelmann PJ, Weiss J, et al. In-depth cell-free DNA sequencing reveals genomic landscape of Hodgkin’s lymphoma and facilitates ultrasensitive residual disease detection. Med. 2021;2(10):1171–e11931111.
Spina V, Bruscaggin A, Cuccaro A, Martini M, Di Trani M, Forestieri G, Manzoni M, Condoluci A, Arribas A, Terzi-Di-Bergamo L, et al. Circulating tumor DNA reveals genetics, clonal evolution, and residual disease in classical Hodgkin lymphoma. Blood. 2018;131(22):2413–25.
Desch AK, Hartung K, Botzen A, Brobeil A, Rummel M, Kurch L, Georgi T, Jox T, Bielack S, Burdach S, et al. Genotyping circulating tumor DNA of pediatric Hodgkin lymphoma. Leukemia. 2020;34(1):151–66.
Wienand K, Chapuy B, Stewart C, Dunford AJ, Wu D, Kim J, Kamburov A, Wood TR, Cader FZ, Ducar MD, et al. Genomic analyses of flow-sorted Hodgkin Reed-Sternberg cells reveal complementary mechanisms of immune evasion. Blood Adv. 2019;3(23):4065–80.
Tiacci E, Ladewig E, Schiavoni G, Penson A, Fortini E, Pettirossi V, Wang Y, Rosseto A, Venanzi A, Vlasevska S, et al. Pervasive mutations of JAK-STAT pathway genes in classical Hodgkin lymphoma. Blood. 2018;131(22):2454–65.
Reichel J, Chadburn A, Rubinstein PG, Giulino-Roth L, Tam W, Liu Y, Gaiolla R, Eng K, Brody J, Inghirami G, et al. Flow sorting and exome sequencing reveal the oncogenome of primary Hodgkin and Reed-Sternberg cells. Blood. 2015;125(7):1061–72.
Camus V, Viennot M, Lequesne J, Viailly PJ, Bohers E, Bessi L, Marcq B, Etancelin P, Dubois S, Picquenot JM, et al. Targeted genotyping of circulating tumor DNA for classical Hodgkin lymphoma monitoring: a prospective study. Haematologica. 2021;106(1):154–62.
Maura F, Ziccheddu B, Xiang JZ, Bhinder B, Rosiene J, Abascal F, Maclachlan KH, Eng KW, Uppal M, He F, et al. Molecular evolution of classic Hodgkin lymphoma revealed through whole-genome sequencing of Hodgkin and Reed Sternberg cells. Blood Cancer Discov. 2023;4(3):208–27.
Hartmann S, Schuhmacher B, Rausch T, Fuller L, Doring C, Weniger M, Lollies A, Weiser C, Thurner L, Rengstl B, et al. Highly recurrent mutations of SGK1, DUSP2 and JUNB in nodular lymphocyte predominant Hodgkin lymphoma. Leukemia. 2016;30(4):844–53.
Schumacher MA, Schmitz R, Brune V, Tiacci E, Doring C, Hansmann ML, Siebert R, Kuppers R. Mutations in the genes coding for the NF-kappaB regulating factors IkappaBalpha and A20 are uncommon in nodular lymphocyte-predominant Hodgkin’s lymphoma. Haematologica. 2010;95(1):153–7.
Lauer EM, Mutter J, Scherer F. Circulating tumor DNA in B-cell lymphoma: technical advances, clinical applications, and perspectives for translational research. Leukemia. 2022;36(9):2151–64.
Schroers-Martin JG, Alig S, Garofalo A, Tessoulin B, Sugio T, Alizadeh AA. Molecular monitoring of lymphomas. Annu Rev Pathol. 2023;18:149–80.
Jimenez C, Chillon Mdel C, Balanzategui A, Puig N, Sebastian E, Alcoceba M, Sarasquete ME, Conde IP, Corral R, Marin LA, et al. Detection of MYD88 L265P mutation by real-time allele-specific oligonucleotide polymerase chain reaction. Appl Immunohistochem Mol Morphol. 2014;22(10):768–73.
Huet S, Salles G. Potential of circulating tumor DNA for the management of patients with lymphoma. JCO Oncol Pract. 2020;16(9):561–8.
Dressman D, Yan H, Traverso G, Kinzler KW, Vogelstein B. Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations. Proc Natl Acad Sci U S A. 2003;100(15):8817–22.
Billadeau D, Blackstadt M, Greipp P, Kyle RA, Oken MM, Kay N, Van Ness B. Analysis of B-lymphoid malignancies using allele-specific polymerase chain reaction: a technique for sequential quantitation of residual disease. Blood. 1991;78(11):3021–9.
Galimberti S, Balducci S, Guerrini F, Del Re M, Cacciola R. Digital droplet PCR in hematologic malignancies: a new useful molecular tool. Diagnostics (Basel). 2022;12(6).
Khagi Y, Goodman AM, Daniels GA, Patel SP, Sacco AG, Randall JM, Bazhenova LA, Kurzrock R. Hypermutated circulating tumor DNA: correlation with response to checkpoint inhibitor-based immunotherapy. Clin Cancer Res. 2017;23(19):5729–36.
Chen Y, De Spiegelaere W, Trypsteen W, Gleerup D, Vandesompele J, Lievens A, Vynck M, Thas O. Benchmarking digital PCR partition classification methods with empirical and simulated duplex data. Brief Bioinform. 2024;25(3).
Yin CC, Luthra R. Molecular detection of t(11;14)(q13;q32) in mantle cell lymphoma. Methods Mol Biol. 2013;999:211–6.
Chen K, Ma Y, Ding T, Zhang X, Chen B, Guan M. Effectiveness of digital PCR for MYD88(L265P) detection in vitreous fluid for primary central nervous system lymphoma diagnosis. Exp Ther Med. 2020;20(1):301–8.
Henriksen TV, Drue SO, Frydendahl A, Demuth C, Rasmussen MH, Reinert T, Pedersen JS, Andersen CL. Error characterization and statistical modeling improves circulating tumor DNA detection by droplet digital PCR. Clin Chem. 2022;68(5):657–67.
Kurtz DM, Soo J, Co Ting Keh L, Alig S, Chabon JJ, Sworder BJ, Schultz A, Jin MC, Scherer F, Garofalo A, et al. Enhanced detection of minimal residual disease by targeted sequencing of phased variants in circulating tumor DNA. Nat Biotechnol. 2021;39(12):1537–47.
McGuire AL, Caulfield T, Cho MK. Research ethics and the challenge of whole-genome sequencing. Nat Rev Genet. 2008;9(2):152–6.
Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, Liu CL, Neal JW, Wakelee HA, Merritt RE, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20(5):548–54.
Wu D, Emerson RO, Sherwood A, Loh ML, Angiolillo A, Howie B, Vogt J, Rieder M, Kirsch I, Carlson C, et al. Detection of minimal residual disease in B lymphoblastic leukemia by high-throughput sequencing of IGH. Clin Cancer Res. 2014;20(17):4540–8.
Hung SS, Meissner B, Chavez EA, Ben-Neriah S, Ennishi D, Jones MR, Shulha HP, Chan FC, Boyle M, Kridel R, et al. Assessment of capture and amplicon-based approaches for the development of a targeted next-generation sequencing pipeline to personalize lymphoma management. J Mol Diagn. 2018;20(2):203–14.
Dubois S, Viailly PJ, Mareschal S, Bohers E, Bertrand P, Ruminy P, Maingonnat C, Jais JP, Peyrouze P, Figeac M, et al. Next-generation sequencing in diffuse large B-cell lymphoma highlights molecular divergence and therapeutic opportunities: a LYSA study. Clin Cancer Res. 2016;22(12):2919–28.
Faham M, Zheng J, Moorhead M, Carlton VE, Stow P, Coustan-Smith E, Pui CH, Campana D. Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia. Blood. 2012;120(26):5173–80.
Kurtz DM, Green MR, Bratman SV, Scherer F, Liu CL, Kunder CA, Takahashi K, Glover C, Keane C, Kihira S, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood. 2015;125(24):3679–87.
Lakhotia R, Roschewski M. Circulating tumour DNA in B-cell lymphomas: current state and future prospects. Br J Haematol. 2021;193(5):867–81.
Newman AM, Lovejoy AF, Klass DM, Kurtz DM, Chabon JJ, Scherer F, Stehr H, Liu CL, Bratman SV, Say C, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol. 2016;34(5):547–55.
Kurtz DM, Scherer F, Jin MC, Soo J, Craig AFM, Esfahani MS, Chabon JJ, Stehr H, Liu CL, Tibshirani R, et al. Circulating tumor DNA measurements as early outcome predictors in diffuse large B-cell lymphoma. J Clin Oncol. 2018;36(28):2845–53.
Chen C, Liu Z, Zhou W, Tian H, Huang J, Yuan H, Yu H. Comparison of the fermentation activities and volatile flavor profiles of Chinese rice wine fermented using an artificial starter, a traditional JIUYAO and a commercial starter. Front Microbiol. 2021;12:716281.
Laurini JA, Aoun P, Iqbal J, Chan W, Greiner TC. Investigation of the BRAF V600E mutation by pyrosequencing in lymphoproliferative disorders. Am J Clin Pathol. 2012;138(6):877–83.
Larose H, Prokoph N, Matthews JD, Schlederer M, Hogler S, Alsulami AF, Ducray SP, Nuglozeh E, Fazaludeen FMS, Elmouna A, et al. Whole exome sequencing reveals NOTCH1 mutations in anaplastic large cell lymphoma and points to Notch both as a key pathway and a potential therapeutic target. Haematologica. 2021;106(6):1693–704.
Nakamura T, Yamashita S, Fukumura K, Nakabayashi J, Tanaka K, Tamura K, Tateishi K, Kinoshita M, Fukushima S, Takami H, et al. Genome-wide DNA methylation profiling identifies primary central nervous system lymphoma as a distinct entity different from systemic diffuse large B-cell lymphoma. Acta Neuropathol. 2017;133(2):321–4.
Kristensen LS, Hansen JW, Kristensen SS, Tholstrup D, Harslof LB, Pedersen OB, De Nully Brown P, Gronbaek K. Aberrant methylation of cell-free circulating DNA in plasma predicts poor outcome in diffuse large B cell lymphoma. Clin Epigenetics. 2016;8(1):95.
Rivas-Delgado A, Nadeu F, Enjuanes A, Casanueva-Eliceiry S, Mozas P, Magnano L, Castrejón de Anta N, Rovira J, Dlouhy I, Martín S, et al. Mutational landscape and tumor burden assessed by cell-free DNA in diffuse large B-cell lymphoma in a population-based study. Clin Cancer Res. 2021;27(2):513–21.
Daigle S, McDonald AA, Morschhauser F, Salles G, Ribrag V, McKay P, Tilly H, Schmitt A, Gerecitano J, Fruchart C, et al. Discovery of candidate predictors of response to Tazemetostat in diffuse large B-cell lymphoma and follicular lymphoma using NGS technology on ctDNA samples collected Pre-treatment. Blood. 2017;130(Supplement 1):4013–4013.
Kurtz DM, Scherer F, Newman AM, Craig A, Jin M, Stehr H, Chabon JJ, Esfahani M, Liu CL, Zhou L, et al. Noninvasive detection of BCL2, BCL6, and MYC Translocations in diffuse large B-cell lymphoma. Blood. 2016;128(22):2930–2930.
Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, Lih CJ, Williams PM, Shaffer AL, Gerecitano J, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21(8):922–6.
Sworder BJ, Kurtz DM, Alig SK, Frank MJ, Shukla N, Garofalo A, Macaulay CW, Shahrokh Esfahani M, Olsen MN, Hamilton J, et al. Determinants of resistance to engineered T cell therapies targeting CD19 in large B cell lymphomas. Cancer Cell. 2023;41(1):210–e225215.
Bruscaggin A, di Bergamo LT, Spina V, Hodkinson B, Forestieri G, Bonfiglio F, Condoluci A, Wu W, Pirosa MC, Faderl MR, et al. Circulating tumor DNA for comprehensive noninvasive monitoring of lymphoma treated with ibrutinib plus nivolumab. Blood Adv. 2021;5(22):4674–85.
Zhang S, Zhang T, Liu H, Zhao J, Zhou H, Su X, Liu X, Li L, Qiu L, Qian Z, et al. Tracking the evolution of untreated high-intermediate/high-risk diffuse large B-cell lymphoma by circulating tumour DNA. Br J Haematol. 2022;196(3):617–28.
Roschewski M, Dunleavy K, Pittaluga S, Moorhead M, Pepin F, Kong K, Shovlin M, Jaffe ES, Staudt LM, Lai C, et al. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study. Lancet Oncol. 2015;16(5):541–9.
Frank MJ, Hossain NM, Bukhari A, Dean E, Spiegel JY, Claire GK, Kirsch I, Jacob AP, Mullins CD, Lee LW, et al. Monitoring of circulating tumor DNA improves early relapse detection after axicabtagene ciloleucel infusion in large B-cell lymphoma: results of a prospective multi-institutional trial. J Clin Oncol. 2021;39(27):3034–43.
Meriranta L, Alkodsi A, Pasanen A, Lepistö M, Mapar P, Blaker YN, Jørgensen J, Karjalainen-Lindsberg ML, Fiskvik I, Mikalsen LTG, et al. Molecular features encoded in the ctDNA reveal heterogeneity and predict outcome in high-risk aggressive B-cell lymphoma. Blood. 2022;139(12):1863–77.
Li M, Ding N, Mi L, Shi Y, Du X, Yi Y, Yang L, Liu W, Zhu J. Liquid biopsy in diffuse large B-cell lymphoma: utility in cell origin determination and survival prediction in Chinese patients. Leuk Lymphoma. 2022;63(3):608–17.
Alig S, Macaulay CW, Kurtz DM, Dührsen U, Hüttmann A, Schmitz C, Jin MC, Sworder BJ, Garofalo A, Shahrokh Esfahani M, et al. Short diagnosis-to-treatment interval is associated with higher circulating tumor DNA levels in diffuse large B-Cell lymphoma. J Clin Oncol. 2021;39(23):2605–16.
Casasnovas RO, Ysebaert L, Thieblemont C, Bachy E, Feugier P, Delmer A, Tricot S, Gabarre J, Andre M, Fruchart C, et al. FDG-PET-driven consolidation strategy in diffuse large B-cell lymphoma: final results of a randomized phase 2 study. Blood. 2017;130(11):1315–26.
Hertzberg M, Gandhi MK, Trotman J, Butcher B, Taper J, Johnston A, Gill D, Ho SJ, Cull G, Fay K, et al. Early treatment intensification with R-ICE and 90Y-ibritumomab tiuxetan (Zevalin)-BEAM stem cell transplantation in patients with high-risk diffuse large B-cell lymphoma patients and positive interim PET after 4 cycles of R-CHOP-14. Haematologica. 2017;102(2):356–63.
Leonard JP, Kolibaba KS, Reeves JA, Tulpule A, Flinn IW, Kolevska T, Robles R, Flowers CR, Collins R, DiBella NJ, et al. Randomized phase II study of R-CHOP with or without Bortezomib in previously untreated patients with non-germinal center B-cell-like diffuse large B-cell lymphoma. J Clin Oncol. 2017;35(31):3538–46.
Moskowitz CH, Schöder H, Teruya-Feldstein J, Sima C, Iasonos A, Portlock CS, Straus D, Noy A, Palomba ML, O’Connor OA, et al. Risk-adapted dose-dense immunochemotherapy determined by interim FDG-PET in advanced-stage diffuse large B-cell lymphoma. J Clin Oncol. 2010;28(11):1896–903.
Nowakowski GS, Chiappella A, Gascoyne RD, Scott DW, Zhang Q, Jurczak W, Özcan M, Hong X, Zhu J, Jin J, et al. ROBUST: a phase III study of Lenalidomide plus R-CHOP Versus Placebo Plus R-CHOP in previously untreated patients with ABC-Type diffuse large B-Cell lymphoma. J Clin Oncol. 2021;39(12):1317–28.
Kovacs G, Robrecht S, Fink AM, Bahlo J, Cramer P, von Tresckow J, Maurer C, Langerbeins P, Fingerle-Rowson G, Ritgen M, et al. Minimal residual disease assessment improves prediction of outcome in patients with chronic lymphocytic leukemia (CLL) who achieve partial response: comprehensive analysis of two phase III studies of the German CLL Study Group. J Clin Oncol. 2016;34(31):3758–65.
Kwok M, Rawstron AC, Varghese A, Evans PA, O’Connor SJ, Doughty C, Newton DJ, Moreton P, Hillmen P. Minimal residual disease is an independent predictor for 10-year survival in CLL. Blood. 2016;128(24):2770–3.
Li M, Mi L, Wang C, Wang X, Zhu J, Qi F, Yu H, Ye Y, Wang D, Cao J, et al. Clinical implications of circulating tumor DNA in predicting the outcome of diffuse large B cell lymphoma patients receiving first-line therapy. BMC Med. 2022;20(1):369.
Kurtz DM, Esfahani MS, Scherer F, Soo J, Jin MC, Liu CL, Newman AM, Dührsen U, Hüttmann A, Casasnovas O, et al. Dynamic risk profiling using serial tumor biomarkers for personalized outcome prediction. Cell. 2019;178(3):699–e713619.
Alcoceba M, Stewart JP, García-Álvarez M, Díaz LG, Jiménez C, Medina A, Chillón MC, Gazdova J, Blanco O, Díaz FJ, et al. Liquid biopsy for molecular characterization of diffuse large B-cell lymphoma and early assessment of minimal residual disease. Br J Haematol. 2024;205(1):109–21.
Herrera AF, Tracy S, Croft B, Opat S, Ray J, Lovejoy AF, Musick L, Paulson JN, Sehn LH, Jiang Y. Risk profiling of patients with relapsed/refractory diffuse large B-cell lymphoma by measuring circulating tumor DNA. Blood Adv. 2022;6(6):1651–60.
Assouline SE, Nielsen TH, Yu S, Alcaide M, Chong L, MacDonald D, Tosikyan A, Kukreti V, Kezouh A, Petrogiannis-Haliotis T, et al. Phase 2 study of panobinostat with or without rituximab in relapsed diffuse large B-cell lymphoma. Blood. 2016;128(2):185–94.
Thompson CA, Ghesquieres H, Maurer MJ, Cerhan JR, Biron P, Ansell SM, Chassagne-Clément C, Inwards DJ, Gargi T, Johnston PB, et al. Utility of routine post-therapy surveillance imaging in diffuse large B-cell lymphoma. J Clin Oncol. 2014;32(31):3506–12.
Huntington SF, Svoboda J, Doshi JA. Cost-effectiveness analysis of routine surveillance imaging of patients with diffuse large B-cell lymphoma in first remission. J Clin Oncol. 2015;33(13):1467–74.
Lakhotia R, Melani C, Dunleavy K, Pittaluga S, Saba N, Lindenberg L, Mena E, Bergvall E, Lucas AN, Jacob A, et al. Circulating tumor DNA predicts therapeutic outcome in mantle cell lymphoma. Blood Adv. 2022;6(8):2667–80.
Agarwal R, Chan YC, Tam CS, Hunter T, Vassiliadis D, Teh CE, Thijssen R, Yeh P, Wong SQ, Ftouni S, et al. Dynamic molecular monitoring reveals that SWI-SNF mutations mediate resistance to ibrutinib plus venetoclax in mantle cell lymphoma. Nat Med. 2019;25(1):119–29.
Martínez-Laperche C, Sanz-Villanueva L, Díaz Crespo FJ, Muñiz P, Martín Rojas R, Carbonell D, Chicano M, Suárez-González J, Menárguez J, Kwon M, et al. EZH2 mutations at diagnosis in follicular lymphoma: a promising biomarker to guide frontline treatment. BMC Cancer. 2022;22(1):982.
Nagy Á, Bátai B, Kiss L, Gróf S, Király PA, Jóna Á, Demeter J, Sánta H, Bátai Á, Pettendi P, et al. Parallel testing of liquid biopsy (ctDNA) and tissue biopsy samples reveals a higher frequency of EZH2 mutations in follicular lymphoma. J Intern Med. 2023;294(3):295–313.
Bödör C, Grossmann V, Popov N, Okosun J, O’Riain C, Tan K, Marzec J, Araf S, Wang J, Lee AM, et al. EZH2 mutations are frequent and represent an early event in follicular lymphoma. Blood. 2013;122(18):3165–8.
Okosun J, Bödör C, Wang J, Araf S, Yang CY, Pan C, Boller S, Cittaro D, Bozek M, Iqbal S, et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet. 2014;46(2):176–81.
Hatipoğlu T, Esmeray Sönmez E, Hu X, Yuan H, Danyeli AE, Şeyhanlı A, Önal-Süzek T, Zhang W, Akman B, Olgun A, et al. Plasma concentrations and cancer-associated mutations in cell-free circulating DNA of treatment-naive Follicular lymphoma for improved non-invasive diagnosis and prognosis. Front Oncol. 2022;12:870487.
Zhao M, Li Q, Yang J, Zhang M, Liu X, Zhang H, Huang Y, Li J, Bao J, Wang J, et al. Application of circulating tumour DNA in terms of prognosis prediction in Chinese follicular lymphoma patients. Front Genet. 2023;14:1066808.
Yoon SE, Shin SH, Nam DK, Cho J, Kim WS, Kim SJ. Feasibility of circulating tumor DNA analysis in patients with Follicular Lymphoma. Cancer Res Treat. 2024.
Delfau-Larue MH, van der Gucht A, Dupuis J, Jais JP, Nel I, Beldi-Ferchiou A, Hamdane S, Benmaad I, Laboure G, Verret B, et al. Total metabolic tumor volume, circulating tumor cells, cell-free DNA: distinct prognostic value in follicular lymphoma. Blood Adv. 2018;2(7):807–16.
Sarkozy C, Huet S, Carlton VE, Fabiani B, Delmer A, Jardin F, Delfau-Larue MH, Hacini M, Ribrag V, Guidez S, et al. The prognostic value of clonal heterogeneity and quantitative assessment of plasma circulating clonal IG-VDJ sequences at diagnosis in patients with follicular lymphoma. Oncotarget. 2017;8(5):8765–74.
Fernández-Miranda I, Pedrosa L, Llanos M, Franco FF, Gómez S, Martín-Acosta P, García-Arroyo FR, Gumá J, Horcajo B, Ballesteros AK, et al. Monitoring of circulating tumor DNA predicts response to treatment and early progression in follicular lymphoma: results of a prospective pilot study. Clin Cancer Research: Official J Am Association Cancer Res. 2023;29(1):209–20.
Jiménez-Ubieto A, Martín-Muñoz A, Poza M, Dorado S, García-Ortiz A, Revilla E, Sarandeses P, Ruiz-Heredia Y, Baumann T, Rodríguez A, et al. Personalized monitoring of circulating tumor DNA with a specific signature of trackable mutations after chimeric antigen receptor T-cell therapy in follicular lymphoma patients. Front Immunol. 2023;14:1188818.
Montoto S, Fitzgibbon J. Transformation of indolent B-cell lymphomas. J Clin Oncol. 2011;29(14):1827–34.
Schroers-Martin JG, Soo J, Brisou G, Scherer F, Kurtz DM, Sworder BJ, Khodadoust MS, Jin MC, Bru A, Liu CL, et al. Tracing founder mutations in circulating and tissue-resident Follicular lymphoma precursors. Cancer Discov. 2023;13(6):1310–23.
Tatarczuch M, Waltham M, Shortt J, Polekhina G, Hawkes EA, Ho S-J, Trotman J, Brasacchio D, Co M, Li J, et al. Molecular associations of response to the new-generation BTK inhibitor zanubrutinib in marginal zone lymphoma. Blood Adv. 2023;7(14):3531–9.
Yoon SE, Kim YJ, Shim JH, Park D, Cho J, Ko YH, Park WY, Mun YC, Lee KE, Cho D, et al. Plasma circulating tumor DNA in patients with primary central nervous system lymphoma. Cancer Res Treat. 2022;54(2):597–612.
He J, Wu J, Jiao Y, Rodriguez FJ, Blakeley JO, Kinzler KW, Papadopoulos N, Vogelstein B, Holdhoff M. Limited detection of IgH gene rearrangements in plasma of patients with primary central nervous system lymphoma. J Neurooncol. 2013;114(3):275–9.
Bobillo S, Crespo M, Escudero L, Mayor R, Raheja P, Carpio C, Rubio-Perez C, Tazón-Vega B, Palacio C, Carabia J, et al. Cell free circulating tumor DNA in cerebrospinal fluid detects and monitors central nervous system involvement of B-cell lymphomas. Haematologica. 2021;106(2):513–21.
Hickmann AK, Frick M, Hadaschik D, Battke F, Bittl M, Ganslandt O, Biskup S, Döcker D. Molecular tumor analysis and liquid biopsy: a feasibility investigation analyzing circulating tumor DNA in patients with central nervous system lymphomas. BMC Cancer. 2019;19(1):192.
Mutter JA, Alig SK, Esfahani MS, Lauer EM, Mitschke J, Kurtz DM, Kühn J, Bleul S, Olsen M, Liu CL, et al. Circulating tumor DNA profiling for detection, risk stratification, and classification of brain lymphomas. J Clin Oncol. 2023;41(9):1684–94.
Wang X, Su W, Gao Y, Feng Y, Wang X, Chen X, Hu Y, Ma Y, Ou Q, Liang D, et al. A pilot study of the use of dynamic analysis of cell-free DNA from aqueous humor and vitreous fluid for the diagnosis and treatment monitoring of vitreoretinal lymphomas. Haematologica. 2022;107(9):2154–62.
Cani AK, Hovelson DH, Demirci H, Johnson MW, Tomlins SA, Rao RC. Next generation sequencing of vitreoretinal lymphomas from small-volume intraocular liquid biopsies: new routes to targeted therapies. Oncotarget. 2017;8(5):7989–98.
Hiemcke-Jiwa LS, Ten Dam-van Loon NH, Leguit RJ, Nierkens S, Ossewaarde-van Norel J, de Boer JH, Roholl FF, de Weger RA, Huibers MMH, de Groot-Mijnes JDF, et al. Potential diagnosis of vitreoretinal lymphoma by detection of MYD88 mutation in aqueous humor with ultrasensitive droplet digital polymerase chain reaction. JAMA Ophthalmol. 2018;136(10):1098–104.
Downs BM, Ding W, Cope LM, Umbricht CB, Li W, He H, Ke X, Holdhoff M, Bettegowda C, Tao W, et al. Methylated markers accurately distinguish primary central nervous system lymphomas (PCNSL) from other CNS tumors. Clin Epigenetics. 2021;13(1):104.
Heger JM, Mattlener J, Schneider J, Gödel P, Sieg N, Ullrich F, Lewis R, Bucaciuc-Mracica T, Schwarz RF, Rueß D, et al. Entirely noninvasive outcome prediction in central nervous system lymphomas using circulating tumor DNA. Blood. 2024;143(6):522–34.
Hattori K, Sakata-Yanagimoto M, Suehara Y, Yokoyama Y, Kato T, Kurita N, Nishikii H, Obara N, Takano S, Ishikawa E, et al. Clinical significance of disease-specific MYD88 mutations in circulating DNA in primary central nervous system lymphoma. Cancer Sci. 2018;109(1):225–30.
Iriyama C, Murate K, Iba S, Okamoto A, Yamamoto H, Kanbara A, Sato A, Iwata E, Yamada R, Okamoto M, et al. Detection of circulating tumor DNA in cerebrospinal fluid prior to diagnosis of spinal cord lymphoma by flow cytometric and cytologic analyses. Ann Hematol. 2022;101(5):1157–9.
Zorofchian S, Lu G, Zhu JJ, Duose DY, Windham J, Esquenazi Y, Ballester LY. Detection of the MYD88 p.L265P mutation in the CSF of a patient with secondary Central Nervous System Lymphoma. Front Oncol. 2018;8:382.
Hiemcke-Jiwa LS, Leguit RJ, Snijders TJ, Bromberg JEC, Nierkens S, Jiwa NM, Minnema MC, Huibers MMH. MYD88 p.(L265P) detection on cell-free DNA in liquid biopsies of patients with primary central nervous system lymphoma. Br J Haematol. 2019;185(5):974–7.
Liang JH, Wu YF, Shen HR, Li Y, Liang JH, Gao R, Hua W, Shang CY, Du KX, Xing TY, et al. Clinical implications of CSF-ctDNA positivity in newly diagnosed diffuse large B cell lymphoma. Leukemia. 2024;38(7):1541–52.
Olszewski AJ, Chorzalska AD, Petersen M, Ollila TA, Zayac A, Kurt H, Treaba DO, Reagan JL, Hsu A, Egan PC, et al. Detection of clonotypic DNA in the cerebrospinal fluid as a marker of central nervous system invasion in lymphoma. Blood Adv. 2021;5(24):5525–35.
Hayashida M, Maekawa F, Chagi Y, Iioka F, Kobashi Y, Watanabe M, Ohno H. Combination of multicolor flow cytometry for circulating lymphoma cells and tests for the RHOA(G17V) and IDH2(R172) hot-spot mutations in plasma cell-free DNA as liquid biopsy for the diagnosis of angioimmunoblastic T-cell lymphoma. Leuk Lymphoma. 2020;61(10):2389–98.
Sakata-Yanagimoto M, Nakamoto-Matsubara R, Komori D, Nguyen TB, Hattori K, Nanmoku T, Kato T, Kurita N, Yokoyama Y, Obara N, et al. Detection of the circulating tumor DNAs in angioimmunoblastic T- cell lymphoma. Ann Hematol. 2017;96(9):1471–5.
Qi F, Cao Z, Chen B, Chai Y, Lin J, Ye J, Wei Y, Liu H, Han-Zhang H, Mao X, et al. Liquid biopsy in extranodal NK/T-cell lymphoma: a prospective analysis of cell-free DNA genotyping and monitoring. Blood Adv. 2021;5(11):2505–14.
Ottolini B, Nawaz N, Trethewey CS, Mamand S, Allchin RL, Dillon R, Fields PA, Ahearne MJ, Wagner SD. Multiple mutations at exon 2 of RHOA detected in plasma from patients with peripheral T-cell lymphoma. Blood Adv. 2020;4(11):2392–403.
Kim JJ, Kim HY, Choi Z, Hwang SY, Jeong H, Choi JR, Yoon SE, Kim WS, Kim SH, Kim HJ, et al. In-depth circulating tumor DNA sequencing for prognostication and monitoring in natural killer/T-cell lymphomas. Front Oncol. 2023;13:1109715.
Li Q, Zhang W, Li J, Xiong J, Liu J, Chen T, Wen Q, Zeng Y, Gao L, Gao L, et al. Plasma circulating tumor DNA assessment reveals KMT2D as a potential poor prognostic factor in extranodal NK/T-cell lymphoma. Biomark Res. 2020;8:27.
Kim SJ, Kim YJ, Yoon SE, Ryu KJ, Park B, Park D, Cho D, Kim HY, Cho J, Ko YH, et al. Circulating tumor DNA-based genotyping and monitoring for predicting disease relapses of patients with peripheral T-cell lymphomas. Cancer Res Treat. 2023;55(1):291–303.
Miljkovic MD, Melani C, Pittaluga S, Lakhotia R, Lucas N, Jacob A, Yusko E, Jaffe ES, Wilson WH, Roschewski M. Next-generation sequencing-based monitoring of circulating tumor DNA reveals clonotypic heterogeneity in untreated PTCL. Blood Adv. 2021;5(20):4198–210.
Jin-Hua L, Wei H, Xin-Yi Z, Haorui S, Jia-Zhu W, Jun-Heng L, Liu-Qing Z, Hua Y, Yue L, Li W et al. Clinical implications of ctdna in residual disease assessment, prognosis prediction, disease monitoring for newly peripheral t-cell lymphoma patients. EHA. 2024.
Gao Y, He H, Li X, Zhang L, Xu W, Feng R, Li W, Xiao Y, Liu X, Chen Y, et al. Sintilimab (anti-PD-1 antibody) plus chidamide (histone deacetylase inhibitor) in relapsed or refractory extranodal natural killer T-cell lymphoma (SCENT): a phase Ib/II study. Signal Transduct Target Ther. 2024;9(1):121.
Tian X-P, Zhang Y-C, Lin N-J, Wang L, Li Z-H, Guo H-G, Ma S-Y, An M-J, Yang J, Hong Y-H, et al. Diagnostic performance and prognostic value of circulating tumor DNA methylation marker in extranodal natural killer/T cell lymphoma. Cell Rep Med. 2023;4(2):100859.
Camus V, Stamatoullas A, Mareschal S, Viailly PJ, Sarafan-Vasseur N, Bohers E, Dubois S, Picquenot JM, Ruminy P, Maingonnat C, et al. Detection and prognostic value of recurrent exportin 1 mutations in tumor and cell-free circulating DNA of patients with classical Hodgkin lymphoma. Haematologica. 2016;101(9):1094–101.
Jardin F, Pujals A, Pelletier L, Bohers E, Camus V, Mareschal S, Dubois S, Sola B, Ochmann M, Lemonnier F, et al. Recurrent mutations of the exportin 1 gene (XPO1) and their impact on selective inhibitor of nuclear export compounds sensitivity in primary mediastinal B-cell lymphoma. Am J Hematol. 2016;91(9):923–30.
Van Slambrouck C, Huh J, Suh C, Song JY, Menon MP, Sohani AR, Duffield AS, Goldberg RC, Dama P, Kiyotani K, et al. Diagnostic utility of STAT6(YE361) expression in classical Hodgkin lymphoma and related entities. Mod Pathol. 2020;33(5):834–45.
Buedts L, Wlodarska I, Finalet-Ferreiro J, Gheysens O, Dehaspe L, Tousseyn T, Fornecker LM, Lazarovici J, Casasnovas RO, Gac AC, et al. The landscape of copy number variations in classical Hodgkin lymphoma: a joint KU Leuven and LYSA study on cell-free DNA. Blood Adv. 2021;5(7):1991–2002.
García-Foncillas J, Alba E, Aranda E, Díaz-Rubio E, López-López R, Tabernero J, Vivancos A. Incorporating BEAMing technology as a liquid biopsy into clinical practice for the management of colorectal cancer patients: an expert taskforce review. Ann Oncol. 2017;28(12):2943–9.
Correia RP, Puga RD, Muto NH, Lee MLM, Torres DC, Hassan R, Bacal NS, Hamerschlak N, Campregher PV. High-throughput sequencing of immunoglobulin heavy chain for minimal residual disease detection in B-lymphoblastic leukemia. Int J Lab Hematol. 2021;43(4):724–31.
Herrera AF, Kim HT, Kong KA, Faham M, Sun H, Sohani AR, Alyea EP, Carlton VE, Chen YB, Cutler CS, et al. Next-generation sequencing-based detection of circulating tumour DNA after allogeneic stem cell transplantation for lymphoma. Br J Haematol. 2016;175(5):841–50.
Acknowledgements
We sincerely thank the members of Dr. Gu’s and Dr. Zhang’s laboratories for their insightful discussions and constructive input during the preparation of this manuscript. We also extend our gratitude to Lingyun Zhou of Hangzhou ShengTing Medical Co. for assistance with figure editing. Additionally, we deeply appreciate the support of colleagues from Dr. Wang’s laboratory at the First Hospital of China Medical University, whose contributions were instrumental to the successful progression of this work.
Funding
HG is supported by the Noncommunicable Chronic Diseases–National Science and Technology Major Project (No. 2024ZD0520100) and the CASHIPS Seed Grant. FZ received the Hundred Talents Program Award from the Chinese Academy of Sciences. XW’s research was funded by the National Natural Science Foundation of China (grant number 81900153) and the Liaoning Province Natural Science Foundation (grant number 2022-YGJC-62).
Author information
Authors and Affiliations
Contributions
LF, XRZ, and XYZ conducted the literature search and drafted the manuscript. XL and FZ edited the figures and proofread the manuscript. HG and XW critically reviewed and edited the manuscript. All authors have read and approved the final version of the manuscript for publication.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Fu, L., Zhou, X., Zhang, X. et al. Circulating tumor DNA in lymphoma: technologies and applications. J Hematol Oncol 18, 29 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-025-01673-7
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-025-01673-7