Abstract
Shallow-depth sequencing of cell-free DNA, an inexpensive and standardized approach to obtain molecular information on tumors non-invasively, has been insufficiently explored for the diagnosis of lymphoma and disease follow-up. This study collected 318 samples, including longitudinal liquid and paired solid biopsies, from a prospectively- recruited cohort of 38 Hodgkin lymphoma (HL) and 85 aggressive B-cell non-HL patients, represented by 81 diffuse large B-cell lymphoma (DLBCL) cases. Following sequencing, copy number alterations and viral read fractions were derived and analyzed. At diagnosis, liquid biopsies showed detectable copy number alterations in 84.2% of HL patients (88.6% for classical HL) and 74.1% of DLBCL patients. Of the DLBCL patients, copy number profiles between liquid-solid pairs were highly concordant (r=0.815±0.043); and, compared to tissue, HL liquid biopsies had abnormalities with higher amplitudes (P=0.010). This implies that tumor DNA is more abundant in plasma. Additionally, 39.5% of HL and 13.6% of DLBCL cases had a significantly elevated number of plasma Epstein-Barr virus DNA fragments, achieving a sensitivity of 100% compared to the current standard. A longitudinal analysis determined that, when detectable, copy number patterns were similar across (re)staging moments in refractory or relapsed patients. Further, the overall profile anomaly correlated highly with the total metabolic tumor volume (P<0.001). To conclude, as a proof of principle, we demonstrate that liquid biopsy-derived copy numbers can aid diagnosis: e.g., by differentiating HL from DLBCL, random forest modeling is represented by an area under the receiver operating characteristic curve of 0.967. This application is potentially useful when tissue is difficult to obtain or when biopsies are small and inconclusive.
Introduction
B-cell-derived lymphomas can develop at different stages along the B-lymphocyte differentiation pathways. This generates a range of tumor entities1 including Hodgkin lymphoma (HL) as well as diverse non-HL (nHL) varieties, with diffuse large B-cell lymphoma (DLBCL) the most common nHL.
Both HL and DLBCL typically display distinct morphological, immunohistochemical and (epi)genetic features.2–5 Their high cell turnover makes them excellent candidates for ‘liquid biopsy’-based research. Necrotic and apoptotic tumor cells have been shown to shed DNA6 into the peripheral circulation and this is informative with regard to diagnosis, prognosis and therapy.7 In theory, this would enable real-time (serial) sampling of the entire (often heterogeneous)8 genomic lymphoma architecture in a convenient and non-invasive manner through traditional blood sampling.
Recent studies9–11 have focused on plasma cell-free DNA (cfDNA) analysis using ultra-deep targeted sequencing12 for single nucleotide variant (SNV) and translocation detection. Overall, these studies have provided two main insights: firstly, that variant allele frequencies often correlate with disease status; and, secondly, that surveilling circulating tumor DNA (ctDNA) might outperform 18F-fluoro- 2-deoxyglucose positron emission tomography/computed tomography (PET/CT) scans in terms of sensitivity. This latter finding holds a great deal of potential for relapse risk assessment by quantifying minimal residual disease.
Despite their potential, ultra-deep sequencing techniques are still expensive and require targeted panels. In contrast, shallow (coverage ~0.25x) whole-genome sequencing (sWGS) for primarily copy number detection is cheaper and fully operative in hospitals that offer noninvasive prenatal testing.13 One of the hallmarks of sWGS is its high specificity in identifying malignant cells: in comparison to SNV, large (i.e., >5 Mb) somatic copy number alterations are rarely detected in unaffected subjects14 whereas SNV accumulate over time. Worth mentioning is that this also occurs in hematopoietic stem cells, a phenomenon referred to as clonal hematopoiesis,15 potentially resulting in incorrect driver identifications.
sWGS of cfDNA may therefore have clinical potential, especially for patients with lesions that are difficult to biopsy (e.g. those in brain; deep lymph nodes in the thoracic cavity or abdomen); or when dealing with ambiguous PET/CT scans, as both imaging and clinical symptoms of lymphoma are often non-specific. Nevertheless, this approach has only been superficially investigated.16 We, therefore, evaluated whether sWGS could serve as a molecular test in addition to established diagnostic methods, using a diverse set of 123 prospectively recruited lymphoma patients, comprising baseline and longitudinal blood samples, supplemented with paired formalin-fixed paraffin-embedded (FFPE) biopsies.
Methods
Ethics statement
This study was approved by the institutional ethics committee at Ghent University Hospital (EC/2016/0307). Written informed consent was obtained from all patients.
Study population
Between January 2016 and November 2019, 123 lymphoma patients were recruited at Ghent University Hospital and AZ Delta Roeselare. The subtypes studied include 38 HL (3 nodular lymphocyte predominant HL; 23 nodular sclerosis classical HL [cHL]; 5 mixed cellularity cHL; 5 lymphocyte-rich cHL; 1 lymphocyte- depleted cHL; and 1 not otherwise specified [NOS]); 81 DLBCL (61 NOS; 4 Epstein-Barr virus [EBV]-positive; 10 primary mediastinal large B-cell lymphomas [PMBCL]; 2 high-grade Bcell lymphomas; 2 T-cell/histiocyte-rich large B-cell lymphomas; 1 intravascular large B-cell lymphoma; and 1 plasmablastic lymphoma) and four grey-zone lymphomas (GZL) (Online Supplementary File S2: Tables S1 and S2). Patients were included at the start, ‘baseline’, of a new line of therapy (i.e., baseline represents initial diagnosis for 115 cases; after ineffective treatment in 6 other cases; and at relapse for the 2 remaining cases). Although patient recruitment was random, the final cohort was enriched for HL patients.
Sample series
Liquid biopsies were collected at baseline for all patients (n=123) and a random selection of paired FFPE tissue was made (n=33); this was obtained from 15 HL and 18 DLBCL patients. Longitudinal liquid biopsies were taken following milestones in treatment with 93 sequenced for 31 patients. These were intentionally enriched for refractory or relapsed disease: interim evaluations after two and four cycles of ABVD in HL; interim evaluations after four cycles of R-CHOP in nHL; at restaging after ineffective treatment or relapse; following successful treatment; and every 6 months for patients in maintained complete remission (CR). For HL (n=9), six patients had obtained CR within a year of inclusion whereas three were refractory. For nHL (n=22), 11 patients reached CR within a year; six were refractory; and five relapsed.
Negative liquid biopsies were included from non-invasive prenatal assays (n=60), supplemented with benign FFPE tissue (n=9) as a control for the solid biopsies. In total, 318 samples were thus analyzed.
Clinical and pathological characteristics
Patient demographics and clinical variables were available through routine clinical practice. The total metabolic tumor volume (MTV) was measured by PET/CT using AW workstation semi-automated segmentation software (GE medical systems, Waukesha, WI, USA), with a standard uptake value cutoff of 2.5.
Pathology diagnosis included standard histological examination of FFPE biopsy material in combination with immunohistochemistry (IHC). Double/triple hit (MYC, BCL2 and/or BCL6) and double expressor (MYC and BCL2) samples were detected using fluorescence in situ hybridization and IHC, respectively. The germinal center B-cell (GCB) cell of origin (COO) status was assigned through the Hans algorithm.17
When using chromogenic in situ hybridization (CISH), the presence of EBV-encoded RNA (EBER) was evaluated via INFORM EBER probes (Ventana Medical Systems, Tucson, AZ, USA). Selected liquid biopsies were additionally tested using the quantitative EBV-polymerase chain reaction (PCR) method, as established by Bordon et al.18
Sequencing and bioinformatic analysis
Each laboratory step and subsequent computational analysis can be found in Online Supplementary File S1: Supplemental methods. These include: copy number profiling;19 defining the tumor burden using the copy number profile abnormality (CPA) score20 and estimated tumor fraction;21 methods used to derive viral read fractions of HIV-1, HIV-2, EBV, JC polyomavirus, human Tlymphotropic virus 1, human herpesvirus 8 and hepatitis C virus; random forest modeling22 of copy number profiles to predict tumor subtype; detection of copy number driver peaks;23 and general statistical testing.
Results
Shallow-depth sequencing of cell-free DNA enables accurate copy number profiling in lymphoma
For healthy individuals, copy number profiles are assumed to be approximately ‘flat’, implying that large alterations are absent. In a previous study,20 in order to statistically distinguish cancerous from control profiles, we developed a score that increases with rising deviance from the naturally-occurring healthy background variance: the CPA score. Based on this variable, 95/123 baseline liquid biopsies were detected as cancerous (Figure 1A; Online Supplementary File S2: Table S3). As expected, the score reached significantly higher (P<0.001; Welch test) values in the lymphoma group compared to the control group. There was no significant difference (P=0.250; Fisher exact test) in the fraction of abnormal observations between HL (32/38; 84.2%) and DLBCL (60/81; 74.1%), nor when specifically considering cHL (31/35; 88.6%; P=0.091). At Ann Arbor stage I, only one of six liquid biopsies showed deviations whereas stage II to IV presented comparable detection rates (Online Supplementary File S1: Figure S1).
In order to confirm whether the copy number profiles derived from blood samples represent those from tissue biopsies, we executed a concordance analysis using 33 liquid-solid pairs (Online Supplementary File 1: Liquid-solid pairs). One representative DLBCL case, patient 59, demonstrated very similar deviations in both samples (Figure 1B), and the Pearson correlation between this patient’s smoothened profiles amounted to 0.916. We can note, from left to right, four clusters distinguished in a corresponding scatter plot (Figure 1C): deletions, copy neutral bins, duplications and amplifications. It should be noted that the amplitudes of alterations, a concept defined as the absolute value of a segment’s log2 ratio,19 are more extreme in liquid than in tissue biopsies (Figure 1B), caused by a greater ctDNA fraction in patient 59’s liquid biopsy. This is also indicated by a steeper identity line than least squares fit in the scatter plot (Figure 1C). Overall, when the CPA score in the liquid biopsy was abnormal, DLBCL pairs showed high concordance (r=0.815±0.043; 95% confidence interval). However, this was not the case for HL (r=0.260±0.055; 95% confidence interval) patients (Figure 1D).
Next, the amplitudes were compared between paired liquid and solid biopsies (Figure 1E). These numbers were averaged per sample and additionally normalized by subtracting the mean across the control samples (0.005 for liquid; 0.026 for solid), which results in a fairer comparison, as FFPE material tends to produce profiles with higher levels of noise,24 naturally increasing the overall deviance. This variable was found to be higher in liquid than in solid biopsies for HL (P=0.010; paired ttest), but not for DLBCL (P=0.316; paired t-test). Worthy of note is that, within HL, the mean of the solid biopsies barely varied from zero, meaning that most of these profiles are hard to differentiate from normal controls, and, therefore, the low liquid-solid correlation observed in HL patients (Figure 1D), seems to be caused by a scarcity of tumor cells in the FFPE samples rather than a low ctDNA fraction. From a biological perspective, this is reasonable: neoplastic Hodgkin and Reed-Sternberg cells are always embedded in an inflammatory background of non-affected cells,25 causing resemblance to healthy tissue.
Shallow-depth sequencing of cell-free DNA enables sensitive Epstein-Barr virus detection in lymphoma
Sequencing reads were mapped to seven different lymphoma- associated viruses. Of the lymphoma liquid biopsies, 39/216 had abnormally-elevated EBV read fractions compared to the negative controls (Online Supplementary File S2: Table S3); whereas one patient tested positive for JC polyomavirus (13 reads) at surveillance (patient 79). In accordance with clinical records, no other viruses were detected. Within control samples, no or exclusively low amounts of EBV reads were noted. The latter was the case for 15/60 liquid (read fraction range 0.032-0.059 parts per million [ppm]) and 6/9 solid controls (0.029-0.08 ppm).
The EBV read fraction was evaluated for all baseline liquid biopsies, with 28/123 patients testing positive (Figure 2A). For GZL, 2/4 patients were positive, and, as expected, more HL samples were EBV-positive (15/38; 39.5%) than DLBCL samples (11/81; 13.6%) (P=0.004; Fisher exact test). All but one EBV-positive sample had an abnormal CPA score in addition (Figure 2B), which significantly associates the latter categorical variables (P=0.004; Fisher exact test).
The sensitivity and specificity of EBV detection through sWGS of cfDNA was established by a comparison with routine CISH for EBER detection results, performed on FFPE tissue. All 18 samples that tested positive for EBER, as well as 8/46 EBER-negative samples, had abnormally-increased EBV levels in plasma, achieving a sensitivity and specificity of 100% and 82.6%, respectively (Figure 2B; Online Supplementary File S2: Tables S3 and S4). The liquid biopsies with the highest viral read fractions of the EBER-negative cases did, however, accommodate increased levels of EBV fragments in blood, as confirmed by a subsequent PCR analysis (Figure 2B). This indicates that routine CISH might have inadequate sensitivity or that viral fragments may have been absent in the tumor cells studied. When considering the latter five as true positives, specificity increases to 93.5%.
As for the copy number alteration amplitudes, liquid biopsies had higher (P=0.039; paired Wilcoxon signed-rank test) EBV read fractions than solid biopsies (Figure 2C). To conclude, three receiver operating characteristic (ROC) analyses (Figure 2D) were included to further demonstrate the performance of this EBV detection approach. Ranked plasma EBV viral read fractions and three different gold standards were used: CISH overridden by PCR (area under the curve [AUC]=0.994); CISH only (AUC=0.948); and solid biopsy reads (AUC=0.868).
Copy numbers derived from liquid biopsies can aid histological classification
When visualizing fractions of aberrant samples across genomic loci per histological subtype, both general (e.g., gains at 2p, holding REL) and subtype-specific fingerprints can be distinguished (Figure 3). Unsupervised clustering26 applied to these summarizing profiles results in the anticipated histological hierarchy. Indeed, nHL and HL are separated as two definite entities, with the exception of GZL and PMBCL, which cluster alongside the HL group. Notwithstanding their non-Hodgkin histology, both have been described as being molecularly related to HL,27,28 hence the structure of the dendrogram. GZL in particular demonstrates overlapping features with cHL and PMBCL.28
To mimic how this translates as a potential clinical application for samples that are difficult to classify morphologically, or in cases in which obtaining tumor tissue is complicated, random forest predictive modeling, in combination with cross-validation, was applied to four differential diagnoses (Figure 4A): HL versus DLBCL; cHL versus PMBCL; DLBCL, NOS versus PMBCL; and within DLBCL, NOS, GCB versus non-GCB. For these analyses, obviously only baseline liquid biopsies were employed. Two ROC AUC can be discussed here: one for all samples and one disregarding flat copy number profiles. The latter statistic is considered in the next paragraph as it represents the performance of a rational application: profiles without aberrations will never be subjected to machine learning algorithms since predictions would be random.
The differential diagnosis of HL and DLBCL, using computationally- processed copy number profiles, was found to be accurate (AUC of 0.967). Out of bag principles (i.e., cross-validation for bootstrapping methods, to overcome train/test bias) were applied to a set of 119 samples, meaning that, at each iteration, sufficient data were available for model training; and, biologically, HL and DLBCL express divergent profiles (Figure 3). PMBCL has features between those of cHL and DLBCL.27 Nonetheless, copy number profiles can aid differential diagnoses, indicated by AUC values of 0.931 and 0.981, respectively. Finally, the GCB versus non-GCB COO status does not seem to trigger specific copy number alterations, as shown by the resulting AUC of 0.450. We could not test whether the activated B-cell versus GCB distinction by gene expression profiling29 would perform better. This should be explored in future work.
During training, learning algorithms assign higher weights to features that improve classification. For the HL versus DLBCL problem, the top three discriminative features - or loci, in our case - are located at 2p, 9p and 11q (Figure 4B, bottom). In addition, the GISTIC software aims to define driver genes in pattern peaks across a set of samples. Of note, there were three interesting peaks in these regions that were significantly enriched (Figure 4B, top), matching several anticipated genes: in both HL and DLBCL, 2p16.1 (REL) gains;30,31 9p24.1 (JAK2 and PD-L1) gains in HL;32 9p21.3 (CDKN2A) losses in DLBCL;33 and to conclude, 11q23.3 (CEP164) gains in DLBCL34 (Online Supplementary File S2: Table S5). Although 2p16.1 was found to be amplified, or duplicated, in both HL and DLBCL, the model gives importance to the 2p arm, as these aberrations are more frequently detected in HL compared to DLBCL (Figure 4B, top).35
Finally, locus importance was examined for the two other well-performing models: to a certain extent, 5p, 9q, and chromosomes 14 and 19 allow separation of cHL and PMBCL, while more specific 7p and, once more, 9p24.1 (JAK2 and PD-L1)32 cytobands are decisive in differentiating DLBCL from PMBCL (Figure 4C).
Longitudinal liquid biopsies correlate with overall disease status
Serial blood samples were analyzed to observe copy number alteration dynamics over time and to study longitudinal changes in CPA and EBV read fractions. One example of an intra-patient evolution is demonstrated by patient 32, who had refractory DLBCL (Figure 5A). In this case, alterations disappeared during treatment (MTV on PET/CT decreased from 915 cm3 at baseline to 4 cm3 between both interim moments), and reappeared when progression was diagnosed after 212 days (MTV increased to 79 cm3). The smoothened profiles at initial staging and subsequent restaging at progression were very similar (r= 0.946; Figure 5B), notwithstanding the long time interval, suggesting that a portion of ‘remnant’ tumor had never been completely eliminated and that minimal residual disease had caused recidivism. Despite the significant correlation, evidence of tumor evolution was present, as indicated by several changes along the copy number alteration pattern, e.g.,on chromosome arms 2q and 6p; and on chromosome 14 Figure 5A). These zones are represented by irregularities in the scatter plot trend between the profiles (Figure 5B).
Overall, observations were consistent for refractory and relapsed patients: when detectable, copy number patterns were very similar across different longitudinal staging moments (Figure 5C). One exception was found in patient 73 (rightmost patient in Figure 5C), who appeared to relapse with a divergent copy number profile (Online Supplementary File S1: Longitudinal samples). This case concerned an aggressive EBV-positive plasmablastic lymphoma that is known to present complex karyotypic abnormalities and, presumably, an instable genome.36
The most recent follow-up of all 17 patients in CR showed that none had remaining copy number alterations; in contrast, this was the case in only 5/14 patients in whom CR was thought not to be reached (Figure 5D). In total, five EBV-positive patients, according to sWGS of cfDNA, were detected at diagnosis of the 31 patients with follow-up. Of these, three went into CR and, consequently, no longer had abnormally elevated EBV levels; one refractory patient (patient 6) was noted to have recurrently high EBV levels and one relapsed patient (patient 73) was detected with an unusually high EBV read fraction, following an earlier drop to 0 ppm at first response evaluation (Figure 5E). It should be noted that, for this patient, no tumor was detected under PET/CT at the fourth time point (Online Supplementary File 2: Table S4), however, the steadily increasing EBV fraction anticipated actual relapse at the fifth time point, after which the MTV increased rapidly along with the plasma concentration of viral fragments (Online Supplementary File S1: Longitudinal samples).
A similar analysis, concerning the CPA score, was performed for all patients with HL (Figure 5F) and nHL (Figure 5G) with follow-up. Similar conclusions were made according to the trend lines, which largely summarize the longitudinal analyses: a prompt drop for patients in CR; varying values for refractory patients; and a drop, followed by a newly-triggered rise, for relapsed patients. It is significant how the CPA decreased rapidly when treatment was initiated: interim samples had normal CPA scores unless they represented a refractory patient. Finally, 6/31 patients never transcended the CPA abnormality cutoff, meaning that these, probably, have insufficient ctDNA. In two cases (patients 66 and 71), however, the MTV reached extreme levels (>1000 cm3) while the CPA score stayed low (Online Supplementary File S1: Longitudinal samples). For these patients, it is more likely that copy number alterations really were absent.
Of the above cases, additional clinical information from four interesting patients further illustrates the potential of shallow-depth sequencing for disease monitoring - e.g., when dealing with ambiguous PET/CT images or tumor and viral entities beyond initial suspicion. These are discussed as short case-reports in Online Supplementary File S1: Case reports.
Copy number anomalies detected in liquid biopsies correlate with clinical parameters
Five lymphoma-related clinical variables were included in an extensive concordance analysis of the baseline and longitudinal liquid biopsies: Ann Arbor stage; International Prognostic Index/Score; lactate dehydrogenase concentration; b2 microglobulin concentration; and MTV (Online Supplementary File S1: Figure S2). The estimated tumor fraction, according to copy number profiles, positively and significantly correlates with all of these clinical variables, rendering the former estimate relevant. The CPA score is intrinsically correlated to the tumor fraction, so similar associations as for the tumor fraction arise. Since all these variables are naturally cross-related, the derived relations are obviously not claimed to be independently significant.
Discussion
To date, molecular profiling of lymphoma has largely been focused on the characterization of actionable targets, such as point mutations and translocations. This profiling requires panel sequencing, currently using tissue biopsies as the DNA source and targeting a substantial number of genes. However, until now, the availability of this precise information has contributed little to actual progress in patient care, despite its tempting rationale.37 In addition, applying these concepts in a minimally-invasive manner requires ultra-deep sequencing (~2000x),38 of which the implementation in routine practice remains challenging.
In this study, we evaluated sWGS of cfDNA for the diagnosis, differential diagnosis, and disease monitoring of HL and DLBCL. In contrast to targeted sequencing, this approach provides genomic insight by copy number profiling in an accessible and standardized manner. Indeed, noninvasive prenatal testing, which involves the same laboratory and computational steps, has evolved into an application performed daily in molecular diagnostic laboratories. The short turnaround time (approximately, 4 days) and lowprice tag (around $200, including processing costs) renders sWGS a fortiori feasible.
Copy number profiling is not a surrogate for targeted sequencing, nor does it enable the characterization of phenotypic traits, such as CD20 expression. However, to a limited extent, it does enable the prediction of targetable genes, such as JAK2, PD-L1 and REL. Further, regarding tumor fraction and specificity, it has the advantage of easier and more stable interpretation: once a copy number alteration is visible, the presence of sufficient ctDNA is most likely ascertained given that large somatic copy number alterations are rarely detected in unaffected subjects.14
For DLBCL, the sensitivity of copy number profiling is a shortcoming: only 74.1% of baseline liquid biopsies were found to have detectable aberrations, which is lower than that reported from targeted ultra-deep duplex sequencing studies (e.g., 98% in Kurtz et al.39). On the other hand, the sensitivity was surprisingly high for HL (84.2% for HL overall and 88.6% for cHL in this study; 88.9% in a smaller proof-of-concept study16) and compares to that of SNVbased studies (e.g., 81.2% in Spina et al.11). It ought to be noted that the lower limit of detection for CNA is often considered to be 3% tumor fraction.21 It is worth mentioning that this limit could be lowered as paired-end sequencing at greater depths (1x-1.5x) is expected to become available for routine diagnosis at a similar cost. This would empower computational tumor-read enrichment by insertsize filtering, as shorter cfDNA fragments are more likely be tumor-derived, thus increasing sensitivity.20,40,41
The signs and symptoms of aggressive lymphoma are often non-specific, having much in common with a broad range of different disorders, including autoimmune, immunodeficiency and systemic diseases, infections and other malignancies. Therefore, currently, accurate lymphoma diagnosis is essentially only possible through invasive tissue biopsy. However, affected sites are sometimes difficult to reach and tissue-based subtyping may be complicated when dealing with inconclusive or small biopsies, or partially-involved lymph nodes. Since several cases of differential diagnosis by computationally-processed copy number profiles have been found to be accurate, copy number profiling of cfDNA might enter clinical practice as a test for patients with: suspected lymphoma with a mediastinal mass (to guide the need for mediastinoscopy); deep-seated pathologic lymph nodes in the abdomen, especially following surgery; a remaining suspicion of lymphoma after a negative biopsy, caused by ambiguous PET/CT images; probable central nervous system lymphoma (to avoid radical surgery); probable intravascular localization;42 and, generally, lymphoma in children (to avoid unnecessary surgery). For these indications, the approach presented here could be easily integrated with other tests in order to decide whether invasive procedures are still necessary.
sWGS of cfDNA was found to be 100% sensitive for the detection of EBV. However, one fact that could preclude its application in lymphoma diagnosis is that all possible causes of elevated EBV levels (e.g., ongoing infectious mononucleosis) are detected simultaneously through blood samples irrespectively of whether these are related to lymphoma. CISH does not have this shortcoming and can be used to assess a neoplastic association. Nonetheless, the combination of an increased EBV level and typical HL or DLBCL alterations strongly indicates an EBV-driven lymphoma. This is especially relevant for the diagnosis of post-transplant or immunodeficiency-associated lymphoma. sWGS could be employed, in part, to detect EBV prior to performing a CISH test (around $140), when required. The latter test should only be performed when EBV is positive.
Future in-house research will focus on the inclusion of other (rarer) lymphoma subtypes, including T-cell lymphomas, to obtain a broader image of the alterations’ specificity compared to subtype. Studies will also concentrate on novel methods to study cfDNA (epi)genetics, including cellfree reduced-representation bisulfite sequencing for methylation detection, which are likely to improve specificity. Since this study was concluded, new liquid biopsies have been analyzed by sWGS at our institutions, mostly in cases of diagnostic ambiguity. While negative results currently never exclude the presence of malignancies, positive results ascertain disease.
Footnotes
- Received August 3, 2020
- Accepted November 25, 2020
Correspondence
Disclosures
No conflicts of interest to disclose.
Contributions
Study supervision: FO and JVD; patient recruitment: CDV, DD, FO, KDG and SB; clinical/pathological data acquisition/generation: CDV, BVDB, KM, DD, FD, SV, AD, FO and JVD; data processing: MVDL; study design: LR, MVDL, CDV, BM, FO and JVD; data analysis: LR; manuscript writing: LR and JVD; all authors read, revised and approved the final manuscript.
Data-sharing statement
Copy number profiles and viral reads counts of all sequenced samples are included in Online Supplementary file: Table S6 and S7, respectively.
Funding
This study was supported by Bijzonder Onderzoeksfonds (BOF), Ghent University, in the form of a doctoral research grant (BOF.STA.2017.0002.01 to LR).
Acknowledgments
The authors would like to thank all collaborators at AZ Delta Roeselare and Ghent University Hospital for the active cohort recruitment, sample collection and provision of individual patient data.
References
- Shaffer AL, Young RM, Staudt LM. Pathogenesis of human B cell lymphomas. Annu Rev Immunol. 2012; 30:565-610. https://doi.org/10.1146/annurev-immunol-020711-075027PubMedPubMed CentralGoogle Scholar
- Re D, Küppers R, Diehl V. Molecular pathogenesis of Hodgkin’s lymphoma. J Clin Oncol. 2005; 23(26):6379-6386. https://doi.org/10.1200/JCO.2005.55.013PubMedGoogle Scholar
- Steidl C, Connors JM, Gascoyne RD. Molecular pathogenesis of Hodgkin’s lymphoma: increasing evidence of the importance of the microenvironment. J Clin Oncol. 2011; 29(14):1812-1826. https://doi.org/10.1200/JCO.2010.32.8401PubMedGoogle Scholar
- Schmitz R, Wright GW, Huang DW. Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med. 2018; 378(15):1396-1407. https://doi.org/10.1056/NEJMoa1801445PubMedPubMed CentralGoogle Scholar
- Steidl C, Gascoyne RD. The molecular pathogenesis of primary mediastinal large B-cell lymphoma. Blood. 2011; 118(10):2659-2669. https://doi.org/10.1182/blood-2011-05-326538PubMedGoogle Scholar
- Schwarzenbach H, Hoon DSB, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011; 11(6):426-437. https://doi.org/10.1038/nrc3066PubMedGoogle Scholar
- Domínguez-Vigil IG, Moreno-Martínez AK, Wang JY, Roehrl MHA, Barrera- Saldaña HA. The dawn of the liquid biopsy in the fight against cancer. Oncotarget. 2017; 9(2):2912-2922. https://doi.org/10.18632/oncotarget.23131PubMedPubMed CentralGoogle Scholar
- Araf S, Wang J, Korfi K. Genomic profiling reveals spatial intra-tumor heterogeneity in follicular lymphoma. Leukemia. 2018; 32(5):1261-1265. https://doi.org/10.1038/s41375-018-0043-yPubMedPubMed CentralGoogle Scholar
- Rossi D, Diop F, Spaccarotella E. Diffuse large B-cell lymphoma genotyping on the liquid biopsy. Blood. 2017; 129(14):1947-1957. https://doi.org/10.1182/blood-2016-05-719641PubMedGoogle Scholar
- Bohers E, Viailly P-J, Becker S. Noninvasive monitoring of diffuse large B-cell lymphoma by cell-free DNA high-throughput targeted sequencing: analysis of a prospective cohort. Blood. Cancer J. 2018; 8(8):74. https://doi.org/10.1038/s41408-018-0111-6PubMedPubMed CentralGoogle Scholar
- Spina V, Bruscaggin A, Cuccaro A. Circulating tumor DNA reveals genetics, clonal evolution, and residual disease in classical Hodgkin lymphoma. Blood. 2018; 131(22):2413-2425. https://doi.org/10.1182/blood-2017-11-812073PubMedGoogle Scholar
- Newman AM, Bratman SV, To J. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014; 20(5):548-554. https://doi.org/10.1038/nm.3519PubMedPubMed CentralGoogle Scholar
- Raman L, Baetens M, De Smet M, Dheedene A, Van Dorpe J, Menten B. PREFACE: In silico pipeline for accurate cell-free fetal DNA fraction prediction. Prenat Diagn. 2019; 39(10):925-933. https://doi.org/10.1002/pd.5508PubMedPubMed CentralGoogle Scholar
- Redon R, Ishikawa S, Fitch KR. Global variation in copy number in the human genome. Nature. 2006; 444(7118):444-454. https://doi.org/10.1038/nature05329PubMedPubMed CentralGoogle Scholar
- Busque L, Patel JP, Figueroa M. Recurrent Somatic TET2 Mutations in Normal Elderly Individuals With Clonal Hematopoiesis. Nat Genet. 2012; 44(11):1179-1181. https://doi.org/10.1038/ng.2413PubMedPubMed CentralGoogle Scholar
- Vandenberghe P, Wlodarska I, Tousseyn T. Non-invasive detection of genomic imbalances in Hodgkin/Reed-Sternberg cells in early and advanced stage Hodgkin’s lymphoma by sequencing of circulating cell-free DNA: a technical proof-of-principle study. Lancet Haematol. 2015; 2(2):e55-65. https://doi.org/10.1016/S2352-3026(14)00039-8PubMedGoogle Scholar
- Hans CP, Weisenburger DD, Greiner TC. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood. 2004; 103(1):275-282. https://doi.org/10.1182/blood-2003-05-1545PubMedGoogle Scholar
- Bordon V, Padalko E, Benoit Y, Dhooge C, Laureys G. Incidence, kinetics, and risk factors of Epstein-Barr virus viremia in pediatric patients after allogeneic stem cell transplantation. Pediatr Transplant. 2012; 16(2):144-150. https://doi.org/10.1111/j.1399-3046.2011.01634.xPubMedGoogle Scholar
- Raman L, Dheedene A, De Smet M, Van Dorpe J, Menten B. WisecondorX: improved copy number detection for routine shallow whole-genome sequencing. Nucleic Acids Res. 2019; 47(4):1605-1614. https://doi.org/10.1093/nar/gky1263PubMedPubMed CentralGoogle Scholar
- Raman L, Van der Linden M, Van der Eecken K. Shallow whole-genome sequencing of plasma cell-free DNA accurately differentiates small from non-small cell lung carcinoma. Genome Med. 2020; 12(1):35. https://doi.org/10.1186/s13073-020-00735-4PubMedPubMed CentralGoogle Scholar
- Adalsteinsson VA, Ha G, Freeman SS. Scalable whole-exome sequencing of cellfree DNA reveals high concordance with metastatic tumors. Nat Commun. 2017; 8(1):1324. https://doi.org/10.1038/s41467-017-00965-yPubMedPubMed CentralGoogle Scholar
- Liaw A, Wiener M. Classification and Regression by random Forest. R News. 2002; 2(3):18-22. Google Scholar
- Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 2011; 12(4):R41. https://doi.org/10.1186/gb-2011-12-4-r41PubMedPubMed CentralGoogle Scholar
- Van der Linden M, Raman L, Vander Trappen A. Detection of copy number alterations by shallow whole-genome sequencing of formalin-fixed, paraffinembedded tumor tissue. Arch Pathol Lab Med. 2020; 144(8):974-981. https://doi.org/10.5858/arpa.2019-0010-OAPubMedGoogle Scholar
- Mathas S, Hartmann S, Küppers R. Hodgkin lymphoma: Pathology and biology. Semin Hematol. 2016; 53(3):139-147. https://doi.org/10.1053/j.seminhematol.2016.05.007PubMedGoogle Scholar
- Ward JH. Hierarchical grouping to optimize an objective function. J Am Stat Assoc. 1963; 58(301):236-244. https://doi.org/10.1080/01621459.1963.10500845Google Scholar
- Rosenwald A, Wright G, Leroy K. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J Exp Med. 2003; 198(6):851-862. https://doi.org/10.1084/jem.20031074PubMedPubMed CentralGoogle Scholar
- Eberle FC, Rodriguez-Canales J, Wei L. Methylation profiling of mediastinal gray zone lymphoma reveals a distinctive signature with elements shared by classical Hodgkin’s lymphoma and primary mediastinal large B-cell lymphoma. Haematologica. 2011; 96(4):558-566. https://doi.org/10.3324/haematol.2010.033167PubMedPubMed CentralGoogle Scholar
- Alizadeh AA, Eisen MB, Davis RE. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000; 403(6769):503-511. https://doi.org/10.1038/35000501PubMedGoogle Scholar
- Joos S, Menz CK, Wrobel G. Classical Hodgkin lymphoma is characterized by recurrent copy number gains of the short arm of chromosome 2. Blood. 2002; 99(4):1381-1387. https://doi.org/10.1182/blood.V99.4.1381PubMedGoogle Scholar
- Houldsworth J, Mathew S, Rao PH. REL proto-oncogene is frequently amplified in extranodal diffuse large cell lymphoma. Blood. 1996; 87(1):25-29. https://doi.org/10.1182/blood.V87.1.25.25PubMedGoogle Scholar
- Green MR, Monti S, Rodig SJ. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010; 116(17):3268-3277. https://doi.org/10.1182/blood-2010-05-282780PubMedPubMed CentralGoogle Scholar
- Lenz G, Wright GW, Emre NCT. Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways. Proc Natl Acad Sci U S A. 2008; 105(36):13520-13525. https://doi.org/10.1073/pnas.0804295105PubMedPubMed CentralGoogle Scholar
- Pasqualucci L, Trifonov V, Fabbri G. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat Genet. 2011; 43(9):830-837. https://doi.org/10.1038/ng.892PubMedPubMed CentralGoogle Scholar
- Kober-Hasslacher M, Schmidt-Supprian M. The unsolved puzzle of c-Rel in B cell lymphoma. Cancers. 2019; 11(7):941. https://doi.org/10.3390/cancers11070941PubMedPubMed CentralGoogle Scholar
- Castillo JJ, Reagan JL. Plasmablastic lymphoma: a systematic review. Scientific World Journal. 2011; 11:687-696. https://doi.org/10.1100/tsw.2011.59PubMedPubMed CentralGoogle Scholar
- Iacoboni G, Zucca E, Ghielmini M, Stathis A. Methodology of clinical trials evaluating the incorporation of new drugs in the firstline treatment of patients with diffuse large B-cell lymphoma (DLBCL): a critical review. Ann Oncol. 2018; 29(5):1120-1129. https://doi.org/10.1093/annonc/mdy113PubMedGoogle Scholar
- Scherer F, Kurtz DM, Newman AM. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016; 8(364):364ra155. https://doi.org/10.1126/scitranslmed.aai8545PubMedPubMed CentralGoogle Scholar
- Kurtz DM, Scherer F, Jin MC. Circulating tumor DNA measurements as early outcome predictors in diffuse large Bcell lymphoma. J Clin Oncol. 2018; 36(28):2845-2853. https://doi.org/10.1200/JCO.2018.78.5246PubMedPubMed CentralGoogle Scholar
- Mouliere F, Mair R, Chandrananda D. Detection of cell-free DNA fragmentation and copy number alterations in cerebrospinal fluid from glioma patients. EMBO Mol Med. 2018; 10(12):e9323. https://doi.org/10.15252/emmm.201809323PubMedPubMed CentralGoogle Scholar
- Cristiano S, Leal A, Phallen J. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature. 2019; 570(7761):385-389. https://doi.org/10.1038/s41586-019-1272-6PubMedPubMed CentralGoogle Scholar
- Deeren D, Van Der Linden M, Dedeurwaerdere F. Circulating cellfree DNA for response evaluation of intravascular lymphoma. Ann Hematol. 2019; 98(8):2021-2023. https://doi.org/10.1007/s00277-019-03677-zPubMedGoogle Scholar
- Rinaldi A, Kwee I, Poretti G. Comparative genome-wide profiling of post-transplant lymphoproliferative disorders and diffuse large B-cell lymphomas. Br J Haematol. 2006; 134(1):27-36. https://doi.org/10.1111/j.1365-2141.2006.06114.xPubMedGoogle Scholar
- Morscio J, Dierickx D, Tousseyn T. Molecular pathogenesis of B-cell posttransplant lymphoproliferative disorder: what do we know so far?. Clin Dev Immunol. 2013; 2013:150835. https://doi.org/10.1155/2013/150835PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.