scholarly journals Phased Variant Enrichment for Enhanced Minimal Residual Disease Detection from Cell-Free DNA

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 552-552 ◽  
Author(s):  
David M. Kurtz ◽  
Joanne Soo ◽  
Stefan Alig ◽  
Lyron Co Ting Keh ◽  
Charles Macaulay ◽  
...  

Background: Circulating tumor DNA (ctDNA) is an emerging biomarker in non-Hodgkin lymphomas (NHLs). Current methods for ctDNA minimal residual disease (MRD) are limited by two factors - low input DNA amounts and high background error rates. VDJ sequencing (i.e., IgHTS) has low background but is limited by low cell-free DNA (cfDNA). Tracking multiple mutations via CAPP-Seq improves sensitivity, but detection is limited by background errors. Clustered mutations have been described in multiple cancers including NHLs and potentially have lower error rates. We explored clustered mutations from whole-genome sequencing (WGS) to identify 'phased variants' (PVs), defined as multiple mutations on a single DNA molecule (Fig 1A). We designed a method to capture PVs for improved ctDNA detection and explored its utility for MRD in DLBCL. Methods: We reanalyzed WGS from 1455 tumors across 11 cancer types. We identified genomic regions recurrently containing PVs and designed an assay for deep cfDNA sequencing. We applied this assay to 171 patients with large B-cell lymphomas. We compared the performance of PVs for disease detection to current ctDNA techniques, including CAPP-Seq and duplex sequencing. Results: To utilize PVs, mutations must occur within a typical cfDNA strand (~170bp). We measured the frequency of putative PVs in WGS, focusing on pairs of mutations occurring within <170bp. PVs were more frequent in NHLs than any other histology (median: DLBCL, 642; FL, 307; Burkitt, 89.5; CLL, 34; breast, 46; lung, colorectal, melanoma, bladder, cervical, head & neck < 10 per case; P < 0.001 for NHLs vs others). PVs in NHLs were enriched in single base substitution mutational signatures associated with activation-induced cytidine deaminase (AID) (SBS84 & 85). PVs in NHLs occurred in stereotyped regions, including canonical AID targets such as IGH, IGK, and IGL, as well as 44 other AID targets (Schmitz, NEJM 2018) (Fig 1B). We additionally identified novel regions not previously implicated as targets of AID, including LPP, XBP1, BZRAP1, and HLA-DQ. We designed an approach for enriching PVs from ~115kb (Phased variant Enrichment Sequencing, PhasE-Seq) and other regions recurrently mutated in B-NHLs. We compared PhasE-Seq and CAPP-Seq using tumor and plasma samples from 16 patients. Compared to CAPP-Seq, PhasE-Seq yielded more SNVs and PVs per case (median SNVs: 331 vs 114, P<0.001; PVs: 729 vs 222.5, P <0.001). We next applied PhasE-Seq to 171 patients with untreated lymphomas (DLBCL, 148; primary mediastinal B-cell lymphoma, PMBCL, 23) profiling 58 tumor and 171 plasma samples with matched germline. We observed significant differences in the distribution of PVs between subtypes - for example, GCB-DLBCL had more PVs in BCL2, MYC, and SGK1, while ABC-DLBCL had more in PIM1 and IGHV4-34 (Fig 1C). Similarly, we noted enrichment in PVs in PMBCL in CIITA, SOCS1, CD83, and ITPKB. We then compared PhasE-Seq to alternative methods for MRD detection. We used limiting dilutions of patient ctDNA down to 1:1,000,000 to establish the detection limit (LOD, Fig 1D). PhasE-Seq outperformed CAPP-Seq and duplex sequencing for recovery of expected tumor content, with a high degree of linearity down to ~1:1,000,000. We applied standard CAPP-Seq and PhasE-Seq to patient cfDNA samples after two cycles of front-line therapy (n=92). We previously reported a 2.5-log reduction in ctDNA as prognostic at this time-point (Kurtz, JCO 2018). Using CAPP-Seq, 58% (53/92) of samples were undetectable. Using PhasE-Seq, 30% (16/53) of samples not detected by CAPP-Seq had evidence of MRD, with levels as low as 2:1,000,000. In patients with ctDNA undetected by CAPP-Seq, detection by PhasE-Seq significantly stratified outcomes (Fig 1E). Conclusions: PVs are frequent in NHLs, likely due to AID, and correlate with disease biology. PhasE-Seq allows for superior detection of ctDNA, including MRD detection in the majority of patients after 2 cycles. Targeted sequencing of ctDNA should consider PVs to maximize detection and guide precision approaches. Figure 1: A) Structure of phased variants B) Distribution of putative PVs from WGS data C) Genomic enrichment in PVs in lymphoma subtypes D) Dilution series comparing PhasE-Seq, CAPP-Seq, and duplex sequencing E) Waterfall plot showing ctDNA level vs outcome; undetectable ctDNA by CAPP-Seq is highlighted F) EFS of patients with undetectable ctDNA by CAPP-Seq after 2 cycles, stratified by PhasE-Seq Disclosures Kurtz: Roche: Consultancy. Diehn:Roche: Consultancy; AstraZeneca: Consultancy; Novartis: Consultancy; BioNTech: Consultancy; Quanticell: Consultancy. Alizadeh:Pharmacyclics: Consultancy; Janssen: Consultancy; Genentech: Consultancy; Roche: Consultancy; Gilead: Consultancy; Celgene: Consultancy; Chugai: Consultancy; Pfizer: Research Funding.

2019 ◽  
Author(s):  
Maysun M. Hasan ◽  
Juber Patel ◽  
Ian Johnson ◽  
Fanli Meng ◽  
Grittney K. Tam ◽  
...  

2021 ◽  
Author(s):  
Shilpa Sampathi ◽  
Yelena Chernyavskaya ◽  
Meghan G. Haney ◽  
L. Henry Moore ◽  
Isabel A. Snyder ◽  
...  

AbstractAcute Lymphoblastic Leukemia (ALL) patients that have minimal residual disease (MRD) after therapy or infiltration of ALL into the central nervous system (CNS) are considered high risk. These patients are often given intensified and/or additional rounds of chemotherapy in the hopes of eliminating their disease. Current methods to diagnose MRD and CNS infiltration rely on detecting ALL cells in patient samples using pathology, flow cytometry, or isolation of ALL genomic DNA for next-generation sequencing. However, blasts may be present in the patient but not detectable in a bone marrow biopsy or cerebrospinal (CSF) fluid sample, leading to incorrect or delayed patient diagnosis. We have developed a nanopore sequencing assay to detect B-ALL-associated cell-free DNA in patient blood and CSF samples. Quantitation of B-cell specific VDJ recombination events in cell-free DNA samples defined B-ALL clonal heterogeneity. Monitoring cfDNA in blood and CSF samples allowed us to track the response of individual B-ALL clones throughout each patient’s course of treatment. Detection of cell-free DNA predicted the clinical diagnosis of MRD and CNS disease. We also identified patients diagnosed as CNS negative who had low levels of cell-free DNA in their CSF sample. These data suggest that cell-free DNA assays may be useful in detecting the presence of ALL in the patient even when blasts are not in the biofluid sample. In total, nanopore analysis of cell-free DNA is a simple, rapid, and inexpensive assay that can serve as a useful complement to traditional clinical diagnostic approaches in the treatment of ALL.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 29-29
Author(s):  
Juan Du ◽  
Baoan Chen ◽  
Wanting Qiang ◽  
Yanchun Jia ◽  
Jing Lu ◽  
...  

Background: Minimal residual disease (MRD) is becoming standard diagnostic care for multiple myeloma. Here we investigate cell-free DNA chromosomal instability as minimal invasive biomarker for minimal residual disease monitoring. Methods: 55 patients were recruited, including 47 newly diagnosed and 8 relapsed multiple myelomas. Plasma samples were collected before treatments, end of 2 cycle and 4 cycle of treatments. Treatment response was assessed by using IMWG criteria. Cell-free DNA was analyzed by illumine HiSeq X10, followed by chromosomal instability analyses by a customized bioinformatics workflow, ultrasensitive chromosomal aneuploidy detector (UCAD), and cfDNA CIN responding to treatment was summarized as cfDNA MRD INDEX. Results: In this cohort study, 53 (96.3%) patients were found with treatment responses after 4 cycle of treatments, including 16 (29.1%) complete, 13 (23.6%) very good partial, 17 (30.9%) partial and 6 (10.9%) marginal responses. The other 2 (3.64%) recurrent MM experiences disease progression. At baseline, plasma cfDNA chromosomal aberrations were found in 14/17(82.3%) multiple myeloma patients. Less patients were found with detectable chromosomal changes after treatments (58.8% after C2, 41.2% after C4, versus 82.3% before treatments, P<0.01). The treatment responses identified in plasma cfDNA were summarized as cfDNA MRD INDEX. Low cfDNA MRD INDEX predicts better treatment responses (Fisher exact test, Odds ratio=7.3, P=4.69e-05). And it predicts complete response with sensitivity 100% and specificity 83.3%, with cutoff value 0.044. Furthermore, cfDNA CIN MRD index were found linearly correlated with percentage of malignant plasma cells from bone marrow aspiration as examined by flow cytometry assay (R-square=0.883, P<0.01). Cutoff -0.06 predicts 100% MRD negatives at specificity 100%. Conclusions: Plasma cfDNA CIN might be used for monitoring multiple myeloma patients treatment responses, especially for predicting minimal residual diseases. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document