Molecular Measurable Residual Disease Detection in Acute Myeloid Leukemia Using Error Corrected Next Generation Sequencing

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 5-6
Author(s):  
Nikhil Patkar ◽  
Anam Fatima Shaikh ◽  
Chinmayee Kakirde ◽  
Rakhi Salve ◽  
Prasanna Bhanshe ◽  
...  

Introduction The monitoring of a patient's response to chemotherapy, called, measurable residual disease (MRD) is one of the most important predictors of outcome in Acute Myeloid Leukemia (AML). Although universally applicable, FCM-MRD for AML suffers from low sensitivity as compared to precursor B lineage acute lymphoblastic leukemia. Here, we evaluated the clinical utility of error corrected next generation sequencing (NGS) to detect MRD (NGS-MRD) in AML using single molecule molecular inversion probes (smMIPS). We compare NGS-MRD and FCM-MRD and determine their impact on patient outcome. We demonstrate that error corrected NGS-MRD at early timepoints in therapy is an independent and significant predictor of outcome in patients of AML treated with conventional therapies. Methods We created a 35 gene "hotspot" panel comprising of a pool of 302 smMIPS. In brief, this panel covers regions of 35 commonly mutated genes in AML.FLT3-ITD were detected using a novel one-step PCR based NGS assay. Post mapping, singleton reads (originating from one UMI) were discarded and consensus family based variant calling was performed. We then created a site and mutation specific error model to ascertain the relevance of an observed variant at each site. A limit of detection (LOD) experiment demonstrated a lower detection limit of 0.05%. For FLT3-ITD the LOD was 0.002%. A total of 393 adult patients of AMLwere accrued over a period of six years.Patients were treated with standard 3+7 induction followed by 3 doses of HiDAC. Allogeneic bone marrow transplantation was offered where feasible. Somatic mutations at diagnosis were evaluated using a smMIPS based 50 gene myeloid panel which was applicable to 327 patients [83.2% of AMLs, median 2 mutations per case (range 1 - 6 trackable mutations)].MRD assessment could be performed in 201 adult patients of AML in morphological remission (not performed in the rest because of suboptimal quality DNA at MRD time points or missing sample).Samples were sequenced on multiple S4 flow cells of a NovaSeq 6000 using 150PE chemistry.FCM-MRD was obtained from the bone marrow at end of induction (PI, n=200) and end of first consolidation cycle (PC, n=98). NGS-MRD sample also obtained at the same time points (PI, n=196& PC, n=127) from the bone marrow (n=266) or peripheral blood (n=45). Results The interaction of mutations that were trackable at diagnosis can be seen in Figure 1A. A total of 345 mutations could be detected in 196 patients (Figure 1B) with a median VAF of 1.01% [0.82% after exclusion of mutations in DNMT3A, TET2, ASXL1 (DTA) genes; (median of 2 mutations for PI and one for PC timepoint)]. The median consensus read coverage was 11,127 for the smMIPS assay, whereas for the FLT3-ITD assay it was 13,96,366.The median follow-up of the cohort was 42.3 months. The presence of NGS-MRD (70.9%) was associated with inferior overall survival (OS; p=0.001) [hazard ratio(HR)- 2.24; 95% confidence interval (CI)- 1.47 to 3.43] and relapse free survival (RFS; p=0.0002) [HR- 2.28; 95% CI- 1.58 to 3.31] at PI time point as well as PC time points [40.94% positive; OS (p=0.008)(HR- 1.92; 95% CI- 1.14 to 3.22) and RFS (p=0.004)(HR- 1.90; 95% CI- 1.18 to 3.05)].Similarly, FCM-MRD (44%) was predictive of inferior OS (p=0.0002)(HR- 2.08; 95% CI- 1.38 to 3.13)and RFS (p=0.0008)(HR- 1.81; 95% CI- 1.26 to 2.60) at PI as well as PC time points [21.4% positive, OS (p=0.04)(HR- 1.87; 95% CI- 0.89 to 3.91) and RFS (p=0.001)(HR- 2.38; 95% CI- 1.17 to 4.81)]. On multivariate analysis post induction NGS MRD emerged as the most important independent prognostic factor predictive of inferior outcome for OS [HR- 1.94; 95% CI-1.15 to 3.27; (p<0.0001)]as well asRFS[HR-2.05; 95% CI-1.30 to 3.23; (p<0.0001)].On incorporating results combining both the MRD modalities,patients that were positive by both techniqueshad a significantly inferior outcome with respect to OS (p=0.0002; HR- 4.66; 95% CI- 2.71 to 8.0)and RFS (p=0.0001; HR- 4.03; 95% CI- 2.51 to 6.47) at PI timepoint as well as PC timepoint [OS (p=0.02; HR- 3.73; 95% CI- 1.07 to 12.97) and RFS (p=0.0015; HR- 4.17; 95% CI- 1.27 to 13.7)] as compared to patients negative by both modalities (Figure 1E,F) Conclusion In conclusion, we demonstrate that error corrected panel-based sequencing is feasible for MRD monitoring in AML and may offer an advantage over existing techniques. Maximum clinical utility may be leveraged by combining FCM and NGS modalities. Figure Disclosures No relevant conflicts of interest to declare.

2021 ◽  
Vol 11 (6) ◽  
Author(s):  
Hee-Je Kim ◽  
Yonggoo Kim ◽  
Dain Kang ◽  
Hoon Seok Kim ◽  
Jong-Mi Lee ◽  
...  

AbstractGiven limited studies on next-generation sequencing-based measurable residual disease (NGS-MRD) in acute myeloid leukemia (AML) patients after allogeneic hematopoietic stem cell transplantation (allo-HSCT), we longitudinally collected samples before and after allo-HSCT from two independent prospective cohorts (n = 132) and investigated the prognostic impact of amplicon-based NGS assessment. Persistent mutations were detected pre-HSCT (43%) and 1 month after HSCT (post-HSCT-1m, 20%). All persistent mutations at both pre-HSCT and post-HSCT-1m were significantly associated with post-transplant relapse and worse overall survival. Changes in MRD status from pre-HSCT to post-HSCT-1m indicated a higher risk for relapse and death. Isolated detectable mutations in genes associated with clonal hematopoiesis were also significant predictors of post-transplant relapse. The optimal time point of NGS-MRD assessment depended on the conditioning intensity (pre-HSCT for myeloablative conditioning and post-HSCT-1m for reduced-intensity conditioning). Serial NGS-MRD monitoring revealed that most residual clones at both pre-HSCT and post-HSCT-1m in patients who never relapsed disappeared after allo-HSCT. Reappearance of mutant clones before overt relapse was detected by the NGS-MRD assay. Taken together, NGS-MRD detection has a prognostic value at both pre-HSCT and post-HSCT-1m, regardless of the mutation type, depending on the conditioning intensity. Serial NGS-MRD monitoring was feasible to compensate for the limited performance of the NGS-MRD assay.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1392-1392
Author(s):  
Parsa Hodjat ◽  
Kankana Ghosh ◽  
Priyanka Priyanka ◽  
Beenu Thakral ◽  
Keyur P. Patel ◽  
...  

Abstract INTRODUCTION Acute myeloid leukemia (AML) is known to have numerous genomic aberrations that predict response to treatment and overall survival. We aimed to assess various mutations in newly diagnosed AML cases by next generation sequencing (NGS) and their association with various well-established clinicopathologic parameters and Medical Research Council (MRC) risk groups. MATERIALS AND METHODS We performed molecular studies on DNA extracted from bone marrow aspirate specimens in 276 newly diagnosed treatment na•ve AML patients presenting at a single referral institution from 08/2013 to 03/2015 as part of routine clinical work up in a CLIA certified molecular diagnostics laboratory. Cases met criteria for AML per WHO 2008 criteria. The entire coding sequences of 28 genes (ABL1, ASXL1, BRAF, DNMT3A, EGFR, EZH2, FLT3, GATA1, GATA2, HRAS, IDH1, IDH2, IKZF2, JAK2, KIT, KRAS, MDM2, MLL, MPL, MYD88, NOTCH1, NPM1, NRAS, PTPN11, RUNX1, TET2, TP53, WT1) were sequenced using a NGS-based custom-designed assay using TruSeq chemistry on Illumina MiSeq platform. FLT3 internal tandem duplications (ITD) and codon 835/836 point mutation were detected by PCR followed by capillary electrophoresis. CEBPA mutation analysis was performed on 262 patients by PCR followed by Sanger sequencing. Cases were categorized as favorable, intermediate and adverse groups as per revised MRC cytogenetic risk group classification. RESULTS Median age was 67 years. Patients included 167 (60.5%) males and 109 (39.5%) females. 38 (14%) and 6 (2%) patients had prior diagnosis of myelodysplastic syndrome and myeloproliferative neoplasms respectively. Hematologic parameters are as follows [median (range)]: Hb 8.7 g/dL (2.8-13.9), platelets 50.5 K/μ L (1-1109), WBC 5.4 K/μ L (0.4-620.4), ANC 0.9 K/μ L (0-145.7), AMC 0.3 K/μ L (0-98.1). Bone marrow (BM) blast % [median (range)] was 45.5% (5-96). LDH was 733 IU/dL (225-13156). Of 275 patients with cytogenetic analysis performed, 98 (35.64%) had diploid karyotype, 75 (27.27%) had one, 38 (13.82%) had two, 8 (2.91%) had three, 56 (20.36%) had > three abnormalities, 75 (27.27%) had monosomies and 62 (22.55%) had trisomies. Of 34 cases classified as AML with recurrent cytogenetic abnormalities per WHO 2008, 10 (3.64%) had t(8;21), 13 (4.73%) had inv(16), 1 (0.36%) had t(15;17), 3 (1.09%) had inv (3), 4 (1.45%) had t(9;11)(p22;q23) and 3 (1.09%) had t(6;9)(p23;q34). MRC risk categorization of the cases was as follows: favorable 24 (8.72%), intermediate 161 (58.55%) and adverse 90 (32.73%). Mutations identified by NGS are as detailed in Table 1. Of 56 patients with FLT3 mutations detected by PCR, the breakdown is as follows: FLT3 ITD (39, 14.13%), FLT3 D835 (16, 5.80%), FLT3, ITD + D835 (1, 0.36%). Of 262 patients assessed, CEBPA mutation was detected in 26 (9.92%). Thirty one (11.23%) cases had no mutations detected in the genes analyzed by NGS or PCR, 93 (33.70%) had mutations in one, 80 (28.98%) in two, 42 (15.22%) in three and 30 (10.87%) in > three genes. We found positive associations between mutated genes and various parameters as detailed in Table 2. CONCLUSIONS: AML is a heterogeneous group of myeloid neoplasms at the genetic level. Multiple genetic mutations in a large subset of cases likely indicate clonal evolution. A subset of mutations has significant association with well-established clinico-pathologic parameters like WBC. With longer follow-up, we could use this data to refine prognostic models for AML. Table 1. Genes Number of Cases Percentage of Cases FLT3 61 22.10 NPM1 48 17.39 NRAS 48 17.39 DNMT3A 47 17.03 TP53 45 16.30 IDH2 40 14.49 IDH1 33 11.96 TET2 32 11.59 ASXL1 30 10.87 RUNX1 30 10.87 PTPN11 13 4.71 KRAS 11 3.99 KIT 8 2.90 WT1 8 2.90 GATA2 7 2.54 EZH2 6 2.17 JAK2 4 1.45 MPL 2 0.72 ABL1 1 0.36 EGFR 1 0.36 GATA1 1 0.36 IKZF2 1 0.36 MDM2 1 0.36 MLL 1 0.36 MYD88 1 0.36 NOTCH1 1 0.36 Table 1. Mutated genes p value Hb NRAS, NPM1 <0.05, <0.04 Platelets TP53, IDH2 <0.03, <0.02 WBC FLT3, NRAS, TP53 <0.05, <0.05, <0.05 AMC NRAS, NPM1, TP53 <0.001, <0.02, <0.02 ABC FLT3 NPM1 <0.049, <0.02 PB blast % FLT3, NPM1, TP53, CEBPA <0.000, <0.002, <0.005, <0.000 BM blast % FLT3, NRAS, NPM1, TP53, IDH1, CEBPA >0.000, <0.0000, <0.014, <0.004, <0.002, <0.012 AMC: absolute monocyte count, ABC: absolute basophil count, PB: peripheral blood, BM: bone marrow Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document