First Results of a 31-Gene Panel Targeted to Investigate Myeloid Malignancies by Next-Generation Amplicon Deep-Sequencing

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 883-883 ◽  
Author(s):  
Alexander Kohlmann ◽  
Sandra Weissmann ◽  
Ulrike Schoeck ◽  
Vera Grossmann ◽  
Wolfgang Kern ◽  
...  

Abstract Abstract 883 Introduction: Massively parallel next-generation sequencing data have changed the landscape of molecular mutations in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). The number of molecular markers used to characterize myeloid malignancies continues to constantly increase. As such, physicians and laboratories face a great unmet need to test panels of genes at a high level of sensitivity and throughput. Methods: We developed a sensitive next-generation deep-sequencing assay for routine diagnostics. In total, 31 genes with relevance in myeloid malignancies providing both favorable and adverse molecular prognostic information were chosen: ASXL1, BCOR, BCORL1, BRAF, CBL, DNMT3A, ETV6, EZH2, FLT3, IDH1, IDH2, JAK2, KDM6A, KIT, KRAS, NOTCH1, NPM1, NRAS, PHF6, PRPF40B, PTPN11, RUNX1, SF1, SF3A1, SF3B1, SRSF2, TET2, TP53, U2AF1, U2AF2, and ZRSR2. Targets of interest comprised either complete coding gene regions or hotspots. In summary, 1,375 amplicons were designed with a median length of 175 bp (range 109–194 bp), representing a total target sequence of 140.35 kb. The sequencing library was constructed starting off 2.2 μg genomic DNA per patient using a singleplex microdroplet-based assay (RainDance, Lexington, MA). Sequencing data was generated using the MiSeq instrument (Illumina, San Diego, CA) loading 4 patients per run. Using the 300 cycles sequencing-by-synthesis chemistry in median 6.099 millions of paired-end reads were generated per run. This resulted in a median coverage per gene of 1,766 reads (range 992-2,432). The total turn-around time of the analysis with this assay was less than 4 days. 49 clinically well-annotated patients harboring myeloid malignancies were analyzed during the evaluation phase. These included 9 acute myeloid leukemia (AML), 9 myelodysplastic syndrome (MDS), 13 chronic myelomonocytic leukemia (CMML), and 18 mixed phenotype acute leukemia, T/myeloid (MPAL-TM) cases. The median age was 69 years (range: 23 – 90 years). Results: In median, the coverage per amplicon harboring a mutation was 2,095-fold, thus enabling a sensitive detection of variants. In total, 146 mutations in 28 of the 31 genes were detected in 47/49 patients with a range of 1–7 mutations per case (median: 3). According to chromosome banding analysis 31/49 cases presented with a normal karyotype. In 30/31 cases with a normal karyotype at least one molecular mutation was observed using this screening panel. 42/146 mutations were detected with a clone size <20%, thus being detected only due to the higher sensitivity of this technique in comparison to direct capillary Sanger sequencing. In this cohort, the most frequently mutated genes were RUNX1 (14/49), DNMT3A (14/49), SRSF2 (11/49), ASXL1 (9/49), and TET2 (9/49). The mutation types comprised 97 missense, 17 duplications, 24 deletions, 5 insertions and 3 insertion/deletions alterations. Novel variants were verified using direct capillary Sanger sequencing (n=19) or sensitive amplicon deep-sequencing (n=65) (454 Life Sciences, Branford, CT). With respect to the technical limit of detecting larger insertions or deletions both a 27-bp insertion (RUNX1, p.Thr121delins9) and a 23-bp deletion variant (ASXL1, p.Glu635ArgfsX15) were successfully sequenced. The highest number of mutations was observed for CMML patients (mean of 3.6 per case; CMML vs remainder: P=0.201). Also, in CMML patients we observed the highest frequency of mutations in major splicing machinery genes such as SF1, SF3A1, SF3B1, SRSF2, U2AF1, U2AF2, and ZRSR2 (11/13 CMML, 84.6% vs 14/36 remainder cases, 38.9%; P<0.001). Importantly, a number of patients (39/49) was detected to harbor mutations in genes reported to be associated with decreased overall survival, both in AML (e.g. TP53, RUNX1, ASXL1, DNMT3A, IDH1, or TET2) and low- or intermediate-1 IPSS risk categories in MDS (e.g. ASXL1, EZH2, ETV6, RUNX1, TP53). As such, detecting these adverse somatic alterations may influence the course of therapy for these patients. Conclusion: We here demonstrated that microdroplet-based sample preparation enabled to target 31 candidate genes for next-generation sequencing in myeloid malignancies in a routine diagnostic environment. This approach provides the potential to screen for prognostically relevant mutations in a fast and comprehensive way providing actionable information suitable to guide therapy. Disclosures: Kohlmann: MLL Munich Leukemia Laboratory: Employment. Weissmann:MLL Munich Leukemia Laboratory: Employment. Schoeck:MLL Munich Leukemia Laboratory: Employment. Grossmann:MLL Munich Leukemia Laboratory: Employment. Kern:MLL Munich Leukemia Laboratory: Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership. Schnittger:MLL Munich Leukemia Laboratory: Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 657-657
Author(s):  
Alexander Kohlmann ◽  
Niroshan Nadarajah ◽  
Vera Grossmann ◽  
Tamara Alpermann ◽  
Wolfgang Kern ◽  
...  

Abstract Abstract 657 Introduction: RUNX1 mutations constitute disease-defining aberrations in acute myeloid leukemia (AML) and were demonstrated to be particularly frequent in secondary and de novo AML with normal karyotype or non-complex alterations and to confer an unfavorable prognosis. Monitoring minimal residual disease (MRD) in AML has been shown to provide prognostic information and is increasingly used for treatment decisions. A variety of molecular markers has been identified suitable for MRD assessment, yet there still is a lack of such markers in a significant number of patients. The use of RUNX1 mutations may bridge a gap. Aims: Patients and Methods: RUNX1 mutation screening was prospectively performed in 814 patients with AML at diagnosis (645 de novo, 109 s-AML, and 60 t-AML). The median age of the patients was 69.6 years (range: 1 – 93 years), including 375 female and 439 male patients, respectively. 50.5% (411/814) of cases presented with a normal karyotype, 38.8% (316/814) with non-complex cytogenetic alterations, 9.6% (78/814) with a complex aberrant karyotype, and 1.1% (9/814) with prognostically favorable cytogenetics. Mutation analysis was performed using a sensitive next-generation amplicon deep-sequencing assay (454 Life Sciences, Branford, CT). Moreover, in a subset of 44 AML patients and additional 59 retrospectively analyzed cases the prognostic impact of MRD levels of RUNX1 mutations was studied at a second time point after completion of intensive induction therapy (median sampling interval: 128 days after diagnosis; range 60 – 180 days). In these follow-up samples the RUNX1 mutations already detected at diagnosis were investigated with a higher coverage (835-fold median coverage) as compared to the diagnostic assessment (759-fold median coverage) resulting in a sensitivity level of 1%. Furthermore, in 57 patients paired samples from diagnosis and relapse were analyzed to assess the stability of RUNX1 mutations. Results: 211/814 patients (25.9%) were detected to carry RUNX1 mutations. The median clone size was 39% and revealed a significant heterogeneity ranging from 2% to 96%. 73.9% (156/211) of mutated patients carried one mutation only, whereas 26.1% (55/211) harbored two (n=46) or more (n=9) mutations. In detail, the 211 patients harbored a total number of 275 alterations in RUNX1: 42.5% (117/275) frame-shift mutations, 34.9% (96/275) missense, 14.2% (39/275) nonsense, 4.4% (12/275) exon-skipping/splicing, and 4.0% (11/275) in-frame insertion/deletion alterations, respectively. Regarding MRD assessment, patients were separated according to the median MRD level (3.92%; range 0.03% - 48.00%) into “good responders” (n=78) with MRD levels below 3.92% and “poor responders” (n=25) with MRD levels above 3.92%. This resulted in significant differences in both event-free survival (median 21.4 vs 5.7 months, p<0.001) and overall survival (73.3% vs 66.1% at 2 years, p=0.016). Moreover, in 57 cases the stability of individual RUNX1 mutations was studied at the time of relapse. In 46/57 (80.7%) cases the same alterations detected at diagnosis were present at relapse, whilst in 2/57 (3.5%) cases the RUNX1 mutation from the diagnostic sample was no longer detectable at relapse. Importantly, in 7/57 (12.3%) patients novel RUNX1 mutations were detected in regions different from those affected at diagnosis. Conclusion: Next-generation deep-sequencing accurately detects and quantifies RUNX1 mutations in AML with high sensitivity. RUNX1 mutations qualify as patient-specific markers for individualized disease monitoring. Thus, the measurement of mutation load by next-generation sequencing may contribute to refine the assignment into distinct risk categories in AML. Analysis of RUNX1 mutations should be considered for the complete coding region at relapse to detect new RUNX1 mutations. Disclosures: Kohlmann: MLL Munich Leukemia Laboratory: Employment; Roche Diagnostics: Honoraria. Nadarajah:MLL Munich Leukemia Laboratory: Employment. Grossmann:MLL Munich Leukemia Laboratory: Employment. Alpermann:MLL Munich Leukemia Laboratory: Employment. Kern:MLL Munich Leukemia Laboratory: Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership. Schnittger:MLL Munich Leukemia Laboratory: Equity Ownership.


2020 ◽  
Author(s):  
Biao Wang ◽  
Bin Yang ◽  
Wei Wu ◽  
Xuan Liu ◽  
Haiqian Li

Abstract The purpose of this study was to analyze association between next-generation sequencing (NGS) genomic profile and conventional MICM characteristics in patients with acute myeloid leukemia (AML) with NPM1 mutation (NPM1mut). We selected 238 NPM1mut patients with available NGS information on 112 genes related to blood diseases, using χ2 and Mann-Whitney U test to analyze the distribution correlation between genomic alterations and MICM parameters. Compared with NPM1mut/FLT3-ITD(−), the NPM1mut/FLT3-ITD(+) group presented a slightly common M5 [78/143 (54.5%) vs. 64/95 (67.4%); P=0.048], more higher CD34 and CD7 expression rates (CD34: 20.6% vs. 47.9%, P<0.001; CD7: 29.9% vs. 61.5%, P<0.001), and lack of favorable- and adverse-risk karyotypes (6.4% vs. 0%; P=0.031). In entire NPM1mut cohort, totaling 240 NPM1 mutation events were identified, of whom 10 (10/240, 4.2%) were missense types. When confining analysis to the 205 cases with NPM1mut insertions/deletions type and normal karyotype, multivariable logistic analysis showed that FLT3-ITD was positively correlated with CD34 and CD7 expression (HR=5.29 [95% CI 2.64-10.60], P<0.001; HR=3.47 [95% CI 1.79-6.73], P<0.001; respectively). Ras-pathway mutation was positively correlated with HLA-DR expression (HR=4.05 [95% CI 1.70-9.63], P=0.002), and KRAS mutation negatively with MPO expression (HR=0.18 [95% CI 0.05-0.62], P=0.007). DNMT3A-R882 was positively correlated with CD7 and HLA-DR expression (HR=3.59 [95% CI 1.80-7.16], P<0.001; HR=13.41 [95% CI 4.56-39.45], P<0.001; respectively). DNMT3A mutation was negatively correlated with MPO expression (HR=0.35 [95% CI 1.48-8.38], P=0.004). TET2/IDH1 mutations were negatively correlated with CD34 and CD7 expression (HR=0.26 [95% CI 0.11-0.62], P=0.002; HR=0.30 [95% CI 0.14-0.62], P=0.001; respectively), and positively with MPO expression (HR=3.52 [95% CI 1.48-8.38], P=0.004). NPM1mut co-existing mutations in signaling pathways (FLT3-ITD and Ras-pathway) and methylation modifiers (DNMT3A and TET2/IDH1) are linked with the expression of CD34, CD7, HLA-DR and MPO, thereby providing a mechanism explanation for the immunophenotypic heterogeneity of this AML entity.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 867-867 ◽  
Author(s):  
Alexander Kohlmann ◽  
Andreas Roller ◽  
Andreia Albuquerque ◽  
Sabrina Kuznia ◽  
Sandra Weissmann ◽  
...  

Abstract Introduction Massively parallel next-generation sequencing (NGS) data have changed the landscape of molecular mutations in chronic lymphocytic leukemia (CLL). The number of molecular markers continues to constantly increase. As such, physicians and laboratories face a great unmet yet challenging need to test panels of genes at a high level of sensitivity. Aim To develop an assay that is easily adoptable to adjust gene targets and amplicons according to current state-of-the-art evidence regarding the published landscape of mutations in CLL. Methods We developed a sensitive deep-sequencing assay for routine diagnostics. In total, 13 genes with relevance in CLL providing in part adverse prognostic information were chosen: ATM, BIRC3, BRAF (V600), FBXW7, KLHL6, KRAS, NOTCH1 (PEST domain), NRAS, MYD88, POT1, SF3B1 (HEAT repeats), TP53, and XPO1. Targets of interest comprised either complete coding gene regions or hotspots. In summary, 323 amplicons were designed with a median length of 204 bp (range 150-240 bp), representing a total target sequence of 39.36 kb. The sequencing library was constructed starting off 2.2 μg genomic DNA per patient using a single-plex microdroplet-based assay (RainDance, Lexington, MA). Sequencing data was generated using the MiSeq instrument (Illumina, San Diego, CA) loading up to 10 patients per run. The total turn-around time of the assay was less than 5 days. As a proof-of-principle cohort, 18 clinically well-annotated CLL patients were analyzed during the evaluation phase. The median age was 78 years (range: 52 – 87 years). Results Using the 500 cycles sequencing by-synthesis chemistry, in median 7,262 millions of paired-end reads were generated per run. This resulted in a median coverage per gene of 7,476 reads (range: 5,595 - 10,337). (1) In this cohort of 18 cases, a total of 71 mutation analyses had already been previously performed for eight of the 13 genes using either capillary Sanger sequencing or alternative amplicon deep-sequencing assays (454 LifeSciences or Illumina MiSeq). In detail, in these 8 genes these 71 assays detected 56 known polymorphisms or mutations in ATM (n=8), BIRC3 (n=6), FBXW7 (n=4), MYD88 (n=4), NOTCH1 (n=10), SF3B1 (n=5), TP53 (n=14), and XPO1 (n=4) and 28 analyses revealed a wild-type status. When comparing these results with data obtained using the 13-gene NGS panel, in all 84/84 (100%) parallel assessments concordant results were obtained underlining the robustness of this assay. (2) Overall and extending the previous results, the comprehensive 13-gene NGS panel then detected in 18/18 patients a total of 46 mutations in 10 of the 13 genes with a range of 1-5 mutations per case (median: 2). The mutation types comprised 22 missense, 4 nonsense, 16 frame-shifts, 3 insertions and 1 splice-site alterations. In median, the coverage per variant was 10,390-fold, thus enabling a sensitive detection of mutations at a lower limit of detection set at 3%. The mutation burden ranged from 3.0% to 62.0%. 18/46 (39.13%) mutations were detected with a clone size <20%, thus being detected only due to the higher sensitivity of this assay in comparison to direct capillary Sanger sequencing. With respect to the technical limit of detecting larger alterations, a 34 bp deletion variant (NOTCH1; c.7403_7436del) was successfully sequenced. Moreover, a common theme in hematological malignancies is the emergence of novel prognostic scoring systems, integrating molecular mutations and cytogenetic lesions into revised survival prediction models. Importantly, a number of patients (14/18) was detected to harbor mutations in genes reported to be associated with decreased overall survival, both in high-risk (e.g. TP53, BIRC3) and intermediate-risk (NOTCH1, SF3B1) categories according to Rossi et al., 2013 (Blood;121:1403-12). As such, detecting these adverse somatic alterations may influence the course of therapy for these patients underlining the utility of such a screening panel. Conclusion We demonstrated that microdroplet-based sample preparation enabled to robustly target 13 genes for next-generation sequencing in a routine diagnostics environment. This included also larger gene targets such as ATM, being represented by 119 amplicons. Thus, this approach provides the potential to screen for prognostically relevant mutations in all CLL patients in a fast and comprehensive way providing actionable information suitable to guide therapy. Disclosures: Kohlmann MLL Munich Leukemia Laboratory: Employment. Roller:MLL Munich Leukemia Laboratory: Employment. Albuquerque:MLL Munich Leukemia Laboratory: Employment. Kuznia:MLL Munich Leukemia Laboratory: Employment. Weissmann:MLL Munich Leukemia Laboratory: Employment. Jeromin:MLL Munich Leukemia Laboratory: Employment. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.


2017 ◽  
Vol 65 (8) ◽  
pp. 1155-1158 ◽  
Author(s):  
Thiago Rodrigo de Noronha ◽  
Miguel Mitne-Neto ◽  
Maria de Lourdes Chauffaille

Karyotype (KT) aberrations are important prognostic factors for acute myeloid leukemia (AML); however, around 50% of cases present normal results. Single nucleotide polymorphism array can detect chromosomal gains, losses or uniparental disomy that are invisible to KT, thus improving patients’ risk assessment. However, when both tests are normal, important driver mutations can be detected by the use of next-generation sequencing (NGS). Fourteen adult patients with AML with normal cytogenetics were investigated by NGS for 19 AML-related genes. Every patient presented at least one mutation:DNMT3Ain nine patients;IDH2in six;IDH1in three;NRASandNPM1in two; andTET2,ASXL1,PTPN11, andRUNX1in one patient. No mutations were found inFLT3,KIT,JAK2,CEBPA,GATA2,TP53,BRAF,CBL,KRAS,andWT1genes. Twelve patients (86%) had at least one mutation in genes related with DNA methylation (DNMT3A,IDH1,IDH2,andTET2), which is involved in regulation of gene expression and genomic stability. All patients could be reclassified based on genomic status and nine had their prognosis modified. In summary, NGS offers insights into the molecular pathogenesis and biology of cytogenetically normal AML in Brazilian patients, indicating that the prognosis could be further stratified by different mutation combinations. This study shows a different frequency of mutations in Brazilian population that should be confirmed.


Author(s):  
Edit Porpaczy ◽  
Wolfgang R. Sperr ◽  
Renate Thalhammer ◽  
Gerlinde Mitterbauer-Hohendanner ◽  
Leonhard Müllauer ◽  
...  

AbstractMixed phenotype acute leukemia (MPAL) is an uncommon disease characterized by currently only limited knowledge concerning biology, clinical presentation, and treatment outcome. We here describe a most unusual case of simultaneous occurrence of T-lymphoblastic lymphoma in cervical and mediastinal lymph nodes and acute myeloid leukemia in the bone marrow (BM) successfully treated with allogeneic stem cell transplantation (SCT). Although the blasts in both locations showed additional aberrant expression of other lineage markers (even B-cell markers), diagnostic criteria of MPAL were not fulfilled either in the LN or in the BM. We performed next generation sequencing (NGS) with the objective to look for common genetic aberrations in both tissues. Histology, immunohistochemistry, flow cytometry, AML-associated genetic alterations (FLT3, NPM1, KIT D816V, CEPBA), and clonal T-cell receptor β and γ gene rearrangements were performed according to routine diagnostic workflows. Next generation sequencing and Sanger sequencing were additionally performed in BM and LN. Somatic mutation in the EZH2 gene (p.(Arg684Cys)) was detected in the BM by NGS, and the same mutation was found in the LN. Since an identical genetic aberration (EZH2 mutation) was detected in both locations, a common progenitor with regional dependent differentiation may be involved.


2020 ◽  
Vol 61 (13) ◽  
pp. 3269-3271
Author(s):  
Wade L. Schulz ◽  
Henry M. Rinder ◽  
Thomas J. S. Durant ◽  
Christopher A. Tormey ◽  
Richard Torres ◽  
...  

2019 ◽  
Vol 85 ◽  
pp. S69-S70
Author(s):  
A. Bolaman ◽  
İ. Erdoğdu ◽  
A. Turgutkaya ◽  
C. Selim ◽  
A. Eroglu Kucukerdiler ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document