Whole Genome Sequencing of Therapy-Related Acute Myeloid Leukemia

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 784-784
Author(s):  
Giridharan Ramsingh ◽  
Dong Shen ◽  
Tamara Lamprecht ◽  
Sharon Heath ◽  
Robert S. Fulton ◽  
...  

Abstract Abstract 784 Whole Genome Sequencing of Therapy-Related Acute Myeloid Leukemia Giridharan Ramsingh, Dong Shen, Tamara L. Lamprecht, Sharon E. Heath, Robert S. Fulton, Elaine Mardis, Li Ding, Peter Westervelt, John Welch, Matthew J. Walter, Timothy A. Graubert, John F. DiPersio, Timothy J. Ley, Richard K. Wilson, and Daniel C. Link. Therapy related therapy-related acute myeloid leukemia (t-AML) accounts for 10–20% of all new cases of AML, and its incidence is rising. A fundamental difference in the pathogenesis of de novo AML and t-AML is prior treatment with chemotherapy and/or radiotherapy. The exposure of hematopoietic stem/progenitors cells (HSPCs) to this genotoxic stress is hypothesized to alter the number and spectrum of mutations that arise in t-AML. Moreover, the genotoxic stress may exert selective pressure to expand those HSPC clones that are inherently resistant to chemotherapy, a common feature in t-AML. To test these hypotheses, we sequenced the genomes of 23 cases of t-AML and compared them to the genomes of 24 cases of de novo AML, which we recently reported (Welch et al., Cell, July 2012). We choose to focus our initial studies on the subset of t-AML with normal cytogenetics or simple balanced translocations. Specifically, MLL gene rearrangements were observed in 22% of cases, other balanced translocations in 22%, trisomy 8 in 22%, normal karyotype in 31%, and a complex karyotype in a single case. All patients had received prior alkylator chemotherapy (62%), topoisomerase inhibitor chemotherapy (65%), or radiotherapy (77%). To identify somatic mutations, whole genome sequencing was performed on leukemic bone marrow (average 65% blasts) and skin (normal) DNA. Average haploid coverage was 37.5X and 34.7X for the leukemia and skin genomes, respectively. All somatic mutations were verified using patient-specific custom NimbleGen capture arrays, followed by Illumina sequencing. Although the total number of somatic single nucleotide variants in older patients (>50 years) with t-AML was similar to that observed in de novo AML (484 ± 68 vs. 506 ± 45, respectively), significantly more mutations were present in younger (≤ 50 years) patients with t-AML (743 ± 228) compared with de novo AML (336 ± 179, P=0.04). Exposure to chemotherapy is associated with an increased rate of transversions in relapsed AML (Ding et al., Nature 2012). However, the percentage of somatic mutations that were transversions in t-AML (35.8 ± 1.91%) was similar to that seen in de novo AML (33.5 ± 0.93%), regardless of age. In the 23 t-AML genomes, we identified recurring mutations (present in at least 2 cases) in 20 genes. Many of these mutations were also observed in de novo AML genomes (Figure 1). The most commonly mutated gene in t-AML was TET2, which was mutated in 35% of cases. Of interest, missense mutations of the ABC transporter gene ABCG2 were significantly enriched in t-AML (2/23, 8.7%) compared with de novo AML (0 in 200 cases, P=0.01). ABCG2 (also known as breast cancer resistance protein, BCRP) has been implicated in chemotherapy resistance. ABCG2 is expressed at high levels in hematopoietic stem cells, where it is known to function as a key drug transporter. Studies are underway to define the frequency of ABCG2 mutations (and other ABC transporter genes) in a larger cohort of t-AML, including cases with alterations in chromosome 5 or 7 or with complex cytogenetic abnormalities. In summary, in younger patients with t-AML, the mutational burden is higher than that of de novo AML patients, possibly reflecting prior exposure to chemoradiotherapy, though no increase in transversions was observed. Mutations of ABCG2 may contribute to chemotherapy resistance in a subset of t-AML. Figure 1. Recurring mutations in t-AML (n = 23) compared with de novo AML (n = 24). Figure 1. Recurring mutations in t-AML (n = 23) compared with de novo AML (n = 24). Disclosures: Ley: Washington University: Patents & Royalties.

Blood ◽  
2015 ◽  
Vol 125 (9) ◽  
pp. 1367-1376 ◽  
Author(s):  
R. Coleman Lindsley ◽  
Brenton G. Mar ◽  
Emanuele Mazzola ◽  
Peter V. Grauman ◽  
Sarah Shareef ◽  
...  

Key Points The presence of a mutation in SRSF2, SF3B1, U2AF1, ZRSR2, ASXL1, EZH2, BCOR, or STAG2 is highly specific for secondary AML. Secondary-type mutations define an s-AML–like disease within t-AML and elderly de novo AML that underlies clinical heterogeneity.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Weijia Fu ◽  
Aijie Huang ◽  
Hui Cheng ◽  
Yanrong Luo ◽  
Lei Gao ◽  
...  

Abstract Background The nucleoporin 98 (NUP98)-paired related homeobox 1 (PMX1) fusion gene, which results from t(1;11)(q23;p15), is rare in patients with acute myeloid leukemia (AML). Currently, only two cases of chronic myeloid leukemia in the accelerated phase or blast crisis and three cases of therapy-related AML have been reported. Here, we first report a patient with de novo AML carrying the NUP98-PMX1 fusion gene. Case presentation A 49-year-old man diagnosed with AML presented the karyotype 46,XY,t(1;11)(q23;p15)[20] in bone marrow (BM) cells. Fluorescence in situ hybridization analysis using dual-color break-apart probes showed the typical signal pattern. Reverse transcription-polymerase chain reaction (RT-PCR) analysis suggested the presence of the NUP98-PMX1 fusion transcript. The patient received idarubicin and cytarabine as induction chemotherapy. After 3 weeks, the BM aspirate showed complete remission, and the RT-PCR result for the NUP98-PMX1 fusion gene was negative. Subsequently, the patient received three cycles of high-dose Ara-c as consolidation chemotherapy, after which he underwent partially matched (human leukocyte antigen–DP locus mismatch) unrelated allogeneic hematopoietic stem cell transplantation (HSCT). The follow-up period ended on September 30, 2020 (6 months after HSCT), and the patient exhibited no recurrence or transplantation-related complications. Conclusion This is the first report of a patient with de novo AML carrying the NUP98-PMX1 fusion gene. The reported case may contribute to a more comprehensive profile of the NUP98-PMX1 rearrangement, but mechanistic studies are warranted to fully understand the role of this fusion gene in leukemia pathogenesis.


2021 ◽  
Vol 5 (3) ◽  
pp. 900-912
Author(s):  
Svea Stratmann ◽  
Sara A. Yones ◽  
Markus Mayrhofer ◽  
Nina Norgren ◽  
Aron Skaftason ◽  
...  

Abstract Relapse is the leading cause of death of adult and pediatric patients with acute myeloid leukemia (AML). Numerous studies have helped to elucidate the complex mutational landscape at diagnosis of AML, leading to improved risk stratification and new therapeutic options. However, multi–whole-genome studies of adult and pediatric AML at relapse are necessary for further advances. To this end, we performed whole-genome and whole-exome sequencing analyses of longitudinal diagnosis, relapse, and/or primary resistant specimens from 48 adult and 25 pediatric patients with AML. We identified mutations recurrently gained at relapse in ARID1A and CSF1R, both of which represent potentially actionable therapeutic alternatives. Further, we report specific differences in the mutational spectrum between adult vs pediatric relapsed AML, with MGA and H3F3A p.Lys28Met mutations recurrently found at relapse in adults, whereas internal tandem duplications in UBTF were identified solely in children. Finally, our study revealed recurrent mutations in IKZF1, KANSL1, and NIPBL at relapse. All of the mentioned genes have either never been reported at diagnosis in de novo AML or have been reported at low frequency, suggesting important roles for these alterations predominantly in disease progression and/or resistance to therapy. Our findings shed further light on the complexity of relapsed AML and identified previously unappreciated alterations that may lead to improved outcomes through personalized medicine.


Blood ◽  
2008 ◽  
Vol 111 (9) ◽  
pp. 4809-4812 ◽  
Author(s):  
Zhifu Xiang ◽  
Yu Zhao ◽  
Vesselin Mitaksov ◽  
Daved H. Fremont ◽  
Yumi Kasai ◽  
...  

Abstract Somatic mutations in JAK2 are frequently found in myeloproliferative diseases, and gain-of-function JAK3 alleles have been identified in M7 acute myeloid leukemia (AML), but a role for JAK1 in AML has not been described. We screened the entire coding region of JAK1 by total exonic resequencing of bone marrow DNA samples from 94 patients with de novo AML. We identified 2 novel somatic mutations in highly conserved residues of the JAK1 gene (T478S, V623A), in 2 separate patients and confirmed these by resequencing germ line DNA samples from the same patients. Overexpression of mutant JAK1 did not transform primary murine cells in standard assays, but compared with wild-type JAK1, JAK1T478S, and JAK1V623A expression was associated with increased STAT1 activation in response to type I interferon and activation of multiple downstream signaling pathways. This is the first report to demonstrate somatic JAK1 mutations in AML and suggests that JAK1 mutations may function as disease-modifying mutations in AML pathogenesis.


Blood ◽  
2008 ◽  
Vol 111 (9) ◽  
pp. 4797-4808 ◽  
Author(s):  
Michael H. Tomasson ◽  
Zhifu Xiang ◽  
Richard Walgren ◽  
Yu Zhao ◽  
Yumi Kasai ◽  
...  

Abstract Activating mutations in tyrosine kinase (TK) genes (eg, FLT3 and KIT) are found in more than 30% of patients with de novo acute myeloid leukemia (AML); many groups have speculated that mutations in other TK genes may be present in the remaining 70%. We performed high-throughput resequencing of the kinase domains of 26 TK genes (11 receptor TK; 15 cytoplasmic TK) expressed in most AML patients using genomic DNA from the bone marrow (tumor) and matched skin biopsy samples (“germline”) from 94 patients with de novo AML; sequence variants were validated in an additional 94 AML tumor samples (14.3 million base pairs of sequence were obtained and analyzed). We identified known somatic mutations in FLT3, KIT, and JAK2 TK genes at the expected frequencies and found 4 novel somatic mutations, JAK1V623A, JAK1T478S, DDR1A803V, and NTRK1S677N, once each in 4 respective patients of 188 tested. We also identified novel germline sequence changes encoding amino acid substitutions (ie, nonsynonymous changes) in 14 TK genes, including TYK2, which had the largest number of nonsynonymous sequence variants (11 total detected). Additional studies will be required to define the roles that these somatic and germline TK gene variants play in AML pathogenesis.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 823-823
Author(s):  
Zhifu Xiang ◽  
Yu Zhao ◽  
Vesselin Mitaksov ◽  
Daved H. Fremont ◽  
Yumi Kasai ◽  
...  

Abstract Activating mutations in receptor tyrosine kinase (RTK) genes (including FLT3 and KIT) occur in more than 30% of newly diagnosed patients with acute myeloid leukemia (AML); we and others have speculated that mutations in other TK genes may be present in the remaining 70%. We therefore examined the expression of all annotated RTK and cytoplasmic tyrosine kinase (CTK) genes to prioritize these genes for sequencing. We performed high-throughput re-sequencing of the kinase domains of 24 TK genes (9 RTK and 15 CTK) using amplified genomic DNA from the bone marrow (tumor) and matched skin biopsy samples (“germline”) from 94 patients with de novo AML, and validated positive findings in an additional 94 AML tumor samples (14.4 million base pairs of double-stranded coverage). In addition to previously reported somatic mutations in FLT3, KIT, and JAK2 (which occurred at expected frequencies), we found novel somatic mutations in four patients in JAK1, NTRK1 and DDR1. Unexpectedly, we also identified novel non-synonymous germline sequence changes in 14 genes, including TYK2. We examined frequencies of known polymorphisms in our patients versus controls. We determined that the previously reported JAK3P132T allele is a germline variant that occurs in 19% of normal African Americans. Even when controlling for race, the TYK2G363S allele was found significantly less frequently in AML samples (12/376 alleles, 3.2%) compared to 147 normal controls (27/294 alleles, 9.2%, p=0.0013). Notably, there was loss of heterozygosity (LOH) at TYK2 in 2 patients. Additional population based studies and biologic validation will be required to define the significance of these sequence changes for AML pathogenesis. Lastly, we compared the expression of RTK and CTK genes in AML samples (n=92) to highly enriched normal human CD34+, promyelocyte, or polymorphonuclear neutrophil populations (n=5 each). We found several RTKs (FLT3, KIT, LTK) and CTKs (FYN, LCK, ITK, HCK and FGR) were tightly regulated in normal hematopoietic development but were dysregulated in many AML samples. Taken together, our data suggest that RTK or CTK mutations are not required for AML development but may be disease modifying events. Our data also suggest that germline variants and dysregulated expression of RTK and CTK genes may play significant roles AML pathogenesis.


Sign in / Sign up

Export Citation Format

Share Document