scholarly journals The Genomic Landscape of Pediatric Myelodysplastic Syndromes

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 956-956 ◽  
Author(s):  
Jason R. Schwartz ◽  
Michael P. Walsh ◽  
Jing Ma ◽  
Tamara Lamprecht ◽  
Shuoguo Wang ◽  
...  

Abstract Myelodysplastic syndromes are uncommon in children (incidence of ~2 cases/million) and have a poor prognosis. Despite the wealth of knowledge about the genomic landscape of adult MDS, much less is understood about pediatric MDS, and many recurrent mutations found in adults are not common in children (Hirabayashi, Blood 2012). Furthermore, the clinical presentation, bone marrow morphology, and cytogenetic abnormalities are also different when comparing adult and pediatric MDS, suggesting disparate underlying mechanisms. Here we describe the somatic and germline genomic landscape of pediatric MDS using whole exome sequencing (WES) and RNA-sequencing. We evaluated 88 diagnostic bone marrow samples obtained from the St. Jude Children's Research Hospital Tissue Bank from patients diagnosed between 1988 and 2015. This cohort contains 34 primary MDS, including Refractory Cytopenia of Childhood/RCC (n=19) and Refractory Anemia with Excess Blasts/RAEB (n=15). For comparison, we also included 32 treatment-related (tMDS), 14 MDS/MPN (including 10 Juvenile Myelomonocytic Leukemia/JMML), and 8 cases of AML with Myelodysplasia-Related Changes/AML-MRC (including 5 cases that previously would have been classified as RAEB in transformation/RAEB-T). WES was completed for 87 tumor/normal pairs (tumor only, n=1) using the Nextera Rapid Capture Expanded Exome (Illumina). Normal comparator gDNA was obtained from flow-sorted lymphocytes and variants were classified as germline if present at greater than 30% variant allele frequency (VAF) in the lymphocyte sample; thus, bone marrow mosaicism cannot be excluded. Mean sequencing coverage for the tumor and normal samples were 102x and 105x, respectively. An average of 7.9 variants were observed per patient in the primary MDS cohort (RCC=6.3, RAEB=10.2), compared to 25.5/patient in the tMDS cohort (p=0.001). Copy number information, obtained from WES data, determined that deletions involving chromosome 7 were frequent (n=28, 32%). Approximately 50% of RCC cases had deletions involving chromosome 7 (9 of 19), compared to only 20% of RAEB cases (3 of 15). Amplification of chromosomes 8 (n=7, 8%) and 21 (n=6, 7%), and deletions of 17 (n=5, 6%) were present at low frequency. In total, we detected 43 additional copy number abnormalities (including 9 cryptic chromosome 7 abnormalities) with WES compared to standard conventional karyotyping. RAS/MAPK pathway mutations were present in 42% of the patients (49 total mutations in 37 cases, including 4 germline variants). Fourteen of the 34 primary MDS cases (41%) had at least one RAS/MAPK mutation, including 13 somatic and 2 germline variants. Mutations in RNA splicing genes (germline, n=0; somatic, n=7; 8% of cohort) were less common, unlike what is observed in adult MDS. As expected, 2 patients with JMML harbored germline variants in PTPN11 and NF1. Surprisingly, presumed germline variants were detected in RRAS and NF1 in patients with primary MDS. Germline variants in transcription factors seen in familial MDS/AML (e.g., RUNX1, CEBPA, ETV6, GATA2) were uncommon, although a germline GATA2 variant was found in a single AML-MRC case. RNA-seq using the TruSeq (Illumina) Stranded RNA protocol was performed on 70 samples with suitable RNA. Fusion transcripts were uncommon in primary MDS, while fusions involving KMT2A, NUP98, RUNX1, MECOM, and ETV6were detected in the tMDS and AML-MRC cohorts. Although many of the mutations affecting the RAS/MAPK pathway were in common genes (NRAS, PTPN11, NF1, or CBL), many other mutations were in genes less frequently reported to be mutated in myeloid neoplasms, such as BRAF, SOS1, RIT1 and RRAS. We demonstrated that the mutations in BRAF (G469A, D594N, N581I) and SOS1 (E433K, G328R, S548R) found in our cohort activate the RAS/MAPK pathway to variable levels, as measured by ERK phosphorylation. In addition, expression of the BRAFvariants conferred IL3-independence in Ba/F3 cells. In conclusion, we show that the genomic landscapes of pediatric and adult MDS are different, namely in patterns of RAS/MAPK pathway and RNA splicing gene mutations, thus supporting the notion that MDS in adults and children comprise unique biological entities. The enrichment of RAS/MAPK mutations in pediatric MDS suggests biologic overlap with JMML and may provide direction for future therapeutic options. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
1993 ◽  
Vol 81 (5) ◽  
pp. 1152-1154
Author(s):  
R Ohno ◽  
T Naoe ◽  
M Hirano ◽  
M Kobayashi ◽  
H Hirai ◽  
...  

We treated 23 patients with myelodysplastic syndromes (MDS); 2 refractory anemia (RA) with prior therapy, 11 RA with excess of blasts (RAEB), and 10 RAEB in transformation (RAEB-T), with daily oral 45 mg/m2 all-trans retinoic acid (ATRA) in a multiinstitutional prospective study. In two patients with RAEB and one with RAEB-T, a more than 1,000/microL increase of peripheral neutrophil counts was observed with some reduction of blast percentage in the bone marrow 2 to 9 weeks after the start of ATRA. However, the effect was transient and did not last for more than 5 weeks despite the continuation of ATRA therapy. In one other patient with RA, one patient with RAEB, and one patient with RAEB-T, slight increase of hemoglobin levels or reduction of blast percentage in bone marrow was noted. Toxicities attributable to ATRA were minimal and included cheilitis, xerosis, dermatitis, gastrointestinal disorders, abnormal liver function tests, and high serum triglyceridemia. Although ATRA works remarkably as a differentiation therapy in acute promyelocytic leukemia, its effect in MDS included in this study was modest. Further study of this agent alone or in combination may be warranted in less advanced stages of this disease.


Author(s):  
Eric Padron ◽  
Tariq I. Mughal ◽  
David Sallman ◽  
Alan F. List

The myelodysplastic/myeloproliferative neoplasms (MDS/MPN) are haematologically diverse stem cell malignancies sharing phenotypic features of both myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPN) that display a paradoxical bone marrow phenotype hallmarked by myeloid proliferation in the context of bone marrow dysplasia and ineffective haematopoiesis. The unfolding MDS/MPN genomic landscape has revealed numerous mutations in signalling genes, such as CBL, JAK2, NRAS, KRAS, CSF3R, and others involving the spliceosome complex. These observations suggest that comutation of genes involved in dysplasia and bone marrow failure along with those of cytokine receptor signalling may, in part, explain the dual MDS/MPN phenotype. The respective MDS/MPN diseases are identified by the type of myeloid subset which predominates in the peripheral blood. Currently there are no standard treatment recommendations for most patients with MDS/MPN. To optimize efforts to improve the management and disease outcomes, it is essential to identify meaningful clinical and biologic endpoints and standardized response criteria for clinical trials.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4831-4831
Author(s):  
Dhatri Kodali ◽  
Hector Mesa ◽  
Ajay Rawal ◽  
Pankaj Gupta

Abstract Thrombocytosis at presentation is uncommon in the myelodysplastic syndromes (MDS), and its influence on the clinical course of the disease and on prognosis is uncertain. To determine the clinical course and long-term outcomes of patients (pts) with thrombocytosis at initial diagnosis of MDS, we conducted a retrospective analysis of 503 pts diagnosed with MDS between Jan 1966 and July 2006 at the Minneapolis VA Medical Center. Original bone marrow and peripheral blood slides and reports were reviewed with a hematopathologist (H.M.) in all pts with high platelet counts (> 400 × 103/μL) and evidence of dysplasia. Clinico-pathological correlation was obtained by chart review. Patients with inadequate data, secondary causes of thrombocytosis, transient thrombocytosis, and those without evidence of dysplasia were excluded. Of 503 pts, 41 (8.2%) were found to have thrombocytosis at presentation. Their median age was 74 years. The spleen was enlarged (by imaging) in 6 pts. Peripheral blood counts (mean; range) at diagnosis were: hemoglobin (10.9 g/dL; 7.4 – 17.1), absolute neutrophils (7.9 × 103/μL; 0.8 – 30.7) and platelets (627 × 103/μL; 402 – 1231). The cases were re-classified according to the WHO classification of myeloid disorders as follows: chronic myelomonocytic leukemia-1 (CMML-1) = 17 (41%), refractory anemia with ringed sideroblasts and thrombocytosis (RARS-T) = 13 (32%) and others = 11 (27%; including 2 pts each with RA, MDS/myeloproliferative disorder-unclassified [MDS/MPD-U] and 5q- syndrome, and 1 pt each with RA with excess blasts [RAEB-1], therapy related MDS [tMDS], refractory cytopenia with multilineage dysplasia [RCMD], MDS-U and atypical chronic myeloid leukemia [Aty CML]). Bone marrow fibrosis was increased in 3 pts with CMML-1 and 1 with RARS-T, and was normal or only focally increased in all other pts. The IPSS risk category was Low in 15 pts, Int-1 in 3 pts and Int-2 in 2 pts. Cytogenetic data was not available in the other pts. Jak-2 mutation analysis was positive in 2 pts with RARS-T, negative in 1 pt each with RARS-T and MDS/MPD-U, and is pending in others. On follow-up, 2 pts with CMML-1 and 1 pt with Aty CML transformed to acute myeloid leukemia (AML) and both pts with 5q- syndrome transformed to CMML-2. Two pts with RARS-T progressed to RAEB-2 and 1 pt with CMML-1 progressed to CMML-2. One pt with CMML-1 developed marked myelofibrosis. Marked thrombocytosis required hydroxyurea treatment in 5 pts. One MDS-U pt received 5-azacytidine. Four of 41 (10%) pts experienced major bleeding events; 3 were receiving aspirin. Five pts required ongoing red cell transfusions. The median survival (MS) was 36 months in RARS-T, 60 months in CMML-1 and 27 months in others; the MS of all 41 pts was 48 months. Causes of death were AML in 3 pts, cytopenias due to MDS in 6 pts and unrelated/unknown in 21 pts. Eleven pts are currently alive. In conclusion, the majority of pts presenting with myelodysplasia and thrombocytosis fall in the MDS/MPD category of the new WHO classification (most commonly CMML or RARS-T), be older, and have low-risk features (IPSS Low or Int-1). The risks of spontaneous bleeding, transformation to AML, progression of disease or myelofibrosis are low, and the overall prognosis is relatively favorable. Platelet counts may reach levels requiring cytoreductive therapy. This study helps better understand the natural history and prognosis of this varied spectrum of MDS and overlap syndromes.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1916-1916
Author(s):  
Olga López Villar ◽  
Fermin M. Sánchez-Guijo ◽  
Juan Luis García ◽  
Jose Ramon González Porras ◽  
Eva Villarón ◽  
...  

Abstract Myelodysplastic syndromes (MDS) are a group of clonal disorders of hematopoietic stem cell (HSC). The hematopoietic microenvironment plays a major role in the physiology of the hematopoietic system, and mesenchymal stem cells (MSC) are not only the progenitors but also the key regulators of this microenviroment. Recently, some data has been published showing that these MSC could be involved in the MDS pathophysiology. Moreover, the presence of cytogenetic aberrations on these cells is controversial. The aim of the study was to characterize bone marrow derived MSC from patients with MDS using different approaches: kinetic studies, immunophenotypic analysis and genetic changes by array-based comparative genomic hybridization (array-CGH). FISH was performed with the probe of 1q31 and Q-PCR was performed with the SYBR Green technique in order to confirm array-CGH results. Patients with untreated MDS were included in the study. Median age was 72 years (range: 54–89). Diagnosis of MDS was established according to the WHO classification as follows: 5q- syndrome (n=7), refractory anemia (n=2), refractory anemia with ringed sideroblasts (n=1) and refractory anemia with excess blasts type 2 (n=3). Standard cytogenetic and FISH studies on hematopoietic cells were performed at diagnosis according to standard methods. MSC from MDS were compared to those from 12 healthy donors. MSC were obtained by plating mononuclear cells from bone marrow, and cultured and expanded following standard procedures. According to the international consensus for MSC characterization, in the third passage MSC were harvested to perform phenotypical studies and cytogenetics. To perform Array-CGH a total of 3500 genomic targets were compounded from RP-11 libraries. The PCR products after purification were arrayed onto glass slides using a BioRobot. DNA was labelled, denaturalised and hybridizated. MSC from MDS achieved confluence at a slower rate than donor-MSC [23 days (range 12–90) vs 15 days (range 11–30) p<0,01]. Also some phenotypical markers showed lower expression on patients MSC: CD105 and CD104 (p<0,05%), compared to MSC from bone marrow donors. In all MDS cases analysed MSC showed DNA genomic changes. The most frequent aberrations were 1q31q32 region gains, present in 72% of cases, and 5q31 loss in 46% of patients. Gains in 1q31 were confirmed by FISH using the probe obtained from the BAC. Loss of 17p13 occurred in 3 cases (28%), and this may be relevant since p53 is included in that region, Q-PCR was subsequently performed confirming the loss of p53 in all these cases. The changes were not observed in hematopoietic cells analysed in order to exclude somatic changes. We conclude that MSC from MDS are functionally abnormal since they show a slower growing capacity and a lower expression of adhesion molecules. In the present study it is shown for the first time that MSC from MDS show several genomic aberrations when CGH arrays are used and the data have been confirmed by FISH and Q-PCR.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4598-4598
Author(s):  
Alvaro Aguayo ◽  
Eunice Garcia-Alvarez ◽  
Erick Crespo-Solis ◽  
Deborah Martinez-Banos ◽  
Xavier Lopez-Karpovitch

Abstract Myelodysplastic syndromes (MDS) are a group of disorders characterized by ineffective hematopoiesis along with abnormalities in proliferation, differentiation and apoptosis. The overall clinical picture is peripheral cytopenias in the setting of a normocellular or hypercellular bone marrow. A growing body of evidence suggests that angiogenesis has a major role in the pathophysiology of MDS. Androgens promote hemopoiesis by increasing the levels of erithropoietin and stimulate the stem cell compartment of the bone marrow, although rarely effective as a single agent, it is often used clinically. Thalidomide (thali) is an antiangiogenic and immunomodulatory agent that has shown to be active in patients with MDS. In a recent study, prednisone (PDN) increased the tolerability and clinical efficacy of thali in patients with myeloid metaplasia and was used in the past in patients with MDS. The purpose of this study was to prospectively evaluate response to the combination of Thali-mesterolone (MRL)-PDN by using the International Working Group Criteria of Response in MDS, in a cohort of patients with transfusion-dependent MDS (TD-MDS). Protocol was approved by the Institutional Ethics Committee, was conducted according to the Helsinki declaration, and an informed consent was signed by all participants. Results: From April 2002 to February 2007, 15 patients have been consecutively treated with thali 100 mg/d, MRL 150 mg/d and prednisone starting at 50 mg/d with further, slow tapering over 3 months. Seven (46.7%) males, median age was 59 years (range 36–78) and WHO-MDS as follows: refractory cytopenia with multilineage dysplasia (RCMD) 9/15 (60%); 5q- syndrome 1/15 (6.6%), refractory anemia with excess of blasts (RAEB)-1 2/15 (13.3%), RAEB-2 (13.3%), and unclassified MDS 1/15 (6.6%). Ten patients (66.7%) had received previous therapy for MDS. Good cytogenetic (CG) risk was present in 10 pts (66.7%); 3/15 (20%) had intermediate risk CG, and 2 (13.3%) patients had no metafases to analyze at diagnosis. Intention-to-treat (ITT) analysis after a 6-months period of therapy showed 2/15 (13.3%) complete response (CR); 2/15 (13.3%) major hematologic response (MHR), and 4/15 (26.7%) a minor hematologic response. There was a decrease in the transfusion-dependence in 3/15 patients (20%); 4/15 (26.7%) patients did not decreased their transfusion-dependence, and 3 of them had progresion to acute leukemia. Adverse events where mild (grade 1–2), easily handled, and included 53% of patients with peripheral neuropathy, 40% had headhache, 33% reported frequent infections, and 1 patient had asymptomatic bradycardia that subsided with thali withdrawal. At the last follow-up, 3 patiens progressed to acute leukemia and died. Median survival of the group is 15.2 months (range 1–50.2). Two of the patients presented here where included in the ITT analysis and where part of the patients that did not responded to therapy and died from progression to acute leukemia. Conclusions: The combination of Thali-MRL-PDN is active in TD-MDS, however new options of therapy are needed to treat this subset of patients, specially in countries where lenalidomide, decitabine and 5-azacitidine are not available yet.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 50623-50623
Author(s):  
Esther Natalie Oliva ◽  
Francesco Albano ◽  
Nicola Di Renzo ◽  
Vincenzo Pavone ◽  
Stefano Molica ◽  
...  

Abstract Abstract 50623 Background: Bone marrow karyotype in myelodysplastic syndromes (MDS) is essential to define the prognosis and to guide treatment decisions, including targeted therapies. Due to the lack of an extensive national MDS registry in Italy, epidemiological data on MDS, including cytogenetics, throughout the territory is unknown. Objective: We evaluated the incidence of cytogenetic abnormalities amongst newly diagnosed MDS patients in in 2 southern Italian regions (Calabria and Puglia). Methods: A pilot project, denominated ANDROMEDA (ANalysis of cytOgenetics alteRatiOn in the MyEloDysplAstic syndromes) was developed in 17 centers to offer a service of conventional cytogenetic analysis for all consecutive patients undergoing diagnostic evaluation for cytopenia and suspected MDS between January 1 and December 31, 2011. The study conformed to the ethical standards set out in the Declaration of Helsinki and was approved by institutional review boards at each participating center. Patients were required to provide their written informed consent. Clinical characteristics of patients and bone marrow morphology, iron staining and histology were registered. Bone marrow samples were centralized for standard cytogenetic studies and fluorescence in situ hybridization to two dedicated genetics laboratories (one for each region), blind to patients' data. Results: Two hundred and thirty-five patients were evaluated and MDS diagnosis was confirmed in 220 cases (88. 3%), according to WHO criteria. The overall incidence of clonal chromosome abnormalities detected by conventional analysis was 36. 9%. Single abnormalities included +8 (13 cases, 5. 8%), del(5q) (12 cases, 5. 4%), –Y (11 cases, 5. 0%) and del(7)/-7 (4 cases, 1. 8%). Complex karyotypes were detected in 18 (8. 1%) cases. Among all cases only 10 (4. 5%) bone marrow samples were not evaluable for cytogenetic analysis. FISH revealed additional abnormalities not identified by conventional analysis only in 3 (1. 3%) out of 72 cases. Patients were classified in WHO subtypes: 39. 2% refractory cytopenia with unilineage dysplasia (RCUD), 1. 5% refractory anemia with ring sideroblasts (RARS), 32. 5% refractory cytopenia with multilineage (RCMD), 10. 8% refractory anemia with excess of blast-1 (RAEB-1), 9. 3% refractory anemia with excess of blast-2 (RAEB-2), 4. 1% MDS with deletion 5q (MDS 5q-) and 2. 6% MDS unclassifiable (MDS-U). Conclusions: These preliminary results demonstrate that the incidence of abnormal karyotype patterns and WHO subgroups in MDS patients in Southern Italy is comparable with that described in other geographical areas. It is confirmed that conventional cytogenetic analysis is a standard in the diagnostic workup of MDS of patients with a suspected myeloid malignancy in order to identify primary abnormalities and prognostic models. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2774-2774
Author(s):  
Alexandros Makis ◽  
Stefanos I Papadhimitriou ◽  
John P Panagiotou ◽  
Agapi Parcharidou ◽  
Vassilios Papadakis ◽  
...  

Abstract Abstract 2774 Poster Board II-750 Background-Aim. De novo myelodysplastic syndromes (MDS) arise in previously healthy children, however congenital physical abnormalities often accompanying the disease, strengthen the assumption that predisposing genetic lesions may contribute to the disturbance of hematopoiesis. In our series of Greek Pediatric patients, emphasis is given to the possible clinical correlation of classifiable and unclassifiable coexisting congenital disorders to the pathogenesis of MDS. Patients and Methods. Thirty children with primary MDS (mean age 7.5 y) and 10 familial cases (mean age 8.1 y) were consecutively diagnosed and treated during a period of 21 y (1988–2009). Diagnosis was based on clinical manifestations, morphology of peripheral blood, marrow aspirate and bone marrow trephine and conventional and molecular cytogenetic analysis of bone marrow cells. Patients with secondary MDS on a background of inherited, well characterized, bone marrow failure syndrome were excluded. Results. Thirty two children had refractory cytopenia (RC), 2 refractory anemia with excess of blasts (RAEB) and 6 refractory anemia with excess blasts in transformation (RAEB-T). Dysplastic features of the erythroid, myeloid and megakaryocytic lineage were detected at marrow aspirates in 77.5%, 42.5% and 72.5% of the patients, respectively, while decreased cellularity was found at bone trephine in 25/40 patients (65%). Several chromosomal aberrations were revealed in 18/40 patients: monosomy 7 in 6 children (2 RC, 1 RAEB, 3 RAEB-T), 5q- in 1 (RC), del(20)(q13) in 1 (RC), hypodiploidy in 1 (RAEB-T), inv(8)(p32q12) in 1 (RC), inv(9)(p12q13) in 1 (RC), 1qh+ in 4 (RC), trisomy 8 in 1 (RC) and complex abnormalities in 2 (RAEB-T). Treatment modalities included: Allo HCST in 13 children (9 RC, 1 RAEB, 3 RAEB-T), immunosuppression in 2 (RC), chemotherapy in 2 (RAEB-T), supportive care in 12 (10 RC, 1 RAEB, 1 RAEB-T 1) and “watch and wait” strategy in 11 (all RC). The 5-year survival for primary MDS and familial cases was 78 % and 67% respectively, while for RC cases was 90% and for RAEB/RAEB/T 15%. Physical abnormalities were identified in 16/40 patients (40%) (13/32 RC, 3/8 RAEB/RAEB-T), among whom 14 patients were characterized as having: Naxos disease 2; Velocardiofacial-like syndrome 2; Löwe syndrome 1; Congenital lymphoedema 1; Hemifacial microsomia 1; Immunodeficiency, centromeric region instability, and facial anomalies-like syndrome (ICF-like) 1; Cornelia De Lange's syndrome 1; Dandy Walker syndrome 1 and complex, unclassifiable abnormalities 6. The genes of these disorders are known to be located close to genes implicated in normal or malignant hematopoiesis or involved in chromosomal aberrations often seen in hematological malignancies (Table). Conclusions Low grade MDS prevailed among the primary or familial MDS, in our series. Physical abnormalities were common and classifiable or unclassifiable genetic disorders were identified in both low grade and advanced MDS. We hypothesize that the genes of some of these disorders could be directly or indirectly involved in hemopoiesis/oncogenesis either by being located close to genes implicated in normal or malignant hemopoiesis, or involved in chromosomal aberrations often seen in hematological malignancies. However, to clarify whether these or other underlying genetic abnormalities may be regarded as initiating or contributing events/mechanisms for MDS in childhood, requires further specific and well-planned basic and clinical studies. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 139-139 ◽  
Author(s):  
Yusuke Shiozawa ◽  
Luca Malcovati ◽  
Aiko Sato-Otsubo ◽  
Anna Gallì ◽  
Kenichi Yoshida ◽  
...  

Abstract Introduction Splicing factor (SF) mutations represent a novel class of driver mutations in myelodysplastic syndromes (MDS), where SF3B1 and SRSF2 are most frequently affected. Although abnormal RNA splicing is thought to play a central role in the pathogenic mechanism of mutated SFs, little is known about exact gene targets whose abnormal splicing is responsible for the pathogenesis of MDS. Methods We enrolled a total of 480 patients with MDS, for whom complete clinical and pathological data were available. RNA sequencing was performed for bone marrow mononuclear cells (BM/MNCs) and/or CD34+ cells from 215 MDS patients. Observed splicing junctions were compared between samples with and without each SF mutation. In SF-mutated cases, NMD could cause severe degradation of abnormal transcripts and obscure the effect of SF-mutants. To sensitively detect abnormal transcripts otherwise degraded by nonsense-mediated RNA decay (NMD), analysis was also performed on BM/MNCs from 7 patients and CD34+ bone marrow cells from 3 patients with or without inhibition of NMD by cycloheximide (CHX). Common mutations and copy number variations were also investigated using targeted sequencing. Results SF3B1 and SRSF2 mutations were associated with distinct clinical phenotypes and outcomes. SF3B1-mutated cases typically showed isolated erythroid dysplasia and high proportion of ring sideroblasts, whereas SRSF2 mutations correlated with a significantly higher incidence of myeloid and megakaryocyte dysplasia (P<.001), higher proportion of bone marrow blasts (P=.02) and lower degree of erythroid dysplasia and proportion of ring sideroblasts (P<.001). SF3B1- and SRSF2-mutated myeloid neoplasms were also associated with a significantly different outcome, SRSF2-mutated neoplasms having a significantly shorter survival (HR=5.35, P<.001). To explore the molecular basis of these distinct features in terms of splicing defects, RNA sequencing data from SF3B1-mutated (n = 68) and SRSF2-mutated (n = 39) BM/MNCs and CD34+ cells were compared with those without known SF mutations (n = 91) to detect splicing events significantly enriched in SF-mutated cells. In total 748 and 589 splicing events were enriched in SF3B1- and SRSF2-mutated samples. Among these, 203 (27%) of SF3B1-specific events were observed almost exclusively in SF3B1-mutated samples;193 (95%) were caused by misrecognition of 3' splice sites of which ~50% resulted in frameshift. In contrast, in SRSF2-mutated cases, the predominant defects were alternative exon usage, which accounted for for 80% of the abnormalities. However, the effect of mutant-SRSF2 on abnormal splicing was generally small, with 89% showing only <3× more abnormal transcripts in SRSF2-mutated. Similar results were obtained for BM/MNCs for both mutations. Splicing defects of both SF-mutations involved substantially different set of genes. Aberrant splicing enriched in SF3B1- and SRSF2-mutated samples involved 12 and 7 cancer-related genes defined by the Cancer Gene Census with no genes overlapped in between. Of special interest among these was EZH2, which showed 2 SRSF2-associated alternative exons with a premature termination codon. EZH2 transcripts having these exons are expected to be susceptible to NMD-mediated degradation. A similar defect was also detected in another component of the polycomb repressive complex 2 (PRC2), implicated in compromised function of PRC2 in SRSF2-mutated cases. On the other hand, 2 genes involved in mitochondrial heme synthesis were significantly affected in SF3B1-mutated samples. In addition, an additional gene engaged in heme synthesis, ABCB7, was identified from experiments using NMD inhibition to detect 'masked splicing'. ABCB7 is one of the causative genes for congenital sideroblastic anemia and uniformly showed reduced expression in SF3B1-mutated samples, most likely due to abnormal splicing. Conclusion SF3B1 and SRSF2 mutations have distinct impacts on clinical phenotypes and outcomes together with RNA splicing. SF3B1 mutations caused misrecognition of 3' splice sites, frequently resulting in truncated gene products and/or decreased expression due to NMD. SRSF2 mutations were characterized by modest but more widespread alterations in exon usage of genes including multiple components of PRC2. Our results provide insights into the pathogenesis of SF-mutated MDS. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5514-5514
Author(s):  
Masataka Taguchi ◽  
Tomoko Kohno ◽  
Hiroyuki Mishima ◽  
Hiroaki Taniguchi ◽  
Takeharu Kato ◽  
...  

Abstract Introduction: Myelodysplastic syndromes (MDS) are considered as a "stem cell disorders", in which hematopoietic stem cells and lineage-committed progenitor cells acquire genetic and epigenetic alterations and provide aberrant, clonal hematopoiesis, sometimes resulted in the progression to acute myeloid leukemia (Elias HK et al, Oncogene 2014). We previously reported a rare case of which the patient developed Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL) 2.5 years after being diagnosed with MDS (Kohno T et al, Br J Haematol 1996). p190 BCR-ABL1 mRNA was detected in the Ph+ALL cells. Metaphase cytogenetics showed the karyotypes: 46, XY, 20q- in MDS phase and 46, XY, t(9;22)(q34;q11), 20q- in ALL phase, indicating that MDS and Ph+ALL in this patient were of the same clonal origin. To uncover the detail of the clonal evolution, we analyzed bone marrow samples of MDS and Ph+ALL in this patient by targeted massively parallel sequencing with a panel of 154 genes including known driver genes of hematologic malignancies. Methods: Genomic DNAs (gDNAs) were extracted from the bone marrow mononuclear cells of MDS and Ph+ALL in this patient. Targeted sequencing was performed on the Illumina HiSeq2500 platform. Single nucleotide variants (SNVs) and small insertions and deletions (INDELs) were called using HaplotypeCaller of Genome Analysis Toolkit (GATK) version 3.4-46. We also attempted to detect the breakpoint of BCR-ABL1 translocation from the targeted sequencing data using the computational method, BreaKmer (Abo RP et al, Nucleic Acids Research 2015). The candidates of the mutations and structural variations were validated by amplicon-based deep sequencing and Sanger sequencing. Copy number variations were analyzed using Affymetrix CytoScan HD Array. Results: The mutations in ASXL1 and U2AF1 genes were identified in the MDS sample with variant allele frequencies (VAFs) of about 45%. At the progression of Ph+ALL, the mutations in SETBP1, SMC1A, and SLC5A8 genes were newly acquired while the ASXL1 and U2AF1 mutations were also identified with the same level of VAFs (about 50%) as the other mutations. VAFs of all of the mutations were decreased to about 20% after the chemotherapy for Ph+ALL, and then increased to about 40% at the recurrence of the disease. Furthermore, we identified the breakpoint of BCR-ABL1 translocation at intron 1 of ABL1 genes and intron 1 of BCR genes, that is the well-known cluster region, m-bcr, only among the samples of Ph+ALL. Copy number analysis confirmed that both MDS and Ph+ALL samples harbored the deletion of chromosome 20q. And the deletion of IKZF1 gene, which is frequently identified in Ph+ALL cases (Mullighan CG et al, Nature 2008), was not identified during the progression from MDS to Ph+ALL. These results demonstrated that the MDS clone harboring 20q- and ASXL1 and U2AF1 mutations acquired the mutations in SETBP1, SMC1A, and SLC5A8 genes and the p190 BCR-ABL1, resulted in the development of Ph+ALL in this patient. Conclusion: The alterations of SETBP1, SMC1A, and SLC5A8 genes are usually reported in myeloid malignancies (Makishima H et al, Nat Genet 2013, Kon A et al, Nat Genet 2013, Whitman SP et al, Blood 2008). Previous study in transgenic mouse demonstrated the distinct role of p190 BCR-ABL1 in the development of an ALL (Voncken JW et al, Blood 1995). Recapitulating this scenario, p190 BCR-ABL1 may play a critical role in the development of Ph+ALL from the MDS stem cells in this patient. This study may provide a new insight into the stem cell origin of MDS and the role of p190 BCR-ABL1 in the development of Ph+ALL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1994 ◽  
Vol 84 (2) ◽  
pp. 588-594 ◽  
Author(s):  
H Asano ◽  
H Ohashi ◽  
M Ichihara ◽  
T Kinoshita ◽  
T Murate ◽  
...  

Abstract Clonality of marrow hematopoietic progenitor cells in myelodysplastic syndromes (MDS) was analyzed by X-chromosome inactivation pattern using polymerase chain reaction (PCR). Five female patients were included in this study; two with refractory anemia (RA) and three with RA with excess blasts (RAEB). They were heterozygous for BstXI restriction fragment length polymorphisms (RFLP) of the X-chromosome-linked phosphoglycerate kinase (PGK) gene. In each patient, erythroid and nonerythroid colonies, grown in the presence of erythropoietin and granulocyte-macrophage colony-stimulating factor (GM-CSF), exhibited no remarkable difference in clonal constitution. Two patients showed only one methylation pattern, suggesting the monoclonal origin of hematopoietic progenitor cells. Colonies of two other patients exhibited predominant and minor methylation patterns in PGK gene, indicating that nonclonal progenitor cells remain a minor population. The bone marrow of one patient appeared to contain a greater proportion of nonclonal progenitors. Stem cell factor (SCF), a potent colony- stimulating factor, enhanced both erythroid and nonerythroid colony formation. However, it did not notably alter the clonal constitutions. We conclude that nonclonal hematopoietic progenitor cells can persist in a substantial number of MDS patients.


Sign in / Sign up

Export Citation Format

Share Document