-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

299 Somatic Genetic and Epigenetic Architecture of Myelodysplastic Syndromes Arising from GATA2 Deficiency

Bone Marrow Failure
Program: Oral and Poster Abstracts
Type: Oral
Session: 508. Bone Marrow Failure: Attack on the Clones – Hematopoietic Stem/Progenitor Cell Defects
Sunday, December 6, 2015: 5:30 PM
Valencia A (W415A), Level 4 (Orange County Convention Center)

Victor Bengt Pastor Loyola, MSc1*, Shinsuke Hirabayashi, MD1*, Sandra Pohl, PhD2*, Emilia J Kozyra, MSc1*, Albert Catala, MD3*, Barbara De Moerloose, MD, PhD4*, Michael Dworzak, MD5*, Henrik Hasle, MD6, Riccardo Masetti, MD7*, Markus Schmugge, MD8, Owen Smith9, Jan Starý, MD10*, Marek Ussowicz, MD11*, Marry M. van den Heuvel-Eibrink, MD, PhD12,13*, Ester Mejstrikova, MD, PhD14*, Ulrich Salzer, MD15*, Michael Lübbert, MD16*, Daniel Heudobler, MD2*, David Betts, MD17*, Jose Cervera, MD18*, Gudrun Göhring, MD19*, Oskar A. Haas20, Olga Haus, MD21*, Kyra Michalova, PhD, DSc22*, Francesco Pasquali, MD23*, Joelle Tchinda, MD24*, Nadine van Roy, MD25*, Brigitte Schlegelberger, MD, Prof.26, H. Berna Beverloo, PhD27*, Peter Noellke, MSc28*, Ayami Yoshimi, MD1*, Franco Locatelli, Prof, MD, PhD29*, Brigitte Strahm, MD1*, Jaroslaw P. Maciejewski, MD, Ph.D.30, Michael Rehli, PhD31*, Charlotte M Niemeyer, Prof, MD1 and Marcin W Wlodarski, MD1*

1University Children´s Hospital Freiburg, Division of Pediatric Hematology and Oncology, University of Freiburg, Freiburg, Germany
2Department of Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
3Dep of Hematology, Hospital Sant Joan de Déu, Barcelona, Spain
4Department of Pediatrics, Ghent University, Ghent, Belgium
5Department of Pediatrics, Medical University of Vienna, St. Anna Children's Hospital and Children’s Cancer Research Institute, Vienna, Austria
6Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
7Pediatric Oncology and Hematology, University of Bologna, Bologna, Italy
8Haematology, University Children's Hospital Zurich, Zurich, Switzerland
9Department of Haematology, Our Lady's Children's Hospital, Dublin, Ireland
10Department of Pediatric Hematology and Oncology, Charles University and Univ Hospital Motol, Prague, Czech Republic
11Dep of Pediatric Hematology/ Oncology, Charles University and Univ Hospital Motol, Prague, Czech Republic
12Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
13Pediatric Oncology/Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
14CLIP-DPHO, Charles University, Hospital Motol, Prague, Czech Republic
15Department of Rheumatology and Clinical Immunology, Freiburg University Medical Center, ALU Freiburg, Freiburg, Germany
16Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg, Germany
17Paediatric Oncology and Haematology, Our Lady´s Children´s Hospital Crumlin, Dublin, Ireland
18Hematology Department, Hospital Universitari i Politecnic La Fe, Valencia, Spain
19Institute of Cell and Molecular Pathology, Hannover Medical School, Hannover, Germany
20St. Anna Children's Hospital, Childrens Cancer Research Institute, Vienna, Austria
21Department of Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
22General Teaching Hospital and 1st Faculty of Medicine, Prague, Czech Republic
23Department of Clinical and Experimental Medicine, University of Insubria, Varese, Italy
24Dep of Hematology and Oncology, University Children's Hospital Zurich, Zurich, Switzerland
25Center for Medical Genetics, Ghent University, Ghent, Belgium
26Institute of Human Genetics, Hannover Medical School, Hannover, Germany
27Dutch Childhood Oncology Group (DCOG), The Hague, Netherlands
28University Children´s Hospital Freiburg, Division of Pediatric Hematology and Oncology, University Freiburg, Freiburg, Germany
29Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
30Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
31Internal Medicine III, University Hospital, Regensburg, Germany

The emergence of GATA2 deficiency as a germline predisposition to myeloid malignancies raises questions about the nature of acquired secondary genetic and epigenetic events facilitating leukemogenesis. Previously, mutations in ASXL1 were implicated as a possible somatic driver in single cases of GATA2-related MDS. However the landscape of secondary changes had not yet been systematically examined in larger MDS cohorts, and accounting for confounding factors. In this study, we used next-generation genomic platforms to investigate targeted mutational landscape and global epigenetic profiles in patients with GATA2 deficiency.

In a large cohort of consecutively diagnosed children with MDS we had initially established that GATA2 deficiency accounts for 7% of primary MDS cases. Exploring the known association between GATA2 mutated (GATA2mut) cases and monosomy 7 (-7), the prevalence of GATA2 deficiency was very high in patients with -7 (37%), reaching its peak in adolescence (>70%). We next tested 60 GATA2-deficient patients with MDS for the presence of secondary mutations using targeted NGS for genes involved in myeloid malignancies. Somatic status was confirmed by matched analysis of fibroblasts, hair follicles or T-cells. Single hematopoietic CFU colonies were sequenced to identify subclonal patterns. For comparison, a GATA2 wildtype (GATA2-WT) cohort of 422 children and adolescents with MDS enrolled in the studies of the European Working Group of Childhood MDS were analyzed by targeted NGS. Somatic mutations were detected in 45% (27/60) of GATA2mut as compared to 19% (82/422) GATA2-WT MDS cases (p<0.0001). Recurrently mutated genes in the GATA2mut group included SETBP1, ASXL1, STAG2, RUNX1, CBL, EZH2, NRAS/KRAS, JAK3, and PTPN11. No mutations were found in TP53, BCOR/BCORL and a number of other oncogenes. Because -7 karyotype was significantly overrepresented in GATA2mut cases with somatic mutations (78%), we next focused on this cytogenetic category. Within the -7 subgroup the rate of somatic mutations was the same in GATA2mut (56%) and GATA2-WT (58%) subgroups. However, hotspot SETBP1 mutations were overrepresented in GATA2-deficient patients with -7 (50%) vs. GATA2-WT MDS cohort (22%, p<0.05). Furthermore, STAG2 mutations were found frequently in the GATA2mut group (10%, 6/60) as opposed to only 0.2% (1/422) of the total GATA2-WT cohort (p<0.0001). Next, we aimed to define the clonal hierarchy of concurrent mutations by longitudinal NGS-analysis during disease course in selected patients. Our results indicate that somatic SETBP1 lesions precede the development of ASXL1 mutations. Remarkably, this model of clonal evolution does not depend on preexisting germline GATA2 lesion, as confirmed by sequencing of single CFU colonies cultivated from the bone marrow of 3 GATA2mut and 3 GATA2-WT MDS patients. Finally, to elucidate the epigenetic effects, we compared methylation patterns using methyl-CpG-immunoprecipitation and Illumina-NGS in 25 GATA2mut to 17 GATA2-WT patients and 10 healthy controls. Based on the degree of global methylation, there were no significant alterations allowing for the discrimination of GATA2-deficient patients from the total MDS cohort, when accounted for bias arising from cytogenetic and morphologic subgroups.

In summary, somatic SETBP1 and STAG2 mutations are associated with MDS arising from GATA2 deficiency. The remaining targeted clonal landscape is essentially determined by the presence of monosomy 7. Similarly, the global epigenetic changes correlate with morphological and cytogenetic subgroups, rather than with germline GATA2 status. The prospect of potential drug targetability of mutations frequently found in children, particularly in the SETBP1 oncogene, and in histone modifiers ASXL1 and EZH2, warrants further biological studies.

Disclosures: No relevant conflicts of interest to declare.

*signifies non-member of ASH