Page 76 - Haematologica3
P. 76

V.B. Pastor et al.
months to 3 years, and spontaneous remission was achieved within 1-20 months from diagnosis. It seems that the -7 clones in our patients either vanished spontaneously or were outcompeted by “fitter” UPD7q-corrected clones with a diploid copy of the wild-type SAMD9L allele. Based on these observations, a watch-and-wait strategy might be proposed for younger patients with RCC/-7 who have no additional somatic driver mutations and are clinically sta- ble. However, prolonged “watchful waiting” poses the risk of progression as witnessed in P1, who developed CMML and acquired oncogenic mutations 3.6 years after he was diagnosed with RCC.
In conclusion, our observations establish the molecular basis of a distinct subtype of familial MDS and point to the notion that MDS with chromosome 7 loss can be the sole and common manifestation of SAMD9L-related dis- ease. The negative mutational effect leads to escape and outgrowth of clones carrying -7/del(7q) with only wild- type SAMD9L allele, which might spontaneously dissa- pear or persist and provide the first step towards disease progression. Finally, this is the first description of long- term revertant mosaicism due to non-random UPD7q in SAMD9L disease, and a plausible explanation for transient monosomy 7 syndrome.
Acknowledgments
We are grateful to Sophia Hollander, Christina Jäger, Yahaira Pastor, Alexandra Fischer, Wilfried Truckenmüller (Freiburg),
Tamara Szattler (Stockholm) and Bart Przychodzien (USA) for their excellent laboratory assistance and data management, and to Dr. Anett Schmidt, Dr. Dagmar Möbius, and Dr. Elisabeth Holfeld (Cottbus) for clinical care. The authors are grateful to the Genomics Core Facility at the German Cancer Research Center/DKFZ, Heidelberg, Germany for performing whole exome sequencing and Claritas Genomics (Cambridge, MA, USA) for targeted sequencing in P4.
Funding
Deutsche Krebshilfe (Max Eder grant #109005) to MWW, BMBF (DKTK German Cancer Consortium, topic molecular diagnostics of pediatric malignancies) to CMN and MWW, BMBF (e:Med FKZ 01ZX1409B) to MB, the German Science Foundation (DFG, SFB 850, to MB and EXE306 to HB), the Research Council, Swedish Cancer Society, and Foundation for Strategic Research to YTB. MWW and ME are past trainees of EHA-ASH translational research training in hematology. MV is an EMBO Long-term Fellow (ALTF 206-2015 co-funded by the European Commission (LTFCOFUND2013, GA-2013- 609409). JCA is a Wellcome Trust Senior Research Fellow in Clinical Science (grant 098513/Z/12/Z) with support from the National Institute for Health Research Biomedical Research Centre at Great Ormond Street Hospital for Children, NHS Foundation Trust and UCL, and Great Ormond Street Children's Charity. This study was supported in part by the Excellence Initiative of the German Research Foundation (GSC- 4, Spemann Graduate School).
436
References
1. Churpek JE, Godley LA. How I diagnose and manage individuals at risk for inherited myeloid malignancies. Blood. 2016;128 (14):1800-1813.
2. Feurstein S, Drazer MW, Godley LA. Genetic predisposition to leukemia and other hematologic malignancies. Semin Oncol. 2016;43(5):598-608.
3. HahnCN,ChongCE,CarmichaelCL,etal. Heritable GATA2 mutations associated with familial myelodysplastic syndrome and acute myeloid leukemia. Nat Genet. 2011; 43(10):1012-1017.
4. Smith ML, Cavenagh JD, Lister TA, Fitzgibbon J. Mutation of CEBPA in familial acute myeloid leukemia. N Engl J Med. 2004;351(23):2403-2407.
5. Song WJ, Sullivan MG, Legare RD, et al. Haploinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia. Nat Genet. 1999;23(2):166-175.
6. Pippucci T, Savoia A, Perrotta S, et al. Mutations in the 5' UTR of ANKRD26, the ankirin repeat domain 26 gene, cause an autosomal-dominant form of inherited thrombocytopenia, THC2. Am J Hum Genet. 2011;88(1):115-120.
7. Zhang MY, Churpek JE, Keel SB, et al. Germline ETV6 mutations in familial thrombocytopenia and hematologic malig- nancy. Nat Genet. 2015;47(2):180-185.
8. Polprasert C, Schulze I, Sekeres MA, et al. Inherited and somatic defects in DDX41 in myeloid neoplasms. Cancer Cell. 2015;27 (5):658-670.
9. Wlodarski MW, Hirabayashi S, Pastor V, et
al. Prevalence, clinical characteristics, and prognosis of GATA2-related myelodysplas- tic syndromes in children and adolescents. Blood. 2016;127(11):1387-1397.
10. Pastor V, Hirabayashi S, Karow A, et al. Mutational landscape in children with myelodysplastic syndromes is distinct from adults: specific somatic drivers and novel germline variants. Leukemia. 2017;31(3): 759-762.
11. Churpek JE, Pyrtel K, Kanchi KL, et al. Genomic analysis of germ line and somatic variants in familial myelodysplasia/acute myeloid leukemia. Blood. 2015;126(22): 2484-2490.
12. Gohring G, Michalova K, Beverloo HB, et al. Complex karyotype newly defined: the strongest prognostic factor in advanced childhood myelodysplastic syndrome. Blood. 2010;116(19):3766-3769.
13. Kardos G, Baumann I, Passmore SJ, et al. Refractory anemia in childhood: a retrospec- tive analysis of 67 patients with particular reference to monosomy 7. Blood. 2003;102(6):1997-2003.
14. Mantadakis E, Shannon KM, Singer DA, et al. Transient monosomy 7 - A case series in children and review of the literature. Cancer. 1999;85(12):2655-2661.
15. Parker TM, Klaassen RJ, Johnston DL. Spontaneous remission of myelodysplastic syndrome with monosomy 7 in a young boy. Cancer Genet Cytogenet. 2008;182(2): 122-125.
16. Leung EW, Woodman RC, Roland B, Abdelhaleem M, Freedman MH, Dror Y. Transient myelodysplastic syndrome associ- ated with isochromosome 7q abnormality. Pediatr Hematol Oncol. 2003;20(7):539-545.
17. Asou H, Matsui H, Ozaki Y, et al.
Identification of a common microdeletion cluster in 7q21.3 subband among patients with myeloid leukemia and myelodysplas- tic syndrome. Biochem Biophys Res Commun. 2009;383(2):245-251.
18. Nagamachi A, Matsui H, Asou H, et al. Haploinsufficiency of SAMD9L, an endo- some fusion facilitator, causes myeloid malignancies in mice mimicking human dis- eases with monosomy 7. Cancer Cell. 2013;24(3):305-317.
19. Chen DH, Below JE, Shimamura A, et al. Ataxia-pancytopenia syndrome is caused by missense mutations in SAMD9L. Am J Hum Genet. 2016;98(6):1146-1158.
20. Tesi B, Davidsson J, Voss M, et al. Gain-of- function SAMD9L mutations cause a syn- drome of cytopenia, immunodeficiency, MDS and neurological symptoms. Blood. 2017;129(16):2266-2279.
21. Narumi S, Amano N, Ishii T, et al. SAMD9 mutations cause a novel multisystem disor- der, MIRAGE syndrome, and are associated with loss of chromosome 7. Nat Genet. 2016;48(7):792-797.
22. Buonocore F, Kuhnen P, Suntharalingham JP, et al. Somatic mutations and progressive monosomy modify SAMD9-related pheno- types in humans. J Clin Invest. 2017;127(5): 1700-1713.
23. Schwartz JR, Wang S, Ma J, et al. Germline SAMD9 mutation in siblings with mono- somy 7 and myelodysplastic syndrome. Leukemia. 2017;31(8):1827-1830.
24. Baumann I, Niemeyer CM BJ, Shannon K. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon: IARC Press; 2008, 2008:104-107.
25. Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classifi-
haematologica | 2018; 103(3)


































































































   74   75   76   77   78