Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Clinical evolution, genetic landscape and trajectories of clonal hematopoiesis in SAMD9/SAMD9L syndromes

A Publisher Correction to this article was published on 19 November 2021

This article has been updated

Abstract

Germline SAMD9 and SAMD9L mutations (SAMD9/9Lmut) predispose to myelodysplastic syndromes (MDS) with propensity for somatic rescue. In this study, we investigated a clinically annotated pediatric MDS cohort (n = 669) to define the prevalence, genetic landscape, phenotype, therapy outcome and clonal architecture of SAMD9/9L syndromes. In consecutively diagnosed MDS, germline SAMD9/9Lmut accounted for 8% and were mutually exclusive with GATA2 mutations present in 7% of the cohort. Among SAMD9/9Lmut cases, refractory cytopenia was the most prevalent MDS subtype (90%); acquired monosomy 7 was present in 38%; constitutional abnormalities were noted in 57%; and immune dysfunction was present in 28%. The clinical outcome was independent of germline mutations. In total, 67 patients had 58 distinct germline SAMD9/9Lmut clustering to protein middle regions. Despite inconclusive in silico prediction, 94% of SAMD9/9Lmut suppressed HEK293 cell growth, and mutations expressed in CD34+ cells induced overt cell death. Furthermore, we found that 61% of SAMD9/9Lmut patients underwent somatic genetic rescue (SGR) resulting in clonal hematopoiesis, of which 95% was maladaptive (monosomy 7 ± cancer mutations), and 51% had adaptive nature (revertant UPD7q, somatic SAMD9/9Lmut). Finally, bone marrow single-cell DNA sequencing revealed multiple competing SGR events in individual patients. Our findings demonstrate that SGR is common in SAMD9/9Lmut MDS and exemplify the exceptional plasticity of hematopoiesis in children.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Study cohorts, prevalence of germline SAMD9, SAMD9L and GATA2 mutations and overall survival.
Fig. 2: Mutational landscape in SAMD9 and SAMD9L genes.
Fig. 3: Assessment of SAMD9 and SAMD9L mutations.
Fig. 4: Clonal events in patients with MDS with germline SAMD9 and SAMD9L mutations.
Fig. 5: Trajectories of clonal hematopoiesis arising from germline SAMD9 and SAMD9L mutations.

Similar content being viewed by others

Data availability

All data are available in the manuscript or in the Supplementary Information. Population MAF of studied variants are from the gnomAD v2.1.1 aggregation database (https://gnomad.broadinstitute.org/). The Python-based Mosaic package for analysis of scDNA-seq is available at GitHub (https://github.com/MissionBio/mosaic). Raw sequence datasets have been deposited at the European Genome-Phenome Archive (http://www.ebi.ac.uk/ega/) hosted by the European Bioinformatics Institute under accession numbers EGAS00001005431 (targeted panel sequencing), EGAS00001005432 (WES) and EGAS00001005433 (scDNA-seq). The detailed clinical annotation of the SAMD9/9Lmut cohort is provided in Supplementary Tables 14. Source data are provided with this paper.

Change history

References

  1. Lemos de Matos, A., Liu, J., McFadden, G. & Esteves, P. J. Evolution and divergence of the mammalian SAMD9/SAMD9L gene family. BMC Evol. Biol. 13, 121 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Wu, L. et al. miR-96 induces cisplatin chemoresistance in non-small cell lung cancer cells by downregulating SAMD9. Oncol. Lett. 11, 945–952 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Topaz, O. et al. A deleterious mutation in SAMD9 causes normophosphatemic familial tumoral calcinosis. Am. J. Hum. Genet. 79, 759–764 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Liu, J. & McFadden, G. SAMD9 is an innate antiviral host factor with stress response properties that can be antagonized by poxviruses. J. Virol. 89, 1925–1931 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Nagamachi, A. et al. Haploinsufficiency of SAMD9L, an endosome fusion facilitator, causes myeloid malignancies in mice mimicking human diseases with monosomy 7. Cancer Cell 24, 305–317 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Narumi, S. et al. SAMD9 mutations cause a novel multisystem disorder, MIRAGE syndrome, and are associated with loss of chromosome 7. Nat. Genet. 48, 792–797 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Thomas 3rd, M. E. et al. Pediatric MDS and bone marrow failure-associated germline mutations in SAMD9 and SAMD9L impair multiple pathways in primary hematopoietic cells. Leukemia https://doi.org/10.1038/s41375-021-01212-6 (2021).

  8. Allenspach, E. J. et al. Germline SAMD9L truncation variants trigger global translational repression. J. Exp. Med. 218, e20201195 (2021).

  9. Liu, J., Wennier, S., Zhang, L. & McFadden, G. M062 is a host range factor essential for myxoma virus pathogenesis and functions as an antagonist of host SAMD9 in human cells. J. Virol. 85, 3270–3282 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Meng, X., Krumm, B., Li, Y., Deng, J. & Xiang, Y. Structural basis for antagonizing a host restriction factor by C7 family of poxvirus host-range proteins. Proc. Natl Acad. Sci. USA 112, 14858–14863 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nounamo, B. et al. An interaction domain in human SAMD9 is essential for myxoma virus host-range determinant M062 antagonism of host anti-viral function. Virology 503, 94–102 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Meng, X. et al. A paralogous pair of mammalian host restriction factors form a critical host barrier against poxvirus infection. PLoS Pathog. 14, e1006884 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Mekhedov, S. L., Makarova, K. S. & Koonin, E. V. The complex domain architecture of SAMD9 family proteins, predicted STAND-like NTPases, suggests new links to inflammation and apoptosis. Biol. Direct 12, 13 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Jiang, Q. et al. The Samd9L gene: transcriptional regulation and tissue-specific expression in mouse development. J. Invest. Dermatol 131, 1428–1434 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li, C. F. et al. Human sterile alpha motif domain 9, a novel gene identified as down-regulated in aggressive fibromatosis, is absent in the mouse. BMC Genomics 8, 92 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Chen, D. H. et al. Ataxia-pancytopenia syndrome is caused by missense mutations in SAMD9L. Am. J. Hum. Genet. 98, 1146–1158 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Tesi, B. et al. Gain-of-function SAMD9L mutations cause a syndrome of cytopenia, immunodeficiency, MDS and neurological symptoms. Blood 129, 2266–2279 (2017).

  18. Schwartz, J. R. et al. Germline SAMD9 mutation in siblings with monosomy 7 and myelodysplastic syndrome. Leukemia 31, 1827–1830 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Amano, N. et al. Genetic defects in pediatric-onset adrenal insufficiency in Japan. Eur. J. Endocrinol. 177, 187–194 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Jeffries, L. et al. A novel SAMD9 mutation causing MIRAGE syndrome: an expansion and review of phenotype, dysmorphology, and natural history. Am. J. Med. Genet. A 176, 415–420 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Kim, Y. M. et al. A case of an infant suspected as IMAGE syndrome who were finally diagnosed with MIRAGE syndrome by targeted Mendelian exome sequencing. BMC Med. Genet. 19, 35 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Shima, H. et al. Two patients with MIRAGE syndrome lacking haematological features: role of somatic second-site reversion SAMD9 mutations. J. Med. Genet 55, 81–85 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Shima, H. et al. MIRAGE syndrome is a rare cause of 46,XY DSD born SGA without adrenal insufficiency. PLoS ONE 13, e0206184 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Csillag, B. et al. Somatic mosaic monosomy 7 and UPD7q in a child with MIRAGE syndrome caused by a novel SAMD9 mutation. Pediatr. Blood Cancer 66, e27589 (2019).

    Article  PubMed  CAS  Google Scholar 

  25. Thunstrom, S. & Axelsson, M. Leukoencephalopathia, demyelinating peripheral neuropathy and dural ectasia explained by a not formerly described de novo mutation in the SAMD9L gene, ends 27 years of investigations—a case report. BMC Neurol. 19, 89 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Cheah, J. J. C. et al. A novel germline SAMD9L mutation in a family with ataxia-pancytopenia syndrome and pediatric acute lymphoblastic leukemia. Haematologica 104, e318–e321 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Mengen, E. & Kucukcongar Yavas, A. A rare etiology of 46, XY disorder of sex development and adrenal insufficiency: a case of MIRAGE syndrome caused by mutations in SAMD9 gene. J. Clin. Res. Pediatr. Endocrinol. 12, 206–211 (2019).

  28. Perisa, M. P. et al. A novel SAMD9 variant identified in patient with MIRAGE syndrome: further defining syndromic phenotype and review of previous cases. Pediatr. Blood Cancer 66, e27726 (2019).

    Article  PubMed  Google Scholar 

  29. Buonocore, F. et al. Somatic mutations and progressive monosomy modify SAMD9-related phenotypes in humans. J. Clin. Invest. 127, 1700–1713 (2017).

  30. Schwartz, J. R. et al. The genomic landscape of pediatric myelodysplastic syndromes. Nat. Commun. 8, 1557 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Wong, J. C. et al. Germline SAMD9 and SAMD9L mutations are associated with extensive genetic evolution and diverse hematologic outcomes. JCI Insight 3, e121086 (2018).

  32. Bluteau, O. et al. A landscape of germ line mutations in a cohort of inherited bone marrow failure patients. Blood 131, 717–732 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Pastor, V. B. et al. Constitutional SAMD9L mutations cause familial myelodysplastic syndrome and transient monosomy 7. Haematologica 103, 427–437 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. de Jesus, A. A. et al. Distinct interferon signatures and cytokine patterns define additional systemic autoinflammatory diseases. J. Clin. Invest. 130, 1669–1682 (2020).

  35. Ishiwa, S. et al. A girl with MIRAGE syndrome who developed steroid-resistant nephrotic syndrome: a case report. BMC Nephrol. 21, 340 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ahmed, I. A. et al. Outcomes of hematopoietic cell transplantation in patients with germline SAMD9/SAMD9L mutations. Biol. Blood Marrow Transpl. 25, 2186–2196 (2019).

    Article  CAS  Google Scholar 

  37. Yoshida, M. et al. Prevalence of germline GATA2 and SAMD9/9L variants in paediatric haematological disorders with monosomy 7. Br. J. Haematol. 191, 835–843 (2020).

  38. Bamshad, M. J. et al. Exome sequencing as a tool for Mendelian disease gene discovery. Nat. Rev. Genet. 12, 745–755 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Wlodarski, M. W., Collin, M. & Horwitz, M. S. GATA2 deficiency and related myeloid neoplasms. Semin. Hematol. 54, 81–86 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Revy, P., Kannengiesser, C. & Fischer, A. Somatic genetic rescue in Mendelian haematopoietic diseases. Nat. Rev. Genet. 20, 582–598 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Huisman, S. A., Redeker, E. J., Maas, S. M., Mannens, M. M. & Hennekam, R. C. High rate of mosaicism in individuals with Cornelia de Lange syndrome. J. Med. Genet. 50, 339–344 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Erickson, R. P. Somatic gene mutation and human disease other than cancer: an update. Mutat. Res. 705, 96–106 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Jordan, D. M. et al. Identification of cis-suppression of human disease mutations by comparative genomics. Nature 524, 225–229 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Leipe, D. D., Koonin, E. V. & Aravind, L. STAND, a class of P-loop NTPases including animal and plant regulators of programmed cell death: multiple, complex domain architectures, unusual phyletic patterns, and evolution by horizontal gene transfer. J. Mol. Biol. 343, 1–28 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Cordero, M. D., Alcocer-Gomez, E. & Ryffel, B. Gain of function mutation and inflammasome driven diseases in human and mouse models. J. Autoimmun. 91, 13–22 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Nagamachi, A. et al. Multiorgan failure with abnormal receptor metabolism in mice mimicking Samd9/9L syndromes. J. Clin. Invest. 131, e140147 (2021).

  47. Sarthy, J. et al. Poor outcome with hematopoietic stem cell transplantation for bone marrow failure and MDS with severe MIRAGE syndrome phenotype. Blood Adv. 2, 120–125 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Baumann, I. et al. Childhood myelodysplastic syndrome. In: WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues (International Agency for Research on Cancer, 2017).

  49. Richards, S. et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 17, 405–424 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Bailey, M. H. et al. Comprehensive characterization of cancer driver genes and mutations. Cell 173, 371–385 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ma, X. et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature 555, 371–376 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Grobner, S. N. et al. The landscape of genomic alterations across childhood cancers. Nature 555, 321–327 (2018).

    Article  PubMed  CAS  Google Scholar 

  53. Zhang, M. Y. et al. Germline ETV6 mutations in familial thrombocytopenia and hematologic malignancy. Nat. Genet. 47, 180–185 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lindsley, R. C. et al. Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation. N. Engl. J. Med. 376, 536–547 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Papaemmanuil, E. et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122, 3616–3627 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Pastor, V. et al. Mutational landscape in children with myelodysplastic syndromes is distinct from adults: specific somatic drivers and novel germline variants. Leukemia 31, 759–762 (2017).

    Article  CAS  PubMed  Google Scholar 

  57. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Ioannidis, N. M. et al. REVEL: an ensemble method for predicting the pathogenicity of rare missense variants. Am. J. Hum. Genet. 99, 877–885 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Nykamp, K. et al. Sherloc: a comprehensive refinement of the ACMG-AMP variant classification criteria. Genet. Med. 19, 1105–1117 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Shihab, H. A. et al. An integrative approach to predicting the functional effects of non-coding and coding sequence variation. Bioinformatics 31, 1536–1543 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Mather, C. A. et al. CADD score has limited clinical validity for the identification of pathogenic variants in noncoding regions in a hereditary cancer panel. Genet. Med. 18, 1269–1275 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Quang, D., Chen, Y. & Xie, X. DANN: a deep learning approach for annotating the pathogenicity of genetic variants. Bioinformatics 31, 761–763 (2015).

    Article  CAS  PubMed  Google Scholar 

  63. Ng, P. C. & Henikoff, S. SIFT: predicting amino acid changes that affect protein function. Nucleic Acids Res. 31, 3812–3814 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Adzhubei, I. A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Schwarz, J. M., Rodelsperger, C., Schuelke, M. & Seelow, D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat. Methods 7, 575–576 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. Reva, B., Antipin, Y. & Sander, C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 39, e118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kircher, M. et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 46, 310-+ (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. van der Velde, K. J. et al. GAVIN: Gene-Aware Variant INterpretation for medical sequencing. Genome Biol. 18, 6 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Liu, X., Wu, C., Li, C. & Boerwinkle, E. dbNSFP v3.0: a one-stop database of functional predictions and annotations for human nonsynonymous and splice-site SNVs. Hum. Mutat. 37, 235–241 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Gelb, B. D. et al. ClinGen’s RASopathy Expert Panel consensus methods for variant interpretation. Genet. Med. 20, 1334–1345 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Duployez, N. et al. Familial myeloid malignancies with germline TET2 mutation. Leukemia 34, 1450–1453 (2020).

    Article  PubMed  Google Scholar 

  73. Kaasinen, E. et al. Impact of constitutional TET2 haploinsufficiency on molecular and clinical phenotype in humans. Nat. Commun. 10, 1252 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Takaoka, K. et al. A germline HLTF mutation in familial MDS induces DNA damage accumulation through impaired PCNA polyubiquitination. Leukemia 33, 1773–1782 (2019).

    Article  CAS  PubMed  Google Scholar 

  75. Gohring, G. et al. Complex karyotype newly defined: the strongest prognostic factor in advanced childhood myelodysplastic syndrome. Blood 116, 3766–3769 (2010).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the Deutsche Krebshilfe Max-Eder-Nachwuchsgruppenprogramm 70109005 (to M.W.W.), ERA PerMED GATA2-HuMo German Federal Ministry of Education and Research (BMBF) 2018-123/01KU1904 (to M.W.W.), German Cancer Consortium DKTK (to M.W.W. and C.M.N.), Fritz-Thyssen Foundation 10.17.1.026MN (to M.W.W.), Deutsche Kinderkrebsstifung 2017.03 (to M.W.W.), BMBF MyPred 01GM1911A (to M.W.W., M.E., C.M.N., G.G., B.S. and C.F.), José Carreras Leukämie-Stiftung (to V.B.P.), DFG SFB1160 (to M.K.), AIRC (Associazione Italiana Ricerca sul Cancro) Special Program Metastatic disease: the key unmet need in oncology 5 per mille 2018, project code 21147 (to F.L.), Cancer Center Core Grant (CA021765, to St. Jude) and Cooperative Centers of Excellence in Hematology NIDDK U54 grant (DK106829 to Fred Hutchinson Cancer Research Center). S.S.S. is a previous recipient of a Spemann Graduate School of Biology and Medicine scholarship. We thank S. Krueger, C. Jaeger, S. Zolles, S. Hollander, M. Teller and A.-R. Kaya for excellent laboratory assistance; A. Breier, A. Fischer, W. Truckenmueller, M. Siskou-Zwecker, A. Gebert, M. Boerries and H. Busch for data management (all University of Freiburg); D. Cullins (St. Jude) for FACS services; and P. Mitra for technical support. We extend great appreciation to M. Weiss and J. Crispino (St. Jude) for valuable and constructive discussions. We also acknowledge the Hilda Biobank Freiburg and Genomics Core Facility at the German Cancer Research Center/DKFZ for specimen processing. Patient care within the EWOG-MDS consortium would not have been possible without the continuous effort of the National Reference Pathologists, National Reference Cytogeneticists, physicians, nurses and other staff of pediatric oncology units and transplant centers in all 17 participating countries (www.ewog-mds-saa.org).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

M.W.W., S.S.S. and C.M.N. designed the research. V.B.P., S.S.S., C.G., A.S., D.L., P.K.P., J.W. and R.D.-D. performed genomic studies and analyzed data. S.S.S., E.K., A.S. and C.G. performed functional studies. M.W.W., R.K.V., P.N., M.E., B.S. and C.M.N. analyzed and interpreted clinical data. P.N. performed clinical statistical analysis. M.D., J.S., F.L., R.M., M.S., B.D.M., A.C., K.K., D.T., H.H., J.B., K.J., M.U., S.P., O.P.S., O.F., S.B., V.H., I.B., S.S.-F., M.R.N., M.G.S., B.B., P.L., P.B., R.B., I.M., M.H.A., R.M., A.S., G.C., S.H., G.G., A.Y.N., M.C., M.E., C.F., B.S., C.M.N. and M.W.W. were involved in patient care, testing and data presentation. S.S.S., M.W.W. and C.M.N. wrote the manuscript. All authors contributed to the manuscript and approved its final version.

Corresponding author

Correspondence to Marcin W. Wlodarski.

Ethics declarations

Competing interests

J.B. declares personal fees, advisory board/steering committee honorarium and/or non-financial support from Novartis, Pfizer, Kite and Janssen Pharma. P.B. declares research grants from Neovii, Reimser and Medac to his affiliated institution; is on the advisory board for Novartis, Cellgene, Amgen, Medac and Servier, both for personal and on behalf of his affiliated institution; and is part of the speaker’s bureau for Miltenyi, Jazz, Reimser, Novartis and Amgen, on behalf of his affiliated institution. Also, P.B. holds patents and receives royalties from Medac. R.D.-D. is employed by Mission Bio and owns equity in Mission Bio.

Additional information

Peer review information Nature Medicine thanks Valeria Santini and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Joao Monteiro was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Constitutional abnormalities in patients with germline SAMD9/9L mutations.

Frequency of all (pie charts) and individual (bar graphs) non-hematopoietic abnormalities in n = 67 SAMD9/9Lmut patients (n = 39 SAMD9mut and n = 29 SAMD9Lmut). Detailed phenotypic findings are outlined in Supplemental Table 4.

Extended Data Fig. 2 Computational assessment of germline SAMD9/9L and GATA2 mutations.

Rank scores of GATA2, SAMD9, and SAMD9L population variants (gnomAD) or patient-specific germline mutations (MUT) was assessed using 10 in silico algorithms: REVEL, CADD, DANN, M_CAP, FATHMM, SIFT, Polyphen2 HDIV, Polyphen2 HVAR, Mutation Taster and Mutation Assessor. The Y-axis depicts the rank scores (0, benign; 1, pathogenic). Analyzed number of gnomAD variants: GATA2 n = 217, SAMD9 n = 767, SAMD9L n = 716 and patient germline mutations from this study: GATA2 n = 19, SAMD9 n = 33, SAMD9L n = 20. (a) Box plots depicting the minimum, lower quartile, median, upper quartile and maximum of each analyzed data set, p-values are calculated by comparing the medians of each data set using Kruskal-Wallis one-way analysis of variance. The significant p-values (<0.05) are bolded. (b) Density plot showing the distribution of rank scores from gnomAD and patient missense variants across GATA2, SAMD9 and SAMD9L genes.

Extended Data Fig. 3 Functional consequence and in silico discordance of SAMD9/9L mutations.

Representative Western blot showing the protein levels 24 h after transfection with plasmid expressing SAMD9 wildtype (WT), SAMD9 n = 5 patient mutations (left), and SAMD9L wildtype (WT) with SAMD9L n = 5 patient mutations (right). The mock control was HEK293 transfected with empty vector. (b) Effect of co-occurring germline and second-site (somatic) SAMD9/9Lmut on HEK293 cell proliferation when expressed either alone or in cis from single constructs. Mean ± SEM from n = 3 independent experiments performed in triplicate are presented. P-values using paired t-test are shown for each double mutant in comparison to the respective germline SAMD9/9Lmut. (c) Comparative evaluation of pathogenicity of 49 germline SAMD9/9L mutations by CADD prediction and HEK293 growth assay.

Source data

Extended Data Fig. 4 Transduction of CD34 + cells with SAMD9/9L mutant lentiviruses.

(a) RT-PCR confirming the overexpression of SAMD9 wildtype, SAMD9 E974K, SAMD9L wildtype and SAMD9L V1512M lentiviruses compared to empty control in sorted GFP positive CD34 + cells 24 h post transduction. Mean ± SEM from n = 2 independent experiments are presented. SAMD9 and SAMD9L expression data is normalized to GAPDH, and untransduced CD34 + sample is set to 1. (b) Flow cytometry plots outlining staining strategy with Annexin V-APC and DAPI in GFP positive CD34 + cells transduced with lentiviral constructs (top: SAMD9L, bottom SAMD9). Corresponding data are shown in main manuscript Fig. 3d.

Extended Data Fig. 5 Association of germline SAMD9/9L mutation allelic frequency with karyotype.

Variant allelic frequencies of germline SAMD9/9L mutations in n = 65 patients either with monosomy 7 (-7), n = 37 or normal, karyotype, n = 28. The median allelic frequency of each group is marked, and the p-value is calculated using unpaired t-test.

Extended Data Fig. 6 Loss of heterozygosity (LOH) plots for chromosome 7 and the association of -7 with somatic events.

Allelic frequency of all informative chromosome 7 variants detected by bone marrow whole exome sequencing (variant allele frequency (VAF) between >5% and <95%) plotted for all sequenced SAMD9/9Lmut cases. The karyotypes at time of sequencing are shown, including monosomy 7 (-7), normal karyotype (NK), and uniparental isodisomy of 7q (UPD7q). (b) Chromosome 7 plots for patients with UPD7q. Two types of loss of heterozygosity (LOH) detected in patient D1297 are indicated by blue (-7) and orange (-7 & UPD7q) arrows. (c) Proportion of somatic events in SAMD9/9Lmut patients. Stacked bars demonstrate the percentage of SAMD9/9Lmut patients with different somatic events (UPD7q, somatic second-site SAMD9/9Lmut and somatic cancer gene mutations) within the -7 (blue) and other than -7 karyotype (grey). Within each bar the absolute number of patients for each group is shown.

Extended Data Fig. 7 Age at diagnosis in SAMD9/9Lmut patients with UPD7q.

Patients with detected uniparental isodisomy 7q (UPD7q), n = 7, had a diagnosis (Dx) at a younger age of median 2.1, range 0.5–7.7 years) in comparison to n = 59 patients without detectable UPD7q (median 9.4, range 0.1–18.1 years). The p-value is calculated using unpaired t-test with Welch’s correction (due to unequal cohort size). Dot plots represent the mean ± SD.

Extended Data Fig. 8 Clonal architecture and chromosome 7 copy number inferred from single-cell DNA sequencing (scDNAseq).

Pattern of clonal evolution (created with BioRender.com) and visualization of genotypes of individual clones constructed from high quality single cells (allelic dropout rate <0.9) detected in 3 patients with SAMD9 (a, b) or SAMD9L (c) germline mutations. Mutational phylogeny was inferred from scDNAseq data using Tapestri Insights v2.2 and Mosaic packages. Root denotes the total number of cells analyzed for each sample and bolded circle symbolizes ancestral clone with germline SAMD9/9Lmut. Percentage and number of single cells appear within colored circles; native state hematopoiesis (grey), second-site SAMD9L mutation (blue), UPD7q (green), -7 and -7 with somatic cancer mutations (both red). Variant allele frequency (VAF) from bulk sequencing is shown for reference. Panels (a) and (c) exemplify patients with branching evolution of independent benign and malignant SGR events arising from germline SAMD9/9Lmut hematopoiesis. Panel (b) depicts the linear evolution of malignant -7 clone with SETBP1/ASXL1 mutation to acquire an additional MYB M375_I376dup mutation. Lower part of panels ac depict the genotype annotation of the observed individual clones (shown above each genotype plot). Selected variants flanking SAMD9 or SAMD9L have either wildtype (WT), heterozygous (HET) or homozygous (HOM) genotype states. Right of lower panels show normalized amplicon read distribution of informative variants from scDNAseq, with red line marking the diploid state referenced from diploid cells.

Extended Data Fig. 9 SAMD9/9Lmut patients with stable disease or remission.

Timeline depicting the clinical course in n = 10 SAMD9/9Lmut patients with a follow up longer than 1 year who had stable disease (defined as no need for transfusions or therapeutic interventions, and no infections) or natural hematopoietic improvement. All patients presented with refractory cytopenia of childhood (RCC). The upper panel (black box) shows n = 5 patients with stable disease course characterized by chronic cytopenia, normal karyotype (NK); n = 4 had SAMD9mut. Two patients underwent immunosuppressive therapy (IST). Lower panel (green box) shows cases with spontaneous hematological improvement; 2 SAMD9Lmut patients with previous monosomy 7 (-7) had normalization of karyotype and complete blood counts (CBC).

Extended Data Fig. 10 Age at diagnosis in SAMD9/9Lmut patients according to outcomes.

Patients in remission group (n = 5) were diagnosed at a younger age in comparison to stable disease (n = 6) or high-risk/progression (n = 21) groups. The p-value was calculated using unpaired t-test with Welch’s correction.. The dot plots represent the mean ± SD. Group were defined as follows: Remission - long-lasting resolution of hematologic symptoms; Stable disease - prolonged chronic cytopenia (>1 year) without the need for therapies; High-risk/progressed - advanced MDS, somatic cancer mutations, and disease progression (progression of karyotype from normal to abnormal or MDS subtype from refractory cytopenia of childhood (RCC) to MDS with excess blast (MDS-EB)).

Supplementary information

Source data

Source Data Fig. 3

Unprocessed western blots with relevant samples lanes marked within a box

Source Data Extended Data Fig. 3

Unprocessed western blots with relevant samples lanes marked within a box

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sahoo, S.S., Pastor, V.B., Goodings, C. et al. Clinical evolution, genetic landscape and trajectories of clonal hematopoiesis in SAMD9/SAMD9L syndromes. Nat Med 27, 1806–1817 (2021). https://doi.org/10.1038/s41591-021-01511-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01511-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing