Utility of clinical comprehensive genomic characterization for diagnostic categorization in patients presenting with hypocellular bone marrow failure syndromes

Piers Blombery, Lucy Fox, Georgina L Ryland, Ella R Thompson, Jennifer Lickiss, Michelle McBean, Satwica Yerneni, David Hughes, Anthea Greenway, Francoise Mechinaud, Erica M Wood, Graham J Lieschke, Jeff Szer, Pasquale Barbaro, John Roy, Joel Wight, Elly Lynch, Melissa Martyn, Clara Gaff, David Ritchie, Piers Blombery, Lucy Fox, Georgina L Ryland, Ella R Thompson, Jennifer Lickiss, Michelle McBean, Satwica Yerneni, David Hughes, Anthea Greenway, Francoise Mechinaud, Erica M Wood, Graham J Lieschke, Jeff Szer, Pasquale Barbaro, John Roy, Joel Wight, Elly Lynch, Melissa Martyn, Clara Gaff, David Ritchie

Abstract

Bone marrow failure (BMF) related to hypoplasia of hematopoietic elements in the bone marrow is a heterogeneous clinical entity with a broad differential diagnosis including both inherited and acquired causes. Accurate diagnostic categorization is critical to optimal patient care and detection of genomic variants in these patients may provide this important diagnostic and prognostic information. We performed real-time, accredited (ISO15189) comprehensive genomic characterization including targeted sequencing and whole exome sequencing in 115 patients with BMF syndrome (median age 24 years, range 3 months - 81 years). In patients with clinical diagnoses of inherited BMF syndromes, acquired BMF syndromes or clinically unclassifiable BMF we detected variants in 52% (12/23), 53% (25/47) and 56% (25/45) respectively. Genomic characterization resulted in a change of diagnosis in 30/115 (26%) including the identification of germline causes for 3/47 and 16/45 cases with pre-test diagnoses of acquired and clinically unclassifiable BMF respectively. The observed clinical impact of accurate diagnostic categorization included choice to perform allogeneic stem cell transplantation, disease-specific targeted treatments, identification of at-risk family members and influence of sibling allogeneic stem cell donor choice. Multiple novel pathogenic variants and copy number changes were identified in our cohort including in TERT, FANCA, RPS7 and SAMD9. Whole exome sequence analysis facilitated the identification of variants in two genes not typically associated with a primary clinical manifestation of BMF but also demonstrated reduced sensitivity for detecting low level acquired variants. In conclusion, genomic characterization can improve diagnostic categorization of patients presenting with hypoplastic BMF syndromes and should be routinely performed in this group of patients.

Figures

Figure 1.
Figure 1.
Genes involved in inherited and acquired bone marrow failure syndromes. Genes causative of an inherited bone marrow failure syndrome (IBMFS), and genes associated with a risk of transformation to hematological malignancy or response to immunosuppression in patients with acquired aplastic anemia (aAA) and hypoplastic myelodysplastic syndrome (hMDS) were assessed in this study. Four genes, CSF3R, GATA2, MPL and RUNX1, were assessed in both germline and acquired settings. Genes targeted by whole exome sequencing (WES) only (i.e., not included on targeted panel) are denoted with an asterisk. Genes KMT2D and PSTPIP1 are not represented in this figure; variants detected in these two genes were considered ‘off-target’ findings.
Figure 2.
Figure 2.
Pre- and post-test diagnosis classification of patients. Numbers of patients classified into each diagnostic group, (i) inherited bone marrow failure syndrome (IBMFS), (ii) acquired aplastic anemia (aAA) or hypoplastic MDS (hMDS) or (iii) clinically unclassifiable bone marrow failure (UBMF) as evaluated prior to genomic testing (pre-test diagnosis) and again following genomic testing (posttest diagnosis) with transitions indicated by the connecting lines (in addition three patients changed diagnosis within the aAA/hMDS category from aAA to MDS). RCC: refractory cytopenia of childhood; tMN: therapy related myeloid neoplasm.

References

    1. Swerdlow SH, Campo E, Harris NL, et al. WHO classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edn ed. Lyon, France: International Agency for Research on Cancer, 2017.
    1. Oostra AB, Nieuwint AW, Joenje H, de Winter JP. Diagnosis of fanconi anemia: chromosomal breakage analysis. Anemia. 2012;2012:238731.
    1. Lai TP, Wright WE, Shay JW. Comparison of telomere length measurement methods. Philos Trans R Soc Lond B Biol Sci. 2018; 373(1741).
    1. Ghemlas I, Li H, Zlateska B, et al. Improving diagnostic precision, care and syndrome definitions using comprehensive next-generation sequencing for the inherited bone marrow failure syndromes. J Med Genet. 2015; 52(9):575-584.
    1. Kulasekararaj AG, Jiang J, Smith AE, et al. Somatic mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic syndrome. Blood. 2014; 124(17):2698-2704.
    1. Yoshizato T, Dumitriu B, Hosokawa K, et al. Somatic mutations and clonal hematopoiesis in aplastic anemia. N Engl J Med. 2015;373(1):35-47.
    1. Bono E, McLornan D, Travaglino E, et al. Clinical, histopathological and molecular characterization of hypoplastic myelodysplastic syndrome. Leukemia. 2019; 33(10):2495-2505.
    1. Killick SB, Bown N, Cavenagh J, et al. Guidelines for the diagnosis and management of adult aplastic anaemia. Br J Haematol. 2016;172(2):187-207.
    1. Ryland GL, Jones K, Chin M, et al. Novel genomic findings in multiple myeloma identified through routine diagnostic sequencing. J Clin Pathol. 2018;71(10):895-899.
    1. McKenna A, Hanna M, Banks E, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20(9):1297-1303.
    1. DePristo MA, Banks E, Poplin R, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43(5):491-498.
    1. Van der Auwera GA, Carneiro MO, Hartl C, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics. 2013;43:11.
    1. Markham JF, Yerneni S, Ryland GL, et al. CNspector: a web-based tool for visualisation and clinical diagnosis of copy number variation from next generation sequencing. Sci Rep. 2019;9(1):6426.
    1. Richards S, Aziz N, Bale 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. 2015;17(5):405-423.
    1. Shen W, Clemente MJ, Hosono N, et al. Deep sequencing reveals stepwise mutation acquisition in paroxysmal nocturnal hemoglobinuria. J Clin Invest. 2014;124(10):4529-4538.
    1. 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.
    1. Wehr C, Grotius K, Casadei S, et al. A novel disease-causing synonymous exonic mutation in GATA2 affecting RNA splicing. Blood. 2018;132(11):1211-1215.
    1. Sebert M, Passet M, Raimbault A, et al. Clinical and molecular characteristics of DDX41-mutated patients in a large cohort of sporadic MDS/AML. Blood. 2018; 132(Suppl 1):S797.
    1. Catucci I, Osorio A, Arver B, et al. Individuals with FANCM biallelic mutations do not develop Fanconi anemia, but show risk for breast cancer, chemotherapy toxicity and may display chromosome fragility. Genet Med. 2018;20(4):452-457.
    1. Tesi B, Davidsson J, Voss M, et al. Gain-offunction SAMD9L mutations cause a syndrome of cytopenia, immunodeficiency, MDS, and neurological symptoms. Blood. 2017;129(16):2266-2279.
    1. Lewinsohn M, Brown AL, Weinel LM, et al. Novel germ line DDX41 mutations define families with a lower age of MDS/AML onset and lymphoid malignancies. Blood. 2016;127(8):1017-1023.
    1. Boztug K, Klein C. Genetic etiologies of severe congenital neutropenia. Curr Opin Pediatr. 2011;23(1):21-26.
    1. Sasada K, Yamamoto N, Masuda H, et al. Inter-observer variance and the need for standardization in the morphological classification of myelodysplastic syndrome. Leuk Res. 2018;69:54-59.
    1. Parmentier S, Schetelig J, Lorenz K, et al. Assessment of dysplastic hematopoiesis: lessons from healthy bone marrow donors. Haematologica. 2012;97(5):723-730.
    1. Willig TN, Draptchinskaia N, Dianzani I, et al. Mutations in ribosomal protein S19 gene and diamond blackfan anemia: wide variations in phenotypic expression. Blood. 1999; 94(12):4294-4306.
    1. Farrar JE, Vlachos A, Atsidaftos E, et al. Ribosomal protein gene deletions in Diamond-Blackfan anemia. Blood. 2011; 118(26):6943-6951.
    1. Ulirsch JC, Verboon JM, Kazerounian S, et al. The genetic landscape of Diamond- Blackfan anemia. Am J Hum Genet. 2018;103(6):930-947.
    1. Vulliamy T, Beswick R, Kirwan MJ, Hossain U, Walne AJ, Dokal I. Telomere length measurement can distinguish pathogenic from non-pathogenic variants in the shelterin component, TIN2. Clin Genet. 2012;81(1):76-81.
    1. Walne AJ, Vulliamy T, Beswick R, Kirwan M, Dokal I. TINF2 mutations result in very short telomeres: analysis of a large cohort of patients with dyskeratosis congenita and related bone marrow failure syndromes. Blood. 2008;112(9):3594-3600.
    1. Vaz F, Hanenberg H, Schuster B, et al. Mutation of the RAD51C gene in a Fanconi anemia–like disorder. Nat Genet. 2010;42(5):406-409.
    1. Somyajit K, Saxena S, Babu S, Mishra A, Nagaraju G. Mammalian RAD51 paralogs protect nascent DNA at stalled forks and mediate replication restart. Nucleic Acids Res. 2015;43(20):9835-9855.
    1. Somyajit K, Subramanya S, Nagaraju G. Distinct roles of FANCO/RAD51C protein in DNA damage signaling and repair: implications for Fanconi anemia and breast cancer susceptibility. J Biol Chem. 2012; 287(5):3366-3380.
    1. Savage SA, Ballew BJ, Giri N, et al. Novel FANCI mutations in Fanconi anemia with VACTERL association. Am J Med Genet A. 2016;170A(2):386- 391.
    1. Mantere T, Haanpaa M, Hanenberg H, et al. Finnish Fanconi anemia mutations and hereditary predisposition to breast and prostate cancer. Clin Genet. 2015;88(1):68-73.
    1. Dale DC, Person RE, Bolyard AA, et al. Mutations in the gene encoding neutrophil elastase in congenital and cyclic neutropenia. Blood. 2000;96(7):2317-2322.
    1. Makaryan V, Zeidler C, Bolyard AA, et al. The diversity of mutations and clinical outcomes for ELANE-associated neutropenia. Curr Opin Hematol. 2015;22(1):3-11.
    1. Smith BN, Ancliff PJ, Pizzey A, Khwaja A, Linch DC, Gale RE. Homozygous HAX1 mutations in severe congenital neutropenia patients with sporadic disease: a novel mutation in two unrelated British kindreds. Br J Haematol. 2009;144(5):762-770.
    1. Carlsson G, Elinder G, Malmgren H, et al. Compound heterozygous HAX1 mutations in a Swedish patient with severe congenital neutropenia and no neurodevelopmental abnormalities. Pediatr Blood Cancer. 2009; 53(6):1143-1146.
    1. Bellanne-Chantelot C, Schmaltz-Panneau B, Marty C, et al. Mutations in the SRP54 gene cause severe congenital neutropenia as well as Shwachman-Diamond-like syndrome. Blood. 2018;132(12):1318-1331.
    1. Carapito R, Konantz M, Paillard C, et al. Mutations in signal recognition particle SRP54 cause syndromic neutropenia with Shwachman-Diamond-like features. J Clin Invest. 2017;127(11):4090-4103.
    1. Draptchinskaia N, Gustavsson P, Andersson B, et al. The gene encoding ribosomal protein S19 is mutated in Diamond-Blackfan anaemia. Nat Genet. 1999;21(2):169-175.
    1. Cronkhite JT, Xing C, Raghu G, et al. Telomere shortening in familial and sporadic pulmonary fibrosis. Am J Respir Crit Care Med. 2008;178(7):729-737.
    1. Ameziane N, Errami A, Leveille F, et al. Genetic subtyping of Fanconi anemia by comprehensive mutation screening. Hum Mutat. 2008;29(1):159-166.
    1. Gille JJ, Floor K, Kerkhoven L, Ameziane N, Joenje H, de Winter JP. Diagnosis of Fanconi anemia: mutation analysis by multiplex ligation- dependent probe amplification and PCR-based sanger sequencing. Anemia. 2012;2012:603253.
    1. Bottega R, Nicchia E, Cappelli E, et al. Hypomorphic FANCA mutations correlate with mild mitochondrial and clinical phenotype in Fanconi anemia. Haematologica. 2018;103(3):417-426.
    1. Morgan NV, Tipping AJ, Joenje H, Mathew CG. High frequency of large intragenic deletions in the Fanconi anemia group A gene. Am J Hum Genet. 1999;65(5):1330-1341.
    1. Zheng Z, Geng J, Yao RE, et al. Molecular defects identified by whole exome sequencing in a child with Fanconi anemia. Gene. 2013;530(2):295-300.
    1. Wehr C, Grotius K, Casadei S, et al. A novel disease-causing synonymous exonic mutation in GATA2 affecting RNA splicing. Blood. 2018;132(11):1211-1215.
    1. Holzinger D, Fassl SK, de Jager W, et al. Single amino acid charge switch defines clinically distinct proline-serine-threonine phosphatase- interacting protein 1 (PSTPIP1)- associated inflammatory diseases. J Allergy Clin Immunol. 2015;136(5):1337-1345.

Source: PubMed

3
Subscribe