Vol 52, No 6 (2021)
Review article
Published online: 2021-04-20

open access

Page views 6297
Article views/downloads 370
Get Citation

Connect on Social Media

Connect on Social Media

The impact of clonal hematopoiesis on outcomes in patients with aplastic anemia

Katarzyna Brzeźniakiewicz-Janus1, Joanna Rupa-Matysek2, Anna Hoppe2, Lidia Gil2
Acta Haematol Pol 2021;52(6):543-551.

Abstract

Over the years, not only have the T-cell mediated immune mechanisms of aplastic anemia (AA) involved in AA development started to become better understood, but there is now also a better understanding of the roles played by somatic mutations, cytogenetic abnormalities and defective telomerase functions and other genetically-related factors.

Somatic gene mutations suggestive of clonal hematopoiesis are detected in approximately one third of patients with AA. Recent studies have suggested that some of these may predict a better response to immunosuppressive therapy, whereas others indicate poorer outcomes with higher risks of clonal evolution to myelodysplastic syndrome or acute myeloid leukemia, and that therefore better results may be obtained based on allogeneic stem cell transplantation. Furthermore, recent advances in molecular techniques may be useful in differentiating aplastic anemia from hypocellular myelodysplastic syndrome and other clonal hematopoiesises of indeterminate potential. All of these are summarized in this review which includes further insights into treatment personalization based on the molecular pathogenesis of AA.

Article available in PDF format

View PDF Download PDF file

References

  1. Maciejewski JP, Selleri C, Sato T, et al. A severe and consistent deficit in marrow and circulating primitive hematopoietic cells (long-term culture-initiating cells) in acquired aplastic anemia. Blood. 1996; 88(6): 1983–1991.
  2. Young NS, Calado RT, Scheinberg P. Current concepts in the pathophysiology and treatment of aplastic anemia. Blood. 2006; 108(8): 2509–2519.
  3. Brown KE, Tisdale J, Barrett AJ, et al. Hepatitis-associated aplastic anemia. N Engl J Med. 1997; 336(15): 1059–1064.
  4. Iavorska I, Nowicki M, Grzelak A, et al. Hepatitis associated aplasic anemia. Acta Haematologica Polonica. 2019; 50(4): 199–203.
  5. de Latour RP, Risitano A, Dufour C. Severe Aplastic Anemia and PNH. In: Carreras E, Dufour C, Mohty M, Kroger N. ed. The EBMT Handbook: Hematopoietic Stem Cell Transplantation and Cellular Therapies. Springer Open, Cham 2019: 579–585.
  6. Killick SB, Bown N, Cavenagh J, et al. British Society for Standards in Haematology. Guidelines for the diagnosis and management of adult aplastic anaemia. Br J Haematol. 2016; 172(2): 187–207.
  7. Young NS. Aplastic Anemia. N Engl J Med. 2018; 379(17): 1643–1656.
  8. Incidence of aplastic anemia: the relevance of diagnostic criteria. By the International Agranulocytosis and Aplastic Anemia Study. Blood. 1987; 70(6): 1718–1721.
  9. McCahon E, Tang K, Rogers PCJ, et al. The impact of Asian descent on the incidence of acquired severe aplastic anaemia in children. Br J Haematol. 2003; 121(1): 170–172.
  10. Issaragrisil S, Kaufman DW, Anderson T, et al. An association of aplastic anaemia in Thailand with low socioeconomic status. Aplastic Anemia Study Group. Br J Haematol. 1995; 91(1): 80–84.
  11. Montané E, Ibáñez L, Vidal X, et al. Catalan Group for Study of Agranulocytosis and Aplastic Anemia. Epidemiology of aplastic anemia: a prospective multicenter study. Haematologica. 2008; 93(4): 518–523.
  12. Kojima S. Why is the incidence of aplastic anemia higher in Asia? Expert Rev Hematol. 2017; 10(4): 277–279.
  13. Socié G, Rosenfeld S, Frickhofen N, et al. Late clonal diseases of treated aplastic anemia. Semin Hematol. 2000; 37(1): 91–101.
  14. Scheinberg P, Chen J. Aplastic anemia: what have we learned from animal models and from the clinic. Semin Hematol. 2013; 50(2): 156–164.
  15. Locasciulli A, Oneto R, Bacigalupo A, et al. Severe Aplastic Anemia Working Party of the European Blood and Marrow Transplant Group. Outcome of patients with acquired aplastic anemia given first line bone marrow transplantation or immunosuppressive treatment in the last decade: a report from the European Group for Blood and Marrow Transplantation (EBMT). Haematologica. 2007; 92(1): 11–18.
  16. Chuncharunee S, Wong R, Rojnuckarin P, et al. Efficacy of rabbit antithymocyte globulin as first-line treatment of severe aplastic anemia: an Asian multicenter retrospective study. Int J Hematol. 2016; 104(4): 454–461.
  17. Li W, Fu J, Wang F, et al. Distinct overexpression of Fas ligand on T lymphocytes in aplastic anemia. Cell Mol Immunol. 2004; 1(2): 142–147.
  18. Zoumbos NC, Gascón P, Djeu JY, et al. Circulating activated suppressor T lymphocytes in aplastic anemia. N Engl J Med. 1985; 312(5): 257–265.
  19. de Bruin AM, Demirel Ö, Hooibrink B, et al. Interferon-γ impairs proliferation of hematopoietic stem cells in mice. Blood. 2013; 121(18): 3578–3585.
  20. Hosokawa K, Muranski P, Feng X, et al. Memory stem T cells in autoimmune disease: high frequency of circulating CD8+ memory stem cells in acquired aplastic anemia. J Immunol. 2016; 196(4): 1568–1578.
  21. Ismail M, Gibson FM, Gordon-Smith EC, et al. Bcl-2 and Bcl-x expression in the CD34+ cells of aplastic anaemia patients: relationship with increased apoptosis and upregulation of Fas antigen. Br J Haematol. 2001; 113(3): 706–712.
  22. Dubey S, Shukla P, Nityanand S. Expression of interferon-gamma and tumor necrosis factor-alpha in bone marrow T cells and their levels in bone marrow plasma in patients with aplastic anemia. Ann Hematol. 2005; 84(9): 572–577.
  23. Qi J, Wang TJ, Li HX, et al. Association of HLA class II (-DRB1,-DQB1,-DPB1) alleles and haplotypes on susceptibility to aplastic anemia in northern Chinese Han. Hum Immunol. 2020; 81(12): 685–691.
  24. Liu C, Li Z, Sheng W, et al. Abnormalities of quantities and functions of natural killer cells in severe aplastic anemia. Immunol Invest. 2014; 43(5): 491–503.
  25. Solomou EE, Rezvani K, Mielke S, et al. Deficient CD4+ CD25+ FOXP3+ T regulatory cells in acquired aplastic anemia. Blood. 2007; 110(5): 1603–1606.
  26. Shi J, Ge M, Lu S, et al. Intrinsic impairment of CD4(+)CD25(+) regulatory T cells in acquired aplastic anemia. Blood. 2012; 120(8): 1624–1632.
  27. Kordasti S, Marsh J, Al-Khan S, et al. Functional characterization of CD4+ T cells in aplastic anemia. Blood. 2012; 119(9): 2033–2043.
  28. Smith JNP, Kanwar VS, MacNamara KC. Hematopoietic stem cell regulation by type I and II interferons in the pathogenesis of acquired aplastic anemia. Front Immunol. 2016; 7: 330.
  29. Sieff CA. Introduction to acquired and inherited bone marrow failure. Hematol Oncol Clin North Am. 2018; 32(4): 569–580.
  30. Kirwan M, Walne AJ, Plagnol V, et al. Exome sequencing identifies autosomal-dominant SRP72 mutations associated with familial aplasia and myelodysplasia. Am J Hum Genet. 2012; 90(5): 888–892.
  31. Ballmaier M, Germeshausen M. Congenital amegakaryocytic thrombocytopenia: clinical presentation, diagnosis, and treatment. Semin Thromb Hemost. 2011; 37(6): 673–681.
  32. McReynolds LJ, Calvo KR, Holland SM. Germline GATA2 mutation and bone marrow failure. Hematol Oncol Clin North Am. 2018; 32(4): 713–728.
  33. Shen W, Kerr CM, Przychozen B, et al. Impact of germline CTC1 alterations on telomere length in acquired bone marrow failure. Br J Haematol. 2019; 185(5): 935–939.
  34. Ulirsch JC, Verboon JM, Kazerounian S, et al. The genetic landscape of diamond-blackfan anemia. Am J Hum Genet. 2018; 103(6): 930–947.
  35. Fox LC, Wood EM, Ritchie DS, et al. Diagnostic evaluation and considerations in hypocellular bone marrow failure-A focus on genomics. Int J Lab Hematol. 2020; 42(Suppl 1): 82–89.
  36. Shimamura A. Aplastic anemia and clonal evolution: germ line and somatic genetics. Hematology Am Soc Hematol Educ Program. 2016; 2016(1): 74–82.
  37. Alter BP. Inherited bone marrow failure syndromes: considerations pre- and posttransplant. Blood. 2017; 130(21): 2257–2264.
  38. Kallen ME, Dulau-Florea A, Wang W, et al. Acquired and germline predisposition to bone marrow failure: Diagnostic features and clinical implications. Semin Hematol. 2019; 56(1): 69–82.
  39. Lane AA, Odejide O, Kopp N, et al. Low frequency clonal mutations recoverable by deep sequencing in patients with aplastic anemia. Leukemia. 2013; 27(4): 968–971.
  40. Heuser M, Schlarmann C, Dobbernack V, et al. Genetic characterization of acquired aplastic anemia by targeted sequencing. Haematologica. 2014; 99(9): e165–e167.
  41. Babushok DV, Perdigones N, Perin JC, et al. Emergence of clonal hematopoiesis in the majority of patients with acquired aplastic anemia. Cancer Genet. 2015; 208(4): 115–128.
  42. 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.
  43. DeZern AE, Symons HJ, Resar LS, et al. Detection of paroxysmal nocturnal hemoglobinuria clones to exclude inherited bone marrow failure syndromes. Eur J Haematol. 2014; 92(6): 467–470.
  44. 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.
  45. Hosokawa K, Sugimori N, Katagiri T, et al. Increased glycosylphosphatidylinositol-anchored protein-deficient granulocytes define a benign subset of bone marrow failures in patients with trisomy 8. Eur J Haematol. 2015; 95(3): 230–238.
  46. Pu JJ, Mukhina G, Wang H, et al. Natural history of paroxysmal nocturnal hemoglobinuria clones in patients presenting as aplastic anemia. Eur J Haematol. 2011; 87(1): 37–45.
  47. Patel BJ, Barot SV, Kuzmanovic T, et al. Distinctive and common features of moderate aplastic anaemia. Br J Haematol. 2020; 189(5): 967–975.
  48. Negoro E, Nagata Y, Clemente MJ, et al. Origins of myelodysplastic syndromes after aplastic anemia. Blood. 2017; 130(17): 1953–1957.
  49. Ball SE, Gibson FM, Rizzo S, et al. Progressive telomere shortening in aplastic anemia. Blood. 1998; 91(10): 3582–3592.
  50. Brümmendorf TH, Maciejewski JP, Mak J, et al. Telomere length in leukocyte subpopulations of patients with aplastic anemia. Blood. 2001; 97(4): 895–900.
  51. Calado RT. Telomeres and marrow failure. Hematology Am Soc Hematol Educ Program. 2009: 338–343.
  52. Calado RT, Young NS, et al. Telomere maintenance and human bone marrow failure. Blood. 2008; 111(9): 4446–4455.
  53. Yamaguchi H, Baerlocher GM, Lansdorp PM, et al. Mutations of the human telomerase RNA gene (TERC) in aplastic anemia and myelodysplastic syndrome. Blood. 2003; 102(3): 916–918.
  54. Yamaguchi H, Calado RT, Ly H, et al. Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia. N Engl J Med. 2005; 352(14): 1413–1424.
  55. Scheinberg P, Cooper JN, Sloand EM, et al. Association of telomere length of peripheral blood leukocytes with hematopoietic relapse, malignant transformation, and survival in severe aplastic anemia. JAMA. 2010; 304(12): 1358–1364.
  56. Gadalla SM, Wang T, Dagnall C, et al. Association between donor leukocyte telomere length and survival after unrelated allogeneic hematopoietic cell transplantation for severe aplastic anemia. JAMA. 2015; 313(6): 594–602.
  57. Dumitriu B, Feng X, Townsley DM, et al. Telomere attrition and candidate gene mutations preceding monosomy 7 in aplastic anemia. Blood. 2015; 125(4): 706–709.
  58. Vulliamy T, Marrone A, Dokal I, et al. Association between aplastic anaemia and mutations in telomerase RNA. Lancet. 2002; 359(9324): 2168–2170.
  59. Winkler T, Hong SG, Decker JE, et al. Defective telomere elongation and hematopoiesis from telomerase-mutant aplastic anemia iPSCs. J Clin Invest. 2013; 123(5): 1952–1963.
  60. Kim SY, Le Rademacher J, Antin JH, et al. Myelodysplastic syndrome evolving from aplastic anemia treated with immunosuppressive therapy: efficacy of hematopoietic stem cell transplantation. Haematologica. 2014; 99(12): 1868–1875.
  61. Calado RT, Yewdell WT, Wilkerson KL, et al. Sex hormones, acting on the TERT gene, increase telomerase activity in human primary hematopoietic cells. Blood. 2009; 114(11): 2236–2243.
  62. Mortazavi Y, Chopra R, Gordon-Smith EC, et al. Clonal patterns of X-chromosome inactivation in female patients with aplastic anaemia studies using a novel reverse transcription polymerase chain reaction method. Eur J Haematol. 2000; 64(6): 385–395.
  63. Keung YK, Pettenati MJ, Cruz JM, et al. Bone marrow cytogenetic abnormalities of aplastic anemia. Am J Hematol. 2001; 66(3): 167–171, doi: 10.1002/1096-8652(200103)66:3<167::aid-ajh1040>3.0.co;2-r.
  64. Maciejewski JP, Selleri C. Evolution of clonal cytogenetic abnormalities in aplastic anemia. Leuk Lymphoma. 2004; 45(3): 433–440.
  65. Saitoh T, Saiki M, Kumagai T, et al. Spontaneous clinical and cytogenetic remission of aplastic anemia in a patient with del(13q). Cancer Genet Cytogenet. 2002; 136(2): 126–128.
  66. Ishiyama K, Karasawa M, Miyawaki S, et al. Aplastic anaemia with 13q-: a benign subset of bone marrow failure responsive to immunosuppressive therapy. Br J Haematol. 2002; 117(3): 747–750.
  67. Hosokawa K, Katagiri T, Sugimori N, et al. Favorable outcome of patients who have 13q deletion: a suggestion for revision of the WHO 'MDS-U' designation. Haematologica. 2012; 97(12): 1845–1849.
  68. Boddu PC, Kadia TM. Molecular pathogenesis of acquired aplastic anemia. Eur J Haematol. 2019; 102(2): 103–110.
  69. Schoettler ML, Nathan DG. The pathophysiology of acquired aplastic anemia: current concepts revisited. Hematol Oncol Clin North Am. 2018; 32(4): 581–594.
  70. Li Y, Wan D, Guo R, et al. Decreased bone marrow regulatory innate lymphoid cells show a distinctive miRNA profiling in aplastic anemia. Hematology. 2021; 26(1): 37–42.
  71. Lu S, Yadav AK, Qiao X. Identification of potential miRNA-mRNA interaction network in bone marrow T cells of acquired aplastic anemia. Hematology. 2020; 25(1): 168–175.
  72. Srivastava J, Chaturvedi CP, Rahman K, et al. Differential expression of miRNAs and their target genes: Exploring a new perspective of acquired aplastic anemia pathogenesis. Int J Lab Hematol. 2020; 42(5): 501–509.
  73. Bauer M, Vaxevanis C, Heimer N, et al. Expression, regulation and function of microRNA as important players in the transition of MDS to secondary AML and their cross talk to rna-binding proteins. Int J Mol Sci. 2020; 21(19): 7140.
  74. Giudice V, Banaszak LG, Gutierrez-Rodrigues F, et al. Circulating exosomal microRNAs in acquired aplastic anemia and myelodysplastic syndromes. Haematologica. 2018; 103(7): 1150–1159.
  75. Hosokawa K, Kajigaya S, Feng X, et al. A plasma microRNA signature as a biomarker for acquired aplastic anemia. Haematologica. 2017; 102(1): 69–78.
  76. Peffault de Latour R, Tabrizi R, Marcais A, et al. Nationwide survey on the use of horse antithymocyte globulins (ATGAM) in patients with acquired aplastic anemia: a report on behalf of the French Reference Center for Aplastic Anemia. Am J Hematol. 2018; 93(5): 635–642.
  77. Cesaro S. Progress and trends in pediatric hematopoietic cell transplantation in Central-East European countries. Acta Haematologica Polonica. 2020; 51(3): 119.
  78. Czyżewski K, Sedláček P, Štěrba J, et al. Progress and trends in pediatric hematopoietic cell transplantation in Central-East European countries. Acta Haematologica Polonica. 2020; 51(3): 142–150.
  79. Peffault de Latour R, Huynh L, Ivanova JI, et al. Burden of illness among patients with severe aplastic anemia who have had insufficient response to immunosuppressive therapy: a multicenter retrospective chart review study. Ann Hematol. 2020; 99(4): 743–752.
  80. Höchsmann B, Moicean A, Risitano A, et al. Supportive care in severe and very severe aplastic anemia. Bone Marrow Transplant. 2013; 48(2): 168–173.
  81. Peslak SA, Olson T, Babushok DV. Diagnosis and treatment of aplastic anemia. Curr Treat Options Oncol. 2017; 18(12): 70.
  82. Tichelli A, Peffault de Latour R, Passweg J, et al. Long-term outcome of a randomized controlled study in patients with newly diagnosed severe aplastic anemia treated with antithymocyte globuline, cyclosporine, with or without G-CSF: a Severe Aplastic Anemia Working Party Trial from the European Group of Blood and Marrow Transplantation. Haematologica. 2020; 105(5): 1223–1231.
  83. Ding SX, Chen T, Wang T, et al. The risk of clonal evolution of granulocyte colony-stimulating factor for acquired aplastic anemia: a systematic review and meta-analysis. Acta Haematol. 2018; 140(3): 141–145.
  84. Bacigalupo A, Oneto R, Schrezenmeier H, et al. First line treatment of aplastic anemia with thymoglobuline in Europe and Asia: outcome of 955 patients treated 2001–2012. Am J Hematol. 2018; 93(5): 643–648.
  85. Bacigalupo A, Bruno B, Saracco P, et al. Antilymphocyte globulin, cyclosporine, prednisolone, and granulocyte colony-stimulating factor for severe aplastic anemia: an update of the GITMO/EBMT study on 100 patients. Blood. 2000; 95(6): 1931–1934.
  86. Alexander WS, Roberts AW, Nicola NA, et al. Deficiencies in progenitor cells of multiple hematopoietic lineages and defective megakaryocytopoiesis in mice lacking the thrombopoietic receptor c-Mpl. Blood. 1996; 87(6): 2162–2170.
  87. Townsley DM, Scheinberg P, Winkler T, et al. Eltrombopag added to standard immunosuppression for aplastic anemia. N Engl J Med. 2017; 376(16): 1540–1550.
  88. Desmond R, Townsley DM, Dumitriu B, et al. Eltrombopag restores trilineage hematopoiesis in refractory severe aplastic anemia that can be sustained on discontinuation of drug. Blood. 2014; 123(12): 1818–1825.
  89. Olnes MJ, Scheinberg P, Calvo KR, et al. Eltrombopag and improved hematopoiesis in refractory aplastic anemia. N Engl J Med. 2012; 367(1): 11–19.
  90. Lengline E, Drenou B, Peterlin P, et al. Nationwide survey on the use of eltrombopag in patients with severe aplastic anemia: report on behalf of the French Reference Center for Aplastic Anemia. Blood. 2016; 128(22): 2684.
  91. McReynolds LJ, Wang Y, Thompson AS, et al. Population frequency of fanconi pathway gene variants and their association with survival after hematopoietic cell transplantation for severe aplastic anemia. Biol Blood Marrow Transplant. 2020; 26(5): 817–822.
  92. Xu LP, Wang SQ, Ma YR, et al. Who is the best haploidentical donor for acquired severe aplastic anemia? Experience from a multicenter study. J Hematol Oncol. 2019; 12(1): 87.
  93. Marsh JCW, Risitano AM, Mufti GJ. The case for upfront HLA-matched unrelated donor hematopoietic stem cell transplantation as a curative option for adult acquired severe aplastic anemia. Biol Blood Marrow Transplant. 2019; 25(9): e277–e284.
  94. Zhu Y, Gao Q, Hu J, et al. Allo-HSCT compared with immunosuppressive therapy for acquired aplastic anemia: a system review and meta-analysis. BMC Immunol. 2020; 21(1): 10.