Skip to main content

Insights into the Pathobiology of Secondary AML

  • Chapter
  • First Online:
Acute Leukemias

Part of the book series: Hematologic Malignancies ((HEMATOLOGIC))

  • 1020 Accesses

Abstract

Secondary acute myeloid leukemia (s-AML) is recognized as AML arising from prior hematological disease, usually myelodysplastic syndrome, myeloproliferative neoplasm, or aplastic anemia or has myelodysplasia related changes or develops in context of prior cytotoxic or radiation therapy. Understanding the biology of s-AML is of utmost importance as this portends a high-risk disease including a potential for lower responses to chemotherapy. Some therapeutic developments in recent times have been CPX-351 as well as venetoclax-containing combinations. Of particular ongoing and forthcoming interest is the role of targeted therapy itself or in combination with other agents. Lastly, the role of allogeneic stem cell transplantation remains crucial although debated, and with improvement in treatment strategies provides opportunities to use the two in combination for best results.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

eBook
USD 16.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 119.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

Abbreviations

AHD:

Antecedent hematological disorder

AML:

Acute myeloid leukemia

CBF:

Core binding factor

CIBMTR:

Center for International Bone Marrow Transplant Research

EBMT:

European Society for Blood and Bone Marrow Transplantation

HCT:

Hematopoietic cell transplantation

JAK:

Janus kinase

MDS:

Myelodysplastic syndrome

MPN:

Myeloproliferative neoplasm

SNP:

Single-nucleotide polymorphism

TP:

Tumor protein

WHO:

World Health Organization

References

  1. Harris NL, Jaffe ES, Diebold J, et al. World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting-Airlie House, Virginia, November 1997. J Clin Oncol. 1999;17:3835–49.

    Article  CAS  PubMed  Google Scholar 

  2. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127:2391–405.

    Article  CAS  PubMed  Google Scholar 

  3. Friedberg JW, Neuberg D, Stone RM, et al. Outcome in patients with myelodysplastic syndrome after autologous bone marrow transplantation for non-Hodgkin’s lymphoma. J Clin Oncol. 1999;17:3128–35.

    Article  CAS  PubMed  Google Scholar 

  4. Koontz MZ, Horning SJ, Balise R, et al. Risk of therapy-related secondary leukemia in Hodgkin lymphoma: the Stanford University experience over three generations of clinical trials. J Clin Oncol. 2013;31:592–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Le Beau MM, Albain KS, Larson RA, et al. Clinical and cytogenetic correlations in 63 patients with therapy-related myelodysplastic syndromes and acute nonlymphocytic leukemia: further evidence for characteristic abnormalities of chromosomes no. 5 and 7. J Clin Oncol. 1986;4:325–45.

    Article  PubMed  Google Scholar 

  6. Travis LB, Holowaty EJ, Bergfeldt K, et al. Risk of leukemia after platinum-based chemotherapy for ovarian cancer. N Engl J Med. 1999;340:351–7.

    Article  CAS  PubMed  Google Scholar 

  7. Smith SM, Le Beau MM, Huo D, et al. Clinical-cytogenetic associations in 306 patients with therapy-related myelodysplasia and myeloid leukemia: the University of Chicago series. Blood. 2003;102:43–52.

    Article  CAS  PubMed  Google Scholar 

  8. Pedersen-Bjergaard J, Philip P. Balanced translocations involving chromosome bands 11q23 and 21q22 are highly characteristic of myelodysplasia and leukemia following therapy with cytostatic agents targeting at DNA-topoisomerase II. Blood. 1991;78:1147–8.

    Article  CAS  PubMed  Google Scholar 

  9. Rowley JD, Olney HJ. International workshop on the relationship of prior therapy to balanced chromosome aberrations in therapy-related myelodysplastic syndromes and acute leukemia: overview report. Genes Chromosomes Cancer. 2002;33:331–45.

    Article  PubMed  Google Scholar 

  10. Takeyama K, Seto M, Uike N, et al. Therapy-related leukemia and myelodysplastic syndrome: a large-scale Japanese study of clinical and cytogenetic features as well as prognostic factors. Int J Hematol. 2000;71:144–52.

    CAS  PubMed  Google Scholar 

  11. Ertz-Archambault N, Kosiorek H, Taylor GE, et al. Association of therapy for autoimmune disease with myelodysplastic syndromes and acute myeloid leukemia. JAMA Oncol. 2017;3:936–43.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Offman J, Opelz G, Doehler B, et al. Defective DNA mismatch repair in acute myeloid leukemia/myelodysplastic syndrome after organ transplantation. Blood. 2004;104:822–8.

    Article  CAS  PubMed  Google Scholar 

  13. Granfeldt Ostgard LS, Medeiros BC, Sengelov H, et al. Epidemiology and clinical significance of secondary and therapy-related acute myeloid leukemia: a national population-based cohort study. J Clin Oncol. 2015;33:3641–9.

    Article  PubMed  Google Scholar 

  14. Hulegardh E, Nilsson C, Lazarevic V, et al. Characterization and prognostic features of secondary acute myeloid leukemia in a population-based setting: a report from the Swedish Acute Leukemia Registry. Am J Hematol. 2015;90:208–14.

    Article  PubMed  Google Scholar 

  15. Boddu P, Kantarjian HM, Garcia-Manero G, et al. Treated secondary acute myeloid leukemia: a distinct high-risk subset of AML with adverse prognosis. Blood Adv. 2017a;1:1312–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Kayser S, Dohner K, Krauter J, et al. The impact of therapy-related acute myeloid leukemia (AML) on outcome in 2853 adult patients with newly diagnosed AML. Blood. 2011;117:2137–45.

    Article  CAS  PubMed  Google Scholar 

  17. Christiansen DH, Andersen MK, Pedersen-Bjergaard J. Mutations with loss of heterozygosity of p53 are common in therapy-related myelodysplasia and acute myeloid leukemia after exposure to alkylating agents and significantly associated with deletion or loss of 5q, a complex karyotype, and a poor prognosis. J Clin Oncol. 2001;19:1405–13.

    Article  CAS  PubMed  Google Scholar 

  18. Leith CP, Kopecky KJ, Godwin J, et al. Acute myeloid leukemia in the elderly: assessment of multidrug resistance (MDR1) and cytogenetics distinguishes biologic subgroups with remarkably distinct responses to standard chemotherapy. A Southwest Oncology Group study. Blood. 1997;89:3323–9.

    Article  CAS  PubMed  Google Scholar 

  19. Leith CP, Kopecky KJ, Chen IM, et al. Frequency and clinical significance of the expression of the multidrug resistance proteins MDR1/P-glycoprotein, MRP1, and LRP in acute myeloid leukemia: a Southwest Oncology Group Study. Blood. 1999;94:1086–99.

    CAS  PubMed  Google Scholar 

  20. Cole SP, Bhardwaj G, Gerlach JH, et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science. 1992;258:1650–4.

    Article  CAS  PubMed  Google Scholar 

  21. Scheffer GL, Wijngaard PL, Flens MJ, et al. The drug resistance-related protein LRP is the human major vault protein. Nat Med. 1995;1:578–82.

    Article  CAS  PubMed  Google Scholar 

  22. Head DR. Revised classification of acute myeloid leukemia. Leukemia. 1996;10:1826–31.

    CAS  PubMed  Google Scholar 

  23. Gillis NK, Ball M, Zhang Q, et al. Clonal haemopoiesis and therapy-related myeloid malignancies in elderly patients: a proof-of-concept, case-control study. Lancet Oncol. 2017;18:112–21.

    Article  PubMed  Google Scholar 

  24. Takahashi K, Wang F, Kantarjian H, et al. Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: a case-control study. Lancet Oncol. 2017;18:100–11.

    Article  PubMed  Google Scholar 

  25. Schulz E, Kashofer K, Heitzer E, et al. Preexisting TP53 mutation in therapy-related acute myeloid leukemia. Ann Hematol. 2015;94:527–9.

    Article  PubMed  Google Scholar 

  26. Wong TN, Ramsingh G, Young AL, et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature. 2015;518:552–5.

    Article  CAS  PubMed  Google Scholar 

  27. Medyouf H, Mossner M, Jann JC, et al. Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit. Cell Stem Cell. 2014;14:824–37.

    Article  CAS  PubMed  Google Scholar 

  28. Lindsley RC, Mar BG, Mazzola E, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood. 2015;125:1367–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Makishima H, Yoshizato T, Yoshida K, et al. Dynamics of clonal evolution in myelodysplastic syndromes. Nat Genet. 2017;49:204–12.

    Article  CAS  PubMed  Google Scholar 

  30. Kim T, Tyndel MS, Kim HJ, et al. The clonal origins of leukemic progression of myelodysplasia. Leukemia. 2017;31:1928–35.

    Article  CAS  PubMed  Google Scholar 

  31. Shiozawa Y, Malcovati L, Galli A, et al. Gene expression and risk of leukemic transformation in myelodysplasia. Blood. 2017;130:2642–53.

    Article  CAS  PubMed  Google Scholar 

  32. Mesa RA, Li CY, Ketterling RP, Schroeder GS, Knudson RA, Tefferi A. Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases. Blood. 2005;105:973–7.

    Article  CAS  PubMed  Google Scholar 

  33. Tefferi A, Guglielmelli P, Larson DR, et al. Long-term survival and blast transformation in molecularly annotated essential thrombocythemia, polycythemia vera, and myelofibrosis. Blood. 2014;124:2507–13; quiz 615

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Abdel-Wahab O, Manshouri T, Patel J, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Res. 2010;70:447–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Green A, Beer P. Somatic mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N Engl J Med. 2010;362:369–70.

    Article  CAS  PubMed  Google Scholar 

  36. Harutyunyan A, Klampfl T, Cazzola M, Kralovics R. p53 lesions in leukemic transformation. N Engl J Med. 2011;364:488–90.

    Article  CAS  PubMed  Google Scholar 

  37. Zhang SJ, Rampal R, Manshouri T, et al. Genetic analysis of patients with leukemic transformation of myeloproliferative neoplasms shows recurrent SRSF2 mutations that are associated with adverse outcome. Blood. 2012;119:4480–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Thoennissen NH, Krug UO, Lee DH, et al. Prevalence and prognostic impact of allelic imbalances associated with leukemic transformation of Philadelphia chromosome-negative myeloproliferative neoplasms. Blood. 2010;115:2882–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Klampfl T, Harutyunyan A, Berg T, et al. Genome integrity of myeloproliferative neoplasms in chronic phase and during disease progression. Blood. 2011;118:167–76.

    Article  CAS  PubMed  Google Scholar 

  40. Rampal R, Ahn J, Abdel-Wahab O, et al. Genomic and functional analysis of leukemic transformation of myeloproliferative neoplasms. Proc Natl Acad Sci U S A. 2014;111:E5401–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. McNerney ME, Brown CD, Peterson AL, et al. The spectrum of somatic mutations in high-risk acute myeloid leukaemia with −7/del(7q). Br J Haematol. 2014;166:550–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Side LE, Curtiss NP, Teel K, et al. RAS, FLT3, and TP53 mutations in therapy-related myeloid malignancies with abnormalities of chromosomes 5 and 7. Genes Chromosomes Cancer. 2004;39:217–23.

    Article  CAS  PubMed  Google Scholar 

  43. Ok CY, Patel KP, Garcia-Manero G, et al. TP53 mutation characteristics in therapy-related myelodysplastic syndromes and acute myeloid leukemia is similar to de novo diseases. J Hematol Oncol. 2015;8:45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Schoch C, Kern W, Schnittger S, Hiddemann W, Haferlach T. Karyotype is an independent prognostic parameter in therapy-related acute myeloid leukemia (t-AML): an analysis of 93 patients with t-AML in comparison to 1091 patients with de novo AML. Leukemia. 2004;18:120–5.

    Article  CAS  PubMed  Google Scholar 

  45. Lim WS, Tardi PG, Dos Santos N, et al. Leukemia-selective uptake and cytotoxicity of CPX-351, a synergistic fixed-ratio cytarabine:daunorubicin formulation, in bone marrow xenografts. Leuk Res. 2010;34:1214–23.

    Article  CAS  PubMed  Google Scholar 

  46. Mayer LD, Harasym TO, Tardi PG, et al. Ratiometric dosing of anticancer drug combinations: controlling drug ratios after systemic administration regulates therapeutic activity in tumor-bearing mice. Mol Cancer Ther. 2006;5:1854–63.

    Article  CAS  PubMed  Google Scholar 

  47. Tardi P, Johnstone S, Harasym N, et al. In vivo maintenance of synergistic cytarabine:daunorubicin ratios greatly enhances therapeutic efficacy. Leuk Res. 2009;33:129–39.

    Article  CAS  PubMed  Google Scholar 

  48. Lancet JE, Cortes JE, Hogge DE, et al. Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood. 2014;123:3239–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia. J Clin Oncol. 2018;36:2684–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Jiang Y, Dunbar A, Gondek LP, et al. Aberrant DNA methylation is a dominant mechanism in MDS progression to AML. Blood. 2009;113:1315–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ruter B, Wijermans PW, Lubbert M. DNA methylation as a therapeutic target in hematologic disorders: recent results in older patients with myelodysplasia and acute myeloid leukemia. Int J Hematol. 2004;80:128–35.

    Article  PubMed  CAS  Google Scholar 

  52. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10:223–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Seymour JF, Dohner H, Butrym A, et al. Azacitidine improves clinical outcomes in older patients with acute myeloid leukaemia with myelodysplasia-related changes compared with conventional care regimens. BMC Cancer. 2017;17:852.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012;30:2670–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Badar T, Kantarjian HM, Ravandi F, et al. Therapeutic benefit of decitabine, a hypomethylating agent, in patients with high-risk primary myelofibrosis and myeloproliferative neoplasm in accelerated or blastic/acute myeloid leukemia phase. Leuk Res. 2015;39:950–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Thepot S, Itzykson R, Seegers V, et al. Treatment of progression of Philadelphia-negative myeloproliferative neoplasms to myelodysplastic syndrome or acute myeloid leukemia by azacitidine: a report on 54 cases on the behalf of the Groupe Francophone des Myelodysplasies (GFM). Blood. 2010;116:3735–42.

    Article  CAS  PubMed  Google Scholar 

  57. Eghtedar A, Verstovsek S, Estrov Z, et al. Phase 2 study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia. Blood. 2012;119:4614–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Rampal RK, Mascarenhas JO, Kosiorek HE, et al. Safety and efficacy of combined ruxolitinib and decitabine in accelerated and blast-phase myeloproliferative neoplasms. Blood Adv. 2018;2:3572–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 2019;133:7–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Maiti A, DiNardo CD, Cortes JE, et al. Interim analysis of phase II study of venetoclax with 10-day decitabine (DEC10-VEN) in acute myeloid leukemia and myelodysplastic syndrome. Blood. 2018;132:286.

    Article  Google Scholar 

  61. Wei AH, Strickland SA Jr, Hou JZ, et al. Venetoclax combined with low-dose cytarabine for previously untreated patients with acute myeloid leukemia: results from a phase Ib/II study. J Clin Oncol. 2019;37:1277–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Boddu PC, Kantarjian HM, Ravandi F, et al. Characteristics and outcomes of older patients with secondary acute myeloid leukemia according to treatment approach. Cancer. 2017b;123:3050–60.

    Article  CAS  PubMed  Google Scholar 

  63. Cherington C, Slack JL, Leis J, et al. Allogeneic stem cell transplantation for myeloproliferative neoplasm in blast phase. Leuk Res. 2012;36:1147–51.

    Article  PubMed  Google Scholar 

  64. Kennedy JA, Atenafu EG, Messner HA, et al. Treatment outcomes following leukemic transformation in Philadelphia-negative myeloproliferative neoplasms. Blood. 2013;121:2725–33.

    Article  CAS  PubMed  Google Scholar 

  65. Oosterveld M, Muus P, Suciu S, et al. Chemotherapy only compared to chemotherapy followed by transplantation in high risk myelodysplastic syndrome and secondary acute myeloid leukemia; two parallel studies adjusted for various prognostic factors. Leukemia. 2002;16:1615–21.

    Article  CAS  PubMed  Google Scholar 

  66. Oosterveld M, Suciu S, Verhoef G, et al. The presence of an HLA-identical sibling donor has no impact on outcome of patients with high-risk MDS or secondary AML (sAML) treated with intensive chemotherapy followed by transplantation: results of a prospective study of the EORTC, EBMT, SAKK and GIMEMA Leukemia Groups (EORTC study 06921). Leukemia. 2003;17:859–68.

    Article  CAS  PubMed  Google Scholar 

  67. Shin SH, Yahng SA, Yoon JH, et al. Survival benefits with transplantation in secondary AML evolving from myelodysplastic syndrome with hypomethylating treatment failure. Bone Marrow Transplant. 2013;48:678–83.

    Article  PubMed  Google Scholar 

  68. Yoshizato T, Nannya Y, Atsuta Y, et al. Genetic abnormalities in myelodysplasia and secondary acute myeloid leukemia: impact on outcome of stem cell transplantation. Blood. 2017;129:2347–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Litzow MR, Tarima S, Perez WS, et al. Allogeneic transplantation for therapy-related myelodysplastic syndrome and acute myeloid leukemia. Blood. 2010;115:1850–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Sengsayadeth S, Labopin M, Boumendil A, et al. Transplant outcomes for secondary acute myeloid leukemia: acute leukemia working party of the European Society for blood and bone marrow transplantation study. Biol Blood Marrow Transplant. 2018;24:1406–14.

    Article  PubMed  Google Scholar 

  71. Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A. IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms. Leukemia. 2010;24:1146–51.

    Article  CAS  PubMed  Google Scholar 

  72. Tefferi A, Lasho TL, Abdel-Wahab O, et al. IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010;24:1302–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with Ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018;378:2386–98.

    Article  CAS  PubMed  Google Scholar 

  74. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130:722–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tania Jain .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2021 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Jain, T., Rampal, R.K. (2021). Insights into the Pathobiology of Secondary AML. In: Faderl, S.H., Kantarjian, H.M., Estey, E. (eds) Acute Leukemias. Hematologic Malignancies. Springer, Cham. https://doi.org/10.1007/978-3-030-53633-6_3

Download citation

  • DOI: https://doi.org/10.1007/978-3-030-53633-6_3

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-030-53632-9

  • Online ISBN: 978-3-030-53633-6

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics