Skip to main content
Top

01-12-2015 | Hematologic cancers | Article

Molecular therapy for acute myeloid leukaemia

Authors: Catherine C. Coombs, Martin S. Tallman, Ross L. Levine

Abstract

Acute myeloid leukaemia (AML) is a heterogeneous disease that is, in general, associated with a very poor prognosis. Multiple cytogenetic and molecular abnormalities that characterize different forms of AML have been used to better prognosticate patients and inform treatment decisions. Indeed, risk status in patients with this disease has classically been based on cytogenetic findings; however, additional molecular characteristics have been shown to inform risk assessment, including FLT3NPM1KIT, and CEBPA mutation status. Advances in sequencing technology have led to the discovery of novel somatic mutations in tissue samples from patients with AML, providing deeper insight into the mutational landscape of the disease. The majority of patients with AML (>97%) are found to have a clonal somatic abnormality on mutational profiling. Nevertheless, our understanding of the utility of mutation profiling in clinical practice remains incomplete and is continually evolving, and evidence-based approaches to application of these data are needed. In this Review, we discuss the evidence-base for integrating mutational data into treatment decisions for patients with AML, and propose novel therapeutic algorithms in the era of molecular medicine.

Nat Rev Clin Oncol 2016; 13: 305–318. doi:10.1038/nrclinonc.2015.210

Subject terms: Acute myeloid leukaemia • Cancer genetics • Cancer therapy • Outcomes research • Predictive markers

Acute myeloid leukaemia (AML) is the most-common acute leukaemia in adults, and is primarily a disease of older adults (defined in this Review as those aged ≥60 years, unless otherwise stated), with a median age at diagnosis of 67 years1, 2. The survival rates for younger adults with AML (aged <60 years) have improved, to some extent, over time, owing mostly to the development of intensive consolidation chemotherapy regimens, and improvements in supportive care and allogeneic haematopoietic-stem-cell transplantation (allo-HSCT) — the standard induction chemotherapy regimens have not changed substantially over the past 40 years3. In older patients, however, limited or no improvement in survival rates has been achieved, especially in patients aged >75 years, for whom no improvement in outcome has been demonstrated over the past three decades4.

Literature
  1. Estey, E. & Dohner, H. Acute myeloid leukaemia. Lancet 368, 1894–1907 (2006).
  2. National Cancer Institute. SEER Stat Fact Sheets: Acute Myeloid Leukemia (AML) NIH [online], (2015).
  3. Pulte, D., Gondos, A. & Brenner, H. Improvements in survival of adults diagnosed with acute myeloblastic leukemia in the early 21st century. Haematologica 93, 594–600 (2008).
  4. Thein, M. S., Ershler, W. B., Jemal, A., Yates, J. W. & Baer, M. R. Outcome of older patients with acute myeloid leukemia: an analysis of SEER data over 3 decades. Cancer 119, 2720–2727 (2013).
  5. Rockova, V. et al. Risk stratification of intermediate-risk acute myeloid leukemia: integrative analysis of a multitude of gene mutation and gene expression markers. Blood 118, 1069–1076 (2011).
  6. Dohner, K. & Paschka, P. Intermediate-risk acute myeloid leukemia therapy: current and future. Hematology Am. Soc. Hematol. Ed. Program 2014, 34–43 (2014).
  7. National Comprehensive Cancer Network. NCCN Guidelines for Treatment of Cancer by Site: Acute Myeloid Leukemia (Version 1.2015) [online], (2015).
  8. Dohner, H. et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 115, 453–474 (2010).
  9. Krug, U. et al. Complete remission and early death after intensive chemotherapy in patients aged 60 years or older with acute myeloid leukaemia: a web-based application for prediction of outcomes. Lancet 376, 2000–2008 (2010).
  10. Patel, J. P. et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 366, 1079–1089 (2012).
  11. Patel, J. P. & Levine, R. L. How do novel molecular genetic markers influence treatment decisions in acute myeloid leukemia? Hematology Am. Soc. Hematol. Ed. Program 2012, 28–34 (2012).
  12. The Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).
  13. Falini, B. et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N. Engl. J. Med. 352, 254–266 (2005).
  14. Cho, Y. U. et al. Preferential occurrence of spliceosome mutations in acute myeloid leukemia with preceding myelodysplastic syndrome and/or myelodysplasia morphology. Leuk. Lymphoma 56, 2301–2308 (2015).
  15. Aslanyan, M. G. et al. Clinical and biological impact of TET2 mutations and expression in younger adult AML patients treated within the EORTC/GIMEMA AML-12 clinical trial. Ann. Hematol. 93, 1401–1412 (2014).
  16. Gray, S. W., Hicks-Courant, K., Cronin, A., Rollins, B. J. & Weeks, J. C. Physicians' attitudes about multiplex tumor genomic testing. J. Clin. Oncol. 32, 1317–1323 (2014).
  17. Ohtake, S. et al. Randomized study of induction therapy comparing standard-dose idarubicin with high-dose daunorubicin in adult patients with previously untreated acute myeloid leukemia: the JALSG AML201 Study. Blood 117, 2358–2365 (2011).
  18. Li, X., Xu, S., Tan, Y. & Chen, J. The effects of idarubicin versus other anthracyclines for induction therapy of patients with newly diagnosed leukaemia. Cochrane Database of Systematic Reviews, Issue 6. Art. No.: CD010432 http://dx.doi.org/10.1002/ 14651858.CD010432.pub2 (2015).
  19. Fernandez, H. F. et al. Anthracycline dose intensification in acute myeloid leukemia. N. Engl. J. Med. 361, 1249–1259 (2009).
  20. Lee, J. H. et al. A randomized trial comparing standard versus high-dose daunorubicin induction in patients with acute myeloid leukemia. Blood 118, 3832–3841 (2011).
  21. Burnett, A. K. et al. A randomized comparison of daunorubicin 90mg/m2 versus 60mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients. Blood 125, 3878–3885 (2015).
  22. Slovak, M. L. et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood 96, 4075–4083 (2000).
  23. Sehgal, A. R. et al. DNMT3A mutational status affects the results of dose-escalated induction therapy in acute myelogenous leukemia. Clin. Cancer Res. 21, 1614–1620 (2015).
  24. Lo-Coco, F. et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N. Engl. J. Med. 369, 111–121 (2013).
  25. Willemze, R. et al. High-dose cytarabine in induction treatment improves the outcome of adult patients younger than age 46 years with acute myeloid leukemia: results of the EORTC-GIMEMA AML-12 trial. J. Clin. Oncol. 32, 219–228 (2014).
  26. Schlenk, R. F. et al. Phase III study of all-trans retinoic acid in previously untreated patients 61 years or older with acute myeloid leukemia. Leukemia 18, 1798–1803 (2004).
  27. Schlenk, R. F. et al. Gene mutations and response to treatment with all-trans retinoic acid in elderly patients with acute myeloid leukemia. Results from the AMLSG Trial AML HD98B. Haematologica 94, 54–60 (2009).
  28. Schlenk, R. F. et al. All-trans retinoic acid improves outcome in younger adult patients with nucleophosmin-1 mutated acute myeloid leukemia — results of the AMLSG 07–04 Randomized Treatment Trial [online], (2011).
  29. Burnett, A. K. et al. The impact on outcome of the addition of all-trans retinoic acid to intensive chemotherapy in younger patients with nonacute promyelocytic acute myeloid leukemia: overall results and results in genotypic subgroups defined by mutations in NPM1, FLT3, and CEBPA. Blood 115, 948–956 (2010).
  30. El Hajj, H. et al. Retinoic acid and arsenic trioxide trigger degradation of mutated NPM1, resulting in apoptosis of AML cells. Blood 125, 3447–3454 (2015).
  31. Martelli, M. P. et al. Arsenic trioxide and all-trans retinoic acid target NPM1 mutant oncoprotein levels and induce apoptosis in NPM1-mutated AML cells. Blood 125, 3455–3465 (2015).
  32. Tassara, M. et al. Valproic acid in combination with all-trans retinoic acid and intensive therapy for acute myeloid leukemia in older patients. 123, 4027–4036 (2014).
  33. Metzeler, K. H. et al. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia 26, 1106–1107 (2012).
  34. Itzykson, R. et al. Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias. Leukemia 25, 1147–1152 (2011).
  35. Emadi, A. et al. Presence of isocitrate dehydrogenase mutations may predict clinical response to hypomethylating agents in patients with acute myeloid leukemia. Am. J. Hematol. 90, E77–E79 (2015).
  36. Rampal, R. et al. DNA hydroxymethylation profiling reveals that WT1 mutations result in loss of TET2 function in acute myeloid leukemia. Cell Rep. 9, 1841–1855 (2014).
  37. Wang, Y. et al. WT1 recruits TET2 to regulate its target gene expression and suppress leukemia cell proliferation. Mol. Cell 57, 662–673 (2015).
  38. Quintas-Cardama, A. et al. Epigenetic therapy is associated with similar survival compared with intensive chemotherapy in older patients with newly diagnosed acute myeloid leukemia. Blood 120, 4840–4845 (2012).
  39. Renner, A. G. et al. Polo-like kinase 1 is overexpressed in acute myeloid leukemia and its inhibition preferentially targets the proliferation of leukemic cells. Blood 114, 659–662 (2009).
  40. Dohner, H. et al. Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood 124, 1426–1433 (2014).
  41. Knapper, S. The clinical development of FLT3 inhibitors in acute myeloid leukemia. Expert Opin. Investig. Drugs 20, 1377–1395 (2011).
  42. Kayser, S. & Levis, M. J. FLT3 tyrosine kinase inhibitors in acute myeloid leukemia: clinical implications and limitations. Leuk. Lymphoma 55, 243–255 (2014).
  43. Wiernik, P. H. FLT3 inhibitors for the treatment of acute myeloid leukemia. Clin. Adv. Hematol. Oncol. 8, 429–436 (2010).
  44. Wander, S. A., Levis, M. J. & Fathi, A. T. The evolving role of FLT3 inhibitors in acute myeloid leukemia: quizartinib and beyond. Ther. Adv. Hematol. 5, 65–77 (2014).
  45. Inaba, H. et al. Phase I pharmacokinetic and pharmacodynamic study of the multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in pediatric relapsed/refractory leukemia. J. Clin. Oncol. 29, 3293–3300 (2011).
  46. Serve, H. et al. Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial. J. Clin. Oncol. 31, 3110–3118 (2013).
  47. Röllig, C. et al. Sorafenib versus placebo in addition to standard therapy in younger patients with newly diagnosed acute myeloid leukemia: results from 267 patients treated in the randomized placebo-controlled SAL-soraml trial [online], (2014).
  48. Macdonald, D. A. et al. A Phase I/II study of sorafenib in combination with low dose cytarabine in elderly patients with acute myeloid leukemia or high-risk myelodysplastic syndrome from the National Cancer Institute of Canada Clinical Trials Group: trial IND.186. Leuk. Lymphoma 54, 760–766 (2013).
  49. Ravandi, F. et al. Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood 121, 4655–4662 (2013).
  50. Stone, R. M. et al. Phase IB study of the FLT3 kinase inhibitor midostaurin with chemotherapy in younger newly diagnosed adult patients with acute myeloid leukemia. Leukemia 26, 2061–2068 (2012).
  51. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  52. Knapper, S. et al. A randomised comparison of the sequential addition of the FLT3 inhibitor lestaurtinib (CEP701) to standard first line chemotherapy for FLT3-Mutated acute myeloid leukemia: the UK experience [online], (2014).
  53. Knapper, S. et al. A Phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood 108, 3262–3270 (2006).
  54. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  55. US National Library of Medicine. ClinicalTrials.gov [online], (2014).
  56. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  57. Duncavage, E. J. & Tandon, B. The utility of next-generation sequencing in diagnosis and monitoring of acute myeloid leukemia and myelodysplastic syndromes. Int. J. Lab. Hematol. 37 (Suppl. 1), 115–121 (2015).
  58. Ibanez, M. et al. Rapid screening of ASXL1, IDH1, IDH2, and c-CBL mutations in de novo acute myeloid leukemia by high-resolution melting. J. Mol. Diagn. 14, 594–601 (2012).
  59. Cheng, D. T. et al. Detection of mutations in myeloid malignancies through paired-sample analysis of microdroplet-PCR deep sequencing data. J. Mol. Diagn. 16, 504–518 (2014).
  60. Luthra, R. et al. Next-generation sequencing-based multigene mutational screening for acute myeloid leukemia using MiSeq: applicability for diagnostics and disease monitoring. Haematologica 99, 465–473 (2014).
  61. Schlenk, R. F. et al. Prospective evaluation of allogeneic hematopoietic stem-cell transplantation from matched related and matched unrelated donors in younger adults with high-risk acute myeloid leukemia: German–Austrian trial AMLHD98A. J. Clin. Oncol. 28, 4642–4648 (2010).
  62. Anderson, J. R., Cain, K. C. & Gelber, R. D. Analysis of survival by tumor response. J. Clin. Oncol. 1, 710–719 (1983).
  63. Buchner, T., Berdel, W. E. & Kienast, J. Cytogenetically normal acute myeloid leukemia. N. Engl. J. Med. 359, 651 (2008).
  64. Cornelissen, J. J. et al. Results of a HOVON/SAKK donor versus no-donor analysis of myeloablative HLA-identical sibling stem cell transplantation in first remission acute myeloid leukemia in young and middle-aged adults: benefits for whom? Blood 109, 3658–3666 (2007).
  65. Stelljes, M. et al. Allogeneic transplantation as post-remission therapy for cytogenetically high-risk acute myeloid leukemia: landmark analysis from a single prospective multicenter trial. Haematologica 96, 972–979 (2011).
  66. Estey, E. et al. Prospective feasibility analysis of reduced-intensity conditioning (RIC) regimens for hematopoietic stem cell transplantation (HSCT) in elderly patients with acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (MDS). Blood 109, 1395–1400 (2007).
  67. Koreth, J. et al. Allogeneic stem cell transplantation for acute myeloid leukemia in first complete remission: systematic review and meta-analysis of prospective clinical trials. JAMA 301, 2349–2361 (2009).
  68. Stelljes, M. et al. Allogeneic transplantation versus chemotherapy as postremission therapy for acute myeloid leukemia: a prospective matched pairs analysis. J. Clin. Oncol. 32, 288–296 (2014).
  69. Schlenk, R. F. et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N. Engl. J. Med. 358, 1909–1918 (2008).
  70. Röllig, C. et al. Allogeneic stem-cell transplantation in patients with NPM1-mutated acute myeloid leukemia: results from a prospective donor versus no-donor analysis of patients after upfront HLA typing within the SAL-AML 2003 trial. J. Clin. Oncol. 33, 403–410 (2015).
  71. Bornhauser, M. et al. Improved outcome after stem-cell transplantation in FLT3/ITD-positive AML. Blood 109, 2264–2265 (2007).
  72. Laboure, G. et al. Potent graft-versus-leukemia effect after reduced-intensity allogeneic SCT for intermediate-risk AML with FLT3-ITD or wild-type NPM1 and CEBPA without FLT3-ITD. Biol. Blood Marrow Transplant. 18, 1845–1850 (2012).
  73. Schlenk, R. F. et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood 124, 3441–3449 (2014).
  74. Brunet, S. et al. Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis. J. Clin. Oncol. 30, 735–741 (2012).
  75. Deol, A. et al. FLT3 mutation increases relapse risk after allogeneic hematopoietic cell transplant for acute myeloid leukemia in first or second complete remission: a center for international blood and marrow transplant research (CIBMTR) analysis [online], (2014).
  76. Chen, Y. B. et al. Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for patients with FLT3-ITD AML [online], (2014).
  77. Sandmaier, B. M. et al. Results of a phase 1 study of quizartinib (AC220) as maintenance therapy in subjects with acute myeloid leukemia in remission following allogeneic hematopoietic cell transplantation [online], (2014).
  78. University of Ulm. Protocol in acute myeloid leukemia with FLT3-ITD. ClinicalTrials.gov [online], (2015).
  79. Wang, Y. et al. Improved outcome with hematopoietic stem cell transplantation in a poor prognostic subgroup of patients with mixed-lineage-leukemia-rearranged acute leukemia: results from a prospective, multi-center study. Am. J. Hematol. 89, 130–136 (2014).
  80. Groschel, S. et al. Deregulated expression of EVI1 defines a poor prognostic subset of MLL-rearranged acute myeloid leukemias: a study of the German–Austrian Acute Myeloid Leukemia Study Group and the Dutch–Belgian–Swiss HOVON/SAKK Cooperative Group. J. Clin. Oncol. 31, 95–103 (2013).
  81. Gaidzik, V. I. et al. RUNX1 mutations in acute myeloid leukemia: results from a comprehensive genetic and clinical analysis from the AML study group. J. Clin. Oncol. 29, 1364–1372 (2011).
  82. Chou, S. C. et al. Prognostic implication of gene mutations on overall survival in the adult acute myeloid leukemia patients receiving or not receiving allogeneic hematopoietic stem cell transplantations. Leuk. Res. 38, 1278–1284 (2014).
  83. Rucker, F. G. et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood 119, 2114–2121 (2012).
  84. Schlenk, R. F. et al. The value of allogeneic and autologous hematopoietic stem cell transplantation in prognostically favorable acute myeloid leukemia with double mutant CEBPA. Blood 122, 1576–1582 (2013).
  85. Neubauer, A. et al. Patients with acute myeloid leukemia and RAS mutations benefit most from postremission high-dose cytarabine: a Cancer and Leukemia Group B study. J. Clin. Oncol. 26, 4603–4609 (2008).
  86. Koo, H. M. et al. Enhanced sensitivity to 1-β-D-arabinofuranosylcytosine and topoisomerase II inhibitors in tumor cell lines harboring activated ras oncogenes. Cancer Res. 56, 5211–5216 (1996).
  87. Koo, H. M., McWilliams, M. J., Alvord, W. G. & Vande Woude, G. F. Ras oncogene-induced sensitization to 1-β-D-arabinofuranosylcytosine. Cancer Res. 59, 6057–6062 (1999).
  88. Lu, C. et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483, 474–478 (2012).
  89. Wang, F. et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 340, 622–626 (2013).
  90. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  91. Stein, E. M. et al. AG-221, an oral, selective, first-in-class, potent inhibitor of the IDH2 mutant metabolic enzyme, induces durable remissions in a phase I study in patients with IDH2 mutation positive advanced hematologic malignancies [online], (2014).
  92. DiNardo, C. S. et al. AG-221, An oral, selective, first-in-class, potent inhibitor of the IDH2 mutant enzyme, induced durable responses in a phase 1 study of IDH2 mutation-positive advanced hematologic malignancies [online], (2015).
  93. de Botton, S. et al. Clinical safety and activity of AG-120, a first-in-class, potent inhibitor of the IDH1-mutant protein, in a phase 1 study of patients with advanced IDH1-mutant hematologic malignancies [online], (2015).
  94. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  95. Cortes, J. E. et al. Phase I study of quizartinib administered daily to patients with relapsed or refractory acute myeloid leukemia irrespective of FMS-like tyrosine kinase 3-internal tandem duplication status. J. Clin. Oncol. 31, 3681–3687 (2013).
  96. Levis, M. J. et al. Final results of a phase 2 open-label, monotherapy efficacy and safety study of quizartinib (AC220) in patients with FLT3-ITD positive or negative relapsed/refractory acute myeloid leukemia after second-line chemotherapy or hematopoietic stem cell transplantation [online], (2012).
  97. Galanis, A. et al. Crenolanib is a potent inhibitor of FLT3 with activity against resistance-conferring point mutants. Blood 123, 94–100 (2014).
  98. Smith, C. C. et al. Crenolanib is a selective type I pan-FLT3 inhibitor. Proc. Natl Acad. Sci. USA 111, 5319–5324 (2014).
  99. Randhawa, J. K. et al. Results of a phase II study of crenolanib in relapsed/refractory acute myeloid leukemia patients (pts) with activating FLT3 mutations [online], (2014).
  100. Brandwein, J. M. et al. A phase I/II study of imatinib plus reinduction therapy for c-kit-positive relapsed/refractory acute myeloid leukemia: inhibition of Akt activation correlates with complete response. Leukemia 25, 945–952 (2011).
  101. Chevallier, P. et al. A phase II trial of high-dose imatinib mesylate for relapsed or refractory c-kit positive and Bcr-Abl negative acute myeloid leukaemia: the AFR-15 trial. Leuk. Res. 33, 1124–1126 (2009).
  102. Advani, A. S. et al. A phase 1 study of imatinib mesylate in combination with cytarabine and daunorubicin for c-kit positive relapsed acute myeloid leukemia. Leuk. Res. 34, 1622–1626 (2010).
  103. Heidel, F. et al. Results of a multicenter Phase II trial for older patients with c-Kit-positive acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (HR-MDS) using low-dose Ara-C and Imatinib. Cancer 109, 907–914 (2007).
  104. Wardelmann, E. et al. Polyclonal evolution of multiple secondary KIT mutations in gastrointestinal stromal tumors under treatment with imatinib mesylate. Clin. Cancer Res. 12, 1743–1749 (2006).
  105. Hsueh, Y. S. et al. Autophagy is involved in endogenous and NVP-AUY922-induced KIT degradation in gastrointestinal stromal tumors. Autophagy 9, 220–233 (2013).
  106. Boissel, N. et al. Dasatinib in high-risk core binding factor acute myeloid leukemia in first complete remission: a French Acute Myeloid Leukemia Intergroup trial. Haematologica 100, 780–785 (2015).
  107. Levine, R. L. et al. The JAK2V617F activating mutation occurs in chronic myelomonocytic leukemia and acute myeloid leukemia, but not in acute lymphoblastic leukemia or chronic lymphocytic leukemia. Blood 106, 3377–3379 (2005).
  108. Eghtedar, A. et al. Phase 2 study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia. Blood 119, 4614–4618 (2012).
  109. Pemmaraju, N. et al. A phase I/II study of the Janus kinase (JAK)1 and 2 inhibitor ruxolitinib in patients with relapsed or refractory acute myeloid leukemia. Clin. Lymphoma Myeloma Leuk. 15, 171–176 (2015).
  110. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  111. Rowinsky, E. K., Windle, J. J. & Von Hoff, D. D. Ras protein farnesyltransferase: a strategic target for anticancer therapeutic development. J. Clin. Oncol. 17, 3631–3652 (1999).
  112. Lancet, J. E. et al. A phase 2 study of the farnesyltransferase inhibitor tipifarnib in poor-risk and elderly patients with previously untreated acute myelogenous leukemia. Blood 109, 1387–1394 (2007).
  113. Erba, H. P. et al. Four different regimens of farnesyltransferase inhibitor tipifarnib in older, untreated acute myeloid leukemia patients: North American Intergroup Phase II study SWOG S0432. Leukemia Res. 38, 329–333 (2014).
  114. Posch, C. et al. Combined targeting of MEK and PI3K/mTOR effector pathways is necessary to effectively inhibit NRAS mutant melanoma in vitro and in vivo. Proc. Natl Acad. Sci. USA 110, 4015–4020 (2013).
  115. Johnson, D. B., Smalley, K. S. & Sosman, J. A. Molecular pathways: targeting NRAS in melanoma and acute myelogenous leukemia. Clin. Cancer Res. 20, 4186–4192 (2014).
  116. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  117. Daigle, S. R. et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 122, 1017–1025 (2013).
  118. Daigle, S. R. et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53–65 (2011).
  119. Stein, E. M. et al. The DOT1L inhibitor EPZ-5676: safety and activity in relapsed/refractory patients with MLL-rearranged leukemia [online], (2014).
  120. Lund, K., Adams, P. D. & Copland, M. EZH2 in normal and malignant hematopoiesis. Leukemia 28, 44–49 (2014).
  121. McCabe, M. T. et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492, 108–112 (2012).
  122. Ohgami, R. S. et al. Next-generation sequencing of acute myeloid leukemia identifies the significance of TP53, U2AF1, ASXL1, and TET2 mutations. Mod. Pathol. 28, 706–714 (2015).
  123. Sinha, S. et al. Mutant WT1 is associated with DNA hypermethylation of PRC2 targets in AML and responds to EZH2 inhibition. Blood 125, 316–326 (2015).
  124. Valent, P. & Zuber, J. BRD4: a BET(ter) target for the treatment of AML? Cell Cycle 13, 689–690 (2014).
  125. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524–528 (2011).
  126. Dawson, M. A. et al. Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia. Leukemia 28, 311–320 (2014).
  127. Dombret, H. et al. A phase 1 study of the BET-bromodomain inhibitor OTX015 in patients with advanced acute leukemia [online], (2014).
  128. Grassadonia, A. et al. Role of hydroxamate-based histone deacetylase inhibitors (Hb-HDACIs) in the treatment of solid malignancies. Cancers 5, 919–942 (2013).
  129. Silva, G., Cardoso, B. A., Belo, H. & Almeida, A. M. Vorinostat induces apoptosis and differentiation in myeloid malignancies: genetic and molecular mechanisms. PLoS ONE 8, e53766 (2013).
  130. Schaefer, E. W. et al. A phase 2 study of vorinostat in acute myeloid leukemia. Haematologica 94, 1375–1382 (2009).
  131. Walter, R. B. et al. Phase II trial of vorinostat and gemtuzumab ozogamicin as induction and post-remission therapy in older adults with previously untreated acute myeloid leukemia. Haematologica 97, 739–742 (2012).
  132. U.S. Food and Drug Administration. Mylotarg (gemtuzumab ozogamicin): market withdrawal [online], (2010).
  133. Ravandi, F. et al. Gemtuzumab ozogamicin: time to resurrect? J. Clin. Oncol. 30, 3921–3923 (2012).
  134. Hills, R. K. et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 15, 986–996 (2014).
  135. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  136. Tan, P. et al. Dual epigenetic targeting with panobinostat and azacitidine in acute myeloid leukemia and high-risk myelodysplastic syndrome. Blood Cancer J. 4, e170 (2014).
  137. US National Library of Medicine. ClinicalTrials.gov [online], (2015).
  138. Novotny-Diermayr, V. et al. The oral HDAC inhibitor pracinostat (SB939) is efficacious and synergistic with the JAK2 inhibitor pacritinib (SB1518) in preclinical models of AML. Blood Cancer J. 2, e69 (2012).
  139. Bertoli, S. et al. Time from diagnosis to intensive chemotherapy initiation does not adversely impact the outcome of patients with acute myeloid leukemia. Blood 121, 2618–2626 (2013).
  140. Sekeres, M. A. et al. Time from diagnosis to treatment initiation predicts survival in younger, but not older, acute myeloid leukemia patients. Blood 113, 28–36 (2009).
  141. Welch, J. S. et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 150, 264–278 (2012).
  142. Ding, L. et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481, 506–510 (2012).
  143. Hughes, A. E. et al. Clonal architecture of secondary acute myeloid leukemia defined by single-cell sequencing. PLoS Genet. 10, e1004462 (2014).
  144. Walter, M. J. et al. Clonal architecture of secondary acute myeloid leukemia. N. Engl. J. Med. 366, 1090–1098 (2012).
  145. Steudel, C. et al. Comparative analysis of MLL partial tandem duplication and FLT3 internal tandem duplication mutations in 956 adult patients with acute myeloid leukemia. Genes Chromosomes Cancer 37, 237–251 (2003).
  146. Marcucci, G., Haferlach, T. & Dohner, H. Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications. J. Clin. Oncol. 29, 475–486 (2011).
  147. Schneider, F. et al. Age-dependent frequencies of NPM1 mutations and FLT3-ITD in patients with normal karyotype AML (NK-AML). Ann. Hematol. 91, 9–18 (2012).
  148. Ostronoff, F. et al. Prognostic significance of NPM1 mutations in the absence of FLT3-internal tandem duplication in older patients with acute myeloid leukemia: a SWOG and UK National Cancer Research Institute/Medical Research Council report. J. Clin. Oncol. 33, 1157–1164 (2015).
  149. Gaidzik, V. I. et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Blood 121, 4769–4777 (2013).
  150. Thol, F. et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. J. Clin. Oncol. 29, 2889–2896 (2011).
  151. Ribeiro, A. F. et al. Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood 119, 5824–5831 (2012).
  152. Mardis, E. R. et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 361, 1058–1066 (2009).
  153. Link, D. C. et al. Distinct patterns of mutations occurring in de novo AML versus AML arising in the setting of severe congenital neutropenia. Blood 110, 1648–1655 (2007).
  154. Carbuccia, N. et al. Mutual exclusion of ASXL1 and NPM1 mutations in a series of acute myeloid leukemias. Leukemia 24, 469–473 (2010).
  155. Schnittger, S. et al. ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome. Leukemia 27, 82–91 (2013).
  156. Paschka, P. et al. ASXL1 mutations in younger adult patients with acute myeloid leukemia: a study by the German–Austrian Acute Myeloid Leukemia Study Group. Haematologica 100, 324–330 (2015).
  157. Metzeler, K. H. et al. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN favorable genetic category. Blood 118, 6920–6929 (2011).
  158. Dufour, A. et al. Acute myeloid leukemia with biallelic CEBPA gene mutations and normal karyotype represents a distinct genetic entity associated with a favorable clinical outcome. J. Clin. Oncol. 28, 570–577 (2010).
  159. Taskesen, E. et al. Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity. Blood 117, 2469–2475 (2011).
  160. Metzeler, K. H. et al. TET2 mutations improve the new European LeukemiaNet risk classification of acute myeloid leukemia: a Cancer and Leukemia Group B study. J. Clin. Oncol. 29, 1373–1381 (2011).
  161. Weissmann, S. et al. Landscape of TET2 mutations in acute myeloid leukemia. Leukemia 26, 934–942 (2012).
  162. Hou, H. A. et al. WT1 mutation in 470 adult patients with acute myeloid leukemia: stability during disease evolution and implication of its incorporation into a survival scoring system. Blood 115, 5222–5231 (2010).
  163. Paschka, P. et al. Wilms' tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a cancer and leukemia group B study. J. Clin. Oncol. 26, 4595–4602 (2008).
  164. Yamaguchi, S. et al. IDH1 and IDH2 mutations confer an adverse effect in patients with acute myeloid leukemia lacking the NPM1 mutation. Eur. J. Haematol. 92, 471–477 (2014).
  165. Paschka, P. et al. IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J. Clin. Oncol. 28, 3636–3643 (2010).
  166. Mendler, J. H. et al. RUNX1 mutations are associated with poor outcome in younger and older patients with cytogenetically normal acute myeloid leukemia and with distinct gene and MicroRNA expression signatures. J. Clin. Oncol. 30, 3109–3118 (2012).
  167. Van Vlierberghe, P. et al. PHF6 mutations in adult acute myeloid leukemia. Leukemia 25, 130–134 (2011).
  168. Hou, H. A. et al. TP53 mutations in de novo acute myeloid leukemia patients: longitudinal follow-ups show the mutation is stable during disease evolution. Blood Cancer J. 5, e331 (2015).
  169. Wang, X. et al. EZH2 mutations are related to low blast percentage in bone marrow and −7/del(7q) in de novo acute myeloid leukemia. PLoS ONE 8, e61341 (2013).
  170. Lee, J. W. et al. The JAK2 V617F mutation in de novo acute myelogenous leukemias. Oncogene 25, 1434–1436 (2006).