MLN4924
MLN4924 is a small-molecule inhibitor of NEDD8-activating enzyme, a cullin-dependent E3 ligase controlling ubiquitination and hence affecting cellular growth, proliferation, and apoptosis. A phase I trial using various schedules and doses of MLN4924 as a single agent have been conducted in patients with relapsed or refractory AML/MDS, and a recommended phase II dose has been determined for further studies.[92] Trials of MLN4924 in combination with HMAs are now underway.
Epigenetic modulators
HMAs, including decitabine and 5-azacytidine, are commonly considered as potential therapies for patients with previously untreated AML, especially individuals who are not suitable candidates for induction chemotherapy. SGI-110, a second-generation HMA, is a dinucleotide of decitabine with a longer half-life. In a randomized phase II trial of patients with relapsed or refractory AML, and of older patients with previously untreated AML who were not fit for induction chemotherapy, the CR rate in those patients with relapsed/refractory AML was 16%, while the CR rate in the previously untreated group was 53%.[59] A phase III study to definitively investigate the efficacy of SGI-110 will be conducted.
EPZ-5676 is a small-molecule inhibitor of DOT1L, a histone methyltransferase that is crucial for the development and maintenance of MLL-rearranged leukemias. In a phase I clinical trial of EPZ-5676, 2 of 28 patients who were evaluable for antileukemic activity achieved CR, and 2 additional patients had resolution of leukemia cutis.[93]
Bromodomain and extraterminal (BET) family proteins are also attractive targets, since inhibitors can be directed against epigenetic regulators that maintain aberrant chromatin states commonly associated with AML. Both preclinical studies, as well as initial clinical trials with the BET inhibitor OTX015, have shown promise.[94]
Lastly, histone deacetylases (HDACs) are a well-known class of enzymes that affect gene expression via the removal of acetyl groups on core lysine groups in nucleosomal histones and other chromatin proteins.[95] HDAC inhibitors alter the landscape of acetylation, thereby influencing cell-cycle arrest, growth inhibition, and apoptosis. Leukemic cells are one of the many cell lines affected by HDAC inhibitors. HMAs and HDACs have been combined, with the rationale that they would have a possible synergistic effect on aberrant epigenetic changes in AML blasts that would restore the expression of silenced tumor suppressor genes. However, no definitive clinical benefit of the combination has been shown after more than 10 years of study. More recently, newer HDAC inhibitors (eg, pracinostat, panobinostat) are in the process of being investigated in clinical trials, alone or in combination with chemotherapy, HMAs, or other novel molecularly targeted compounds.[95]
ABT-199
ABT-199 (GDC-0199) is a highly selective antagonist of the antiapoptotic BCL-2 protein (which may mediate chemoresistance). In a recent trial, patients with refractory/relapsed AML, and patients who were treatment-naive but not able to undergo intensive therapy, were given ABT-199, with excellent responses, especially in patients with IDH mutations.[96] ABT-199 is now being tested in combination with HMAs or chemotherapy.
Immunotherapeutics
SGN-CD33A is a humanized CD33 antibody conjugated to a pyrrolobenzodiazepine (PBD) dimer that is a highly potent DNA binding agent. Early results from phase I clinical trials have recently been presented. In one such trial-of SGN-CD33A monotherapy in patients who had CD33-positive AML in relapse or who had declined intensive induction chemotherapy-a dose of 40 μg/kg led to a 60% OR rate in treatment-naive patients.[97] In another phase I trial-in treatment-naive CD33-positive AML patients who were not eligible for or who declined conventional intensive chemotherapy-combining SGN-CD33A and an HMA was determined to be tolerable, with an OR rate (CR + CR with incomplete blood count recovery) of 65%.[98]
Chimeric antigen receptors (CARs) that redirect T-cell specificity towards antigens that are enriched on the surface of “bulk” blasts and/or leukemic stem cells have also recently begun to be explored in clinical trials.[99] A phase I trial of CD123 CAR T cells in patients with refractory or relapsed disease (including individuals who have already undergone alloHSCT) is ongoing at our institution.
Whether these immunotherapeutic approaches will benefit from being used in combination with the emerging immune checkpoint inhibitors is an exciting area of intensive clinical research.
Conclusions
Emerging biological insights and new molecular and immunotherapeutic approaches are generating exciting results for AML patients, and while preliminary, these may result in improvement in outcome, in particular for those patients most in need: older patients and those with high-risk disease. The challenge is how to match molecular and clinical information with emerging compounds in order to select the best treatment for individual patients. The success of personalized approaches in AML is likely to depend on our ability to readily attain molecular information and on access to new drugs, in addition to several other key factors. The latter include a critical revision of more traditional clinical trial designs in favor of novel approaches that are more likely to identify drugs’ early biologic and clinical activities; increased referral of patients to tertiary centers by community oncologists; and close collaboration among cancer center networks, oncology cooperative groups, pharmaceutical sponsors, and regulatory authorities.
Financial Disclosure:The authors have no significant financial interest in or other relationship with the manufacturer of any product or provider of any service mentioned in this article.
References:
1. National Institutes of Health/National Cancer Institute. SEER Stat Fact Sheets: acute myeloid leukemia (AML). http://seer.cancer.gov/statfacts/html/amyl.html. Accessed March 15, 2016.
2. Swerdlow SH, Campo E, Harris NL, et al, editors. WHO classification of tumours of haematopoietic and lymphoid tissues. Lyon, France: IARC Press; 2008.
3. Godley LA. Inherited predisposition to acute myeloid leukemia. Semin Hematol. 2014;51:306-21.
4. Polprasert C, Schulze I, Sekeres MA, et al. Inherited and somatic defects in DDX41 in myeloid neoplasms. Cancer Cell. 2015;27:658-70.
5. Bueso-Ramos CE, Kanagal-Shamanna R, Routbort MJ, Hanson CA. Therapy-related myeloid neoplasms. Am J Clin Pathol. 2015;144:207-18.
6. Gale RP, Bennett JM, Hoffman FO. Therapy-related AML: a slip of the lip can sink a ship. Leuk Res. 2014;38:418-20.
7. Döhner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453-74.
8. Mrózek K, Marcucci G, Nicolet D, et al. Prognostic significance of the European LeukemiaNet standardized system for reporting cytogenetic and molecular alterations in adults with acute myeloid leukemia. J Clin Oncol. 2012;30:4515-23.
9. O’Donnell MR, Tallman MS, Abboud CN, et al. National Comprehensive Cancer Network Clinical Guidelines in Oncology (NCCN Guidelines). Acute myeloid leukemia. Version 1.2015. http://www.NCCN.org/publications/physician_gls/pdf/aml.pdf. Accessed March 15, 2016.
10. Grimwade D, Hills RK, Moorman AV, et al; National Cancer Research Institute Adult Leukaemia Working Group. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood. 2010;116:354-65.
11. Grimwade D, Mrózek K. Diagnostic and prognostic value of cytogenetics in acute myeloid leukemia. Hematol Oncol Clin North Am. 2011;25:1135-61.
12. Schanz J. Helpful tool or oversimplification? Concept of the monosomal karyotype from the clinical and cytogenetic point of view. Biol Blood Marrow Transplant. 2016;22:191-2.
13. Marcucci G, Haferlach T, Döhner H. Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications. J Clin Oncol. 2011;29:475-86.
14. Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368:2059-74.
15. Coombs CC, Tallman MS, Levine RL. Molecular therapy for acute myeloid leukaemia. Nat Rev Clin Oncol. 2015 Dec 1. [Epub ahead of print]
16. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98:1752-9.
17. Whitman SP, Archer KJ, Feng L, et al. Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a Cancer and Leukemia Group B study. Cancer Res. 2001;61:7233-9.
18. Schlenk RF, Döhner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358:1909-18.
19. Mead AJ, Linch DC, Hills RK, et al. FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia. Blood. 2007;110:1262-70.
20. Allen C, Hills RK, Lamb K, et al. The importance of relative mutant level for evaluating impact on outcome of KIT, FLT3 and CBL mutations in core-binding factor acute myeloid leukemia. Leukemia. 2013;27:1891-901.
21. Poiré X, Moser BK, Gallagher RE, et al. Arsenic trioxide in front-line therapy of acute promyelocytic leukemia (C9710): prognostic significance of FLT3 mutations and complex karyotype. Leuk Lymphoma. 2014;55:1523-32.
22. Paschka P, Marcucci G, Ruppert AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B study. J Clin Oncol. 2006;24:3904-11.
23. Falini B, Martelli MP. Impact of genomics in the clinical management of patients with cytogenetically normal acute myeloid leukemia. Best Pract Res Clin Haematol. 2015;28:90-7.
24. Falini B, Mecucci C, Tiacci E, et al; GIMEMA Acute Leukemia Working Party. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005;352:254-66.
25. Gale RE, Green C, Allen C, et al. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood. 2008;111:2776-84.
26. Becker H, Marcucci G, Maharry K, et al. Favorable prognostic impact of NPM1 mutations in older patients with cytogenetically normal de novo acute myeloid leukemia and associated gene- and microRNA-expression signatures: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28:596-604.
27. Wouters BJ, Löwenberg B, Erpelinck-Verschueren CA, et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood. 2009;113:3088-91.
28. Mendler JH, Maharry K, Radmacher MD, 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. 2012;30:3109-18.
29. Rücker FG, Schlenk RF, Bullinger L, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012;119:2114-21.
30. Yang L, Rau R, Goodell MA. DNMT3A in haematological malignancies. Nat Rev Cancer. 2015;15:152-65.
31. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363:2424-33.
32. Gaidzik VI, Schlenk RF, Paschka P, et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Blood. 2013;121:4769-77.
33. Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2012;30:742-50.
34. Gale RE, Lamb K, Allen C, et al. Simpson’s paradox and the impact of different DNMT3A mutations on outcome in younger adults with acute myeloid leukemia. J Clin Oncol. 2015;33:2072-83.
35. Patel JP, Gonen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366:1079-89.
36. Luskin MR, Lee JW, Fernandez HF, et al. Benefit of high dose daunorubicin in AML induction extends across cytogenetic and molecular groups: updated analysis of E1900. Blood. 2016 Jan 11. [Epub ahead of print]
37. Metzeler KH, Walker A, Geyer S, et al. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia. 2012;26:1106-7.
38. Parker SJ, Metallo CM. Metabolic consequences of oncogenic IDH mutations. Pharmacol Ther. 2015;152:54-62.
39. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18:553-67.
40. Mardis ER, Ding L, Dooling DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009;361:1058-66.
41. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28:2348-55.
42. Paschka P, Schlenk RF, Gaidzik VI, 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. 2010;28:3636-43.
43. Metzeler KH, Maharry K, Radmacher MD, 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. 2011;29:1373-81.
44. Gaidzik VI, Paschka P, Spath D, et al. TET2 mutations in acute myeloid leukemia (AML): results from a comprehensive genetic and clinical analysis of the AML Study Group. J Clin Oncol. 2012;30:1350-7.
45. Metzeler KH, Becker H, Maharry K, et al. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN favorable genetic category. Blood. 2011;118:6920-9.
46. Whitman SP, Ruppert AS, Marcucci G, et al. Long-term disease-free survivors with cytogenetically normal acute myeloid leukemia and MLL partial tandem duplication: a Cancer and Leukemia Group B study. Blood. 2007;109:5164-7.
47. Becker H, Marcucci G, Maharry K, et al. Mutations of the Wilms tumor 1 gene (WT1) in older patients with primary cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. Blood. 2010;116:788-92.
48. Gaidzik VI, Schlenk RF, Moschny S, et al. Prognostic impact of WT1 mutations in cytogenetically normal acute myeloid leukemia: a study of the German-Austrian AML Study Group. Blood. 2009;113:4505-11.
49. Pautas C, Merabet F, Thomas X, et al. Randomized study of intensified anthracycline doses for induction and recombinant interleukin-2 for maintenance in patients with acute myeloid leukemia age 50 to 70 years: results of the ALFA-9801 study. J Clin Oncol. 2010;28:808-14.
50. Fernandez HF, Sun Z, Yao X, et al. Anthracycline dose intensification in acute myeloid leukemia. N Engl J Med. 2009;361:1249-59.
51. Löwenberg B, Ossenkoppele GJ, van Putten W, et al; Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON), German AML Study Group (AMLSG), Swiss Group for Clinical Cancer Research (SAKK) Collaborative Group. High-dose daunorubicin in older patients with acute myeloid leukemia. N Engl J Med. 2009;361:1235-48.
52. Burnett AK, Russell NH, Hills RK, et al. A randomized comparison of daunorubicin 90mg/m2 vs 60mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients. Blood. 2015;125:3878-85.
53. Löwenberg B. Sense and nonsense of high-dose cytarabine for acute myeloid leukemia. Blood. 2013;121:26-8.
54. Wang ES. Treating acute myeloid leukemia in older adults. Hematology Am Soc Hematol Educ Program. 2014;2014:14-20.
55. Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose cytarabine and hydoxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 2007;109:1114-24.
56. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126:291-9.
57. 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.
58. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proc Natl Acad Sci USA. 2010;107:7473-8.
59. Issa JP, Roboz G, Rizzieri D, et al. Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: a multicentre, randomised, dose-escalation phase 1 study. Lancet Oncol. 2015;16:1099-110.
60. Bloomfield CD, Lawrence D, Byrd JC, et al. Frequency of prolonged remission duration after high-dose cytarabine intensification in acute myeloid leukemia varies by cytogenetic subtype. Cancer Res. 1998;58:4173-9.
61. Cornelissen JJ, Blaise D. Hematopoietic stem cell transplantation for patients with AML in first complete remission. Blood. 2016;127:62-70.
62. Versluis J, Hazenberg CL, Passweg JR, et al; HOVON and SAKK Leukemia Groups. Post-remission treatment with allogeneic stem cell transplantation in patients aged 60 years and older with acute myeloid leukaemia: a time-dependent analysis. Lancet Haematol. 2015;2:e427-e436.
63. Grimwade D, Freeman SD. Defining minimal residual disease in acute myeloid leukemia: Which platforms are ready for “prime time”? Blood. 2014;124:3345-55.
64. Hokland P, Ommen HB, Mulé MP, Hourigan CS. Advancing the minimal residual disease concept in acute myeloid leukemia. Semin Hematol. 2015;52:184-92.
65. Paietta E. Should minimal residual disease guide therapy in AML? Best Pract Res Clin Haematol. 2015;28:98-105.
66. Walter RB, Buckley SA, Pagel JM, et al. Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission. Blood. 2013;122:1813-21.
67. Yin JA, O’Brien MA, Hills RK, et al. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: results of the United Kingdom MRC AML-15 trial. Blood. 2012;120:2826-35.
68. Krönke J, Schlenk RF, Jensen KO, et al. Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: a study from the German-Austrian acute myeloid leukemia study group. J Clin Oncol. 2011;29:2709-16.
69. Stone RM. Should older adults with AML receive post-remission therapy? Best Pract Res Clin Haematol. 2015 Oct 20. [Epub ahead of print]
70. Thol F, Schlenk RF, Heuser M, Ganser A. How I treat refractory and early relapsed acute myeloid leukemia. Blood. 2015;126:319-27.
71. Marcucci G, Yan P, Maharry K, et al. Epigenetics meets genetics in acute myeloid leukemia: clinical impact of a novel seven-gene score. J Clin Oncol. 2014;32:548-56.
72. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481:506-10.
73. Ustun C, Marcucci G. Emerging diagnostic and therapeutic approaches in core binding factor acute myeloid leukaemia. Curr Opin Hematol. 2015;22:85-91.
74. Burnett AK, Hills RK, Milligan D, et al. Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial. J Clin Oncol. 2011;29:369-77.
75. Hills RK, Castaigne S, Appelbaum FR, 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. 2014;15:986-96.
76. Marcucci G, Geyer S, Zhao J. Adding the KIT inhibitor dasatinib (DAS) to standard induction and consolidation therapy for newly diagnosed patients (pts) with core binding factor (CBF) acute myeloid leukemia (AML): initial results of the CALGB 10801 (Alliance) study. Blood. 2013;122:357.
77. Grunwald MR, Levis MJ. FLT3 tyrosine kinase inhibition as a paradigm for targeted drug development in acute myeloid leukemia. Semin Hematol. 2015;52:193-9.
78. Röllig C, Serve H, Hüttmann A, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16:1691-9.
79. Serve H, Krug U, Wagner R, 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. 2013;31:3110-8.
80. Stone RM, Mandrekar S, Sanford BL, et al. The multi-kinase inhibitor midostaurin (M) prolongs survival compared with placebo (P) in combination with daunorubicin (D)/cytarabine (C) induction (ind), high-dose C consolidation (consol), and as maintenance (maint) therapy in newly diagnosed acute myeloid leukemia (AML) patients (pts) age 18-60 with FLT3 mutations (muts): an international prospective randomized (rand) P-controlled double-blind trial (CALGB 10603/RATIFY [Alliance]) [abstract]. Blood. 2015;126:6.
81. Cortes JE, Kantarjian H, Foran JM, 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. 2013;31:3681-7.
82. Randhawa JK, Kantarjian HM, Borthakur G, et al. Results of a phase II study of crenolanib in relapsed/refractory acute myeloid leukemia patients (pts) with activating FLT3 mutations [abstract]. Blood. 2014;124:389.
83. Levis MJ, Perl AE, Altman JK, et al. Results of a first-in-human, phase I/II trial of ASP2215, a selective, potent inhibitor of FLT3/Axl in patients with relapsed or refractory (R/R) acute myeloid leukemia (AML). J Clin Oncol. 2015;33:abstr 7003.
84. Stein EM, Altman JK, Collins R, 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 [abstract]. Blood. 2014;124:115.
85. 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.
86. Cortes JE, Goldberg SL, Feldman EJ, et al. Phase II, multicenter, randomized trial of CPX-351 (cytarabine:daunorubicin) liposome injection versus intensive salvage therapy in adults with first relapse AML. Cancer. 2015;121:234-42.
87. Ravandi F, Ritchie EK, Sayar H, et al. Vosaroxin plus cytarabine versus placebo plus cytarabine in patients with first relapsed or refractory acute myeloid leukaemia (VALOR): a randomised, controlled, double-blind, multinational, phase 3 study. Lancet Oncol. 2015;16:1025-36.
88. Dennis M, Russell N, Hills RK, et al. Vosaroxin and vosaroxin plus low-dose Ara-C (LDAC) vs low-dose Ara-C alone in older patients with acute myeloid leukemia. Blood. 2015;125:2923-32.
89. Döhner H, Lübbert M, Fiedler W, et al. Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood. 2014;124:1426-33.
90. Kojima K, Kornblau SM, Ruvolo V, et al. Prognostic impact and targeting of CRM1 in acute myeloid leukemia. Blood. 2013;121:4166-74.
91. Yee K, Savona M, Sorensen M, et al. A phase 1 dose-escalation study of the oral selective inhibitor of nuclear export (SINE) KPT-330 (selinexor) in patients (pts) with relapsed/refractory acute myeloid leukemia (AML). J Clin Oncol. 2014;32(suppl 5s):abstr 7032.
92. Swords RT, Erba HP, DeAngelo DJ, et al. Pevonedistat (MLN4924), a first-in-class NEDD8-activating enzyme inhibitor, in patients with acute myeloid leukaemia and myelodysplastic syndromes: a phase 1 study. Br J Haematol. 2015;169:534-43.
93. Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L inhibitor EPZ-5676: safety and activity in relapsed/refractory patients with MLL-rearranged leukemia [abstract]. Blood. 2014;124:387.
94. Dombret H, Preudhomme C, Berthon C, et al. A phase 1 study of the BET-bromodomain inhibitor OTX015 in patients with advanced acute leukemia [abstract]. Blood. 2014;124:117.
95. Quintás-Cardama A, Santos FP, Garcia-Manero G. Histone deacetylase inhibitors for the treatment of myelodysplastic syndrome and acute myeloid leukemia. Leukemia. 2011;25:226-35.
96. Konopleva M, Pollyea DA, Potluri J, et al. A phase 2 study of ABT-199 (GDC-0199) in patients with acute myelogenous leukemia (AML) [abstract]. Blood. 2014;124:118.
97. Stein EM, Stein A, Walter RB, et al. Interim analysis of a phase 1 trial of SGN-CD33A in patients with CD33-positive acute myeloid leukemia (AML). Blood. 2014;124:abstr 623.
98. Fathi AT, Erba HP, Lancet JE, et al. SGN-CD33A plus hypomethylating agents: a novel, well-tolerated regimen with high remission rate in frontline unfit AML [abstract]. Blood. 2015;126:454.
99. Mardiros A, Forman SJ, Budde LE. T cells expressing CD123 chimeric antigen receptors for treatment of acute myeloid leukemia. Curr Opin Hematol. 2015;22:484-8.