Abstract
The recently updated World Health Organization (WHO) Classification of myeloid neoplasms and leukemia reflects the fact that research in the underlying pathogenic mechanisms of acute myeloid leukemia (AML) has led to remarkable advances in our understanding of the disease. Gene mutations now allow us to explore the enormous diversity among cytogenetically defined subsets of AML, particularly the large subset of cytogenetically normal AML. Despite the progress in unraveling the tumor genome, only a small number of recurrent mutations have been incorporated into risk-stratification schemes and have been proven to be clinically relevant, targetable lesions. We here discuss the utility of molecular markers in AML in prognostication and treatment decision making, specifically highlighting the aberrations included in the current WHO classification.
Keywords: acute myeloid leukemia, molecular markers, prognostic impact
1 |. INTRODUCTION
In acute myeloid leukemia (AML), recurrent cytogenetic abnormalities are established diagnostic and prognostic markers, suggesting that acquired genetic abnormalities play an essential role in leukemogenesis.1 The prognosis for patients with AML is determined to a large degree by the biology of the disease. Roughly 45% of the patients harbor a normal karyotype as detected by conventional cytogenetics at diagnosis, yet a somatic mutation can be identified in 97.3% of the cases.2 Targeted sequencing has identified several mutations that carry prognostic information, including mutations in FLT3, NPM1, KIT, CEBPA, and TET2.3 In addition, massively parallel sequencing led to the discovery of recurrent mutations in DNMT3A and IDH.4,5
The recently updated World Health Organization (WHO) classification of myeloid neoplasms and leukemia reflects the fact that research in the underlying pathogenic mechanisms of AML has led to remarkable advances in our understanding of the disease.5 Currently, the WHO classification includes eight AML categories defined by recurrent genetic abnormalities (including AML with t(9;22)(q34;q11.2)/BCR-ABL1 as a provisional entity) as well as three categories defined by gene mutations, that is, AML with NPM1, AML with biallelic mutated CEBPA, and the provisional entity AML with mutated RUNX1. Consistently, AMLs defined by recurrent genetic abnormalities are associated with distinctive clinicopathological features and impact prognosis. AML with FLT3, however, is not included as a separate entity, because it occurs across multiple subtypes. However, the WHO classification acknowledges that FLT3 should be tested in all AML cases.5
Although new molecular analysis techniques, such as ultra-deep sequencing, has helped to identify numerous recurrent genetic abnormalities, to date, however, only a limited number have been incorporated into risk-stratification schemes, such as the National Comprehensive Cancer Network or European LeukemiaNet (ELN) Guidelines (Table 1).6 In addition, increasing evidence indicates that the presence of minimal residual disease (MRD), measured either molecularly or by multiparameter flow cytometry, identifies patients at particularly high risk of relapse and provides powerful prognostic information beyond pretreatment characteristics such as cytogenetic or molecular abnormalities.7 Nonetheless, in adult AML MRD as a tool to fine-tune risk assessment during postremission therapy with adaption of treatment strategy is lagging behind acute lymphoblastic leukemia (ALL), acute promyelocytic leukemia (APL), or chronic myeloid leukemia (CML), in which MRD is now routinely used to guide treatment decisions at predefined check-points during therapy.8–12 Here, we review genetic abnormalities that should be used for treatment stratification in AML and their clinical implications.
TABLE 1.
Risk category | Genetic abnormality |
---|---|
Favorable | t(8;21)(q22;q22.1); RUNX1-RUNX1T1 inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11 Mutated NPM1 without FLT3-ITD or with FLT3-ITD lowb Biallelic mutated CEBPA |
Intermediate | Mutated NPM1 and FLT3-ITD highb Wild-type NPM1 without FLT3-ITD or with FLT3-ITD lowb (w/o adverse-risk genetic lesions) t(9;11)(p21.3;q23.3); MLLT3-KMT2Ac Cytogenetic abnormalities not classified as favorable or adverse |
Adverse | t(6;9)(p23;q34.1); DEK-NUP214 t(v;11q23.3); KMT2A rearranged t(9;22)(q34.1;q11.2); BCR-ABL1 inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2); GATA2, MECOM(EVI1) −5 or del(5q); −7; −17/abn(17p) Complex karyotype,d monosomal karyotypea Wild-type NPM1 and FLT3-ITD highb Mutated RUNX1e Mutated ASXL1e Mutated TP53f |
Modified according to Ref.13 Defined by the presence of one single monosomy (excluding loss of X or Y) in association with at least one additional monosomy or structural chromosome abnormality (excluding core-binding factor AML).13
Low, low allelic ratio (<0.5); high, high allelic ratio (>0.5); as determined by GeneScan analysis.
The presence of t(9;11)(p21.3;q23.3) takes precedence over rare, concurrent adverse-risk gene mutations.
Three or more unrelated chromosome abnormalities in the absence of one of the World Health Organization-designated recurring translocations or inversions, that is, t(8;21), inv(16) or t(16;16), t(9;11), t(v;11)(v;q23.3), t(6;9), inv(3) or t(3;3); AML with BCR-ABL1.
These markers should not be used as an adverse prognostic marker if they co-occur with favorable-risk AML subtypes.
TP53 mutations are significantly associated with AML with complex and monosomal karyotype.132
2 |. CORE-BINDING FACTOR LEUKEMIA
In this category, AMLs characterized by the balanced translocation t(8;21)(q22;q22.1) and with the pericentric inversion inv(16) (p13.1q22) or the less frequent balanced translocation t(16;16) (p13.1;q22) are grouped and considered as AML regardless of bone marrow blast cell counts at diagnosis.6 Both t(8;21), involving RUNX1-RUNX1T1, and inv(16)/t(16;16), involving CBFB-MYH11, harbor chimeric fusions involving genes of the core-binding factor (CBF) complex, a major regulator of hematopoiesis, providing the common designation CBF-AML. Patients with CBF-AML are considered to have a favorable prognosis as compared to other genetic risk groups.13 After an anthracycline- and cytarabine-based induction chemotherapy, about 90% of CBF-AML patients achieve a complete remission (CR)14,15 and repeated cycles of postremission chemotherapy with high-dose cytarabine (HiDAC) (usually 3–4) have emerged as preferred treatment of CBF-AML.16–18 The relapse risk may not exceed 20–35% in 3–5 years with repeated courses of intensive consolidation therapy.15,19–22 Consequently, these patients have, on average, no survival advantage with allogeneic stem cell transplantation (allo-SCT) while in first remission because the transplant-related mortality is greater than the decrease in relapse rates afforded by the transplant.23,24
Recently published data from the Medical Research Council (MRC) AML15 trial in younger adult patients with favorable-risk AML indicated an improved survival after therapy with fludarabine, cytarabine, granulocyte colony-stimulating factor, and idarubicin (FLAG-Ida) followed by two cycles of HiDAC consolidation with an 8-year survival rate of 95%.25 In addition, the combination of gemtuzumab ozogamicin (GO) with chemotherapy has been shown to improve overall survival (OS) in subgroup analysis of CBF-AML,26–28 suggesting that higher intensity regimens may lead to deeper log reductions after the first course of chemotherapy as has been shown for the addition of GO to intensive chemotherapy20 as well as for anthracycline dose intensification during intensive induction chemotherapy.29 Patients who received daunorubicin of 90 mg/m2 showed a faster and deeper MRD reduction and achieved a higher proportion of complete molecular responses (at least 3 log reduction as compared to diagnosis as measured by real-time quantitative polymerase chain reaction (RT-qPCR) with at least 10−4 sensitivity) that translated into a reduced relapse rate as compared to those patients receiving 60 mg/m2. As a consequence, the combination of FLAG-GO (GO 3 mg/m2 intravenously on day 1 during induction and two of six postremission cycles) has been evaluated as front-line therapy in 45 younger adult patients (median age, 48 years) with CBF-AML.30 This regimen resulted in a high CR rate of 95% with 5% induction deaths. The OS and relapse-free survival (RFS) probability at 3 years were 78% and 85%, respectively, suggesting that a deeper molecular remission could be achieved as compared to the standard HiDAC approach, though no head-to-head comparison of the reduction in measurable disease (MRD) according to treatment strategy had been performed. However, even though MRD-positive CBF-AML patients have higher relapse rates, it has only been shown that therapy intensification based upon the presence of either MRD or pretreatment mutations like KIT improves event-free survival (EFS), but not OS, which is in part reflected by a high response rate to salvage chemotherapy.31 Taken together, CBF-AML is a very chemo-responsive disease so, in this case, more chemotherapy (whichever we choose—HiDAC, FLAG-Ida, dose-intensified daunorubicin, or FLAG-GO) in patients who can tolerate it will lead to a higher cure rate.
In addition, several variables associated with worse outcome have been recognized in at least some studies, including a high white blood cell count (WBC) and the presence of KIT or FLT3 mutations at diagnosis.15,32–36 In CBF-AML, KIT mutations occur in about 20–30% of the patients and have been associated with an adverse prognostic impact on survival.33,34 However, based on a previous report on AML with inv(16)/t(16,16), the unfavorable impact on relapse rate did not translate into an inferior survival.37 In contrast, AML with inv(16)/t(16,16) harboring additional FLT3 mutations was associated with a strong negative impact in multivariable analysis on OS.37 In a study by Boissel et al35 who evaluated 103 pediatric and adult patients with CBF-AML, KIT mutations occurred in 17% and were associated with a shorter EFS and RFS (P = 0.002 and P = 0.003) in t(8;21) but not inv(16) patients. Again, FLT3 mutations (1% ITD; 7% TKD835) were significantly associated with a shorter EFS and OS (P < 0.0001 and P = 0.0002), owing to an excess of early events. In addition, Allen et al38 noted that the relative KIT mutant level should be taken into account, since in their analysis on 354 younger adult patients with CBF-AML, only KIT mutations with a mutant level greater than 25% increased the risk of relapse. Thus, due to the inconsistencies in the available data, cooperating gene mutations in CBF-AML should currently not be used to guide treatment decisions.
In addition, the MRD level seems to be an important prognostic factor. Recent studies have highlighted the heterogeneity of the disease by identifying subsets of patients with distinct risks of disease recurrence based on the degree of reduction in RUNX1-RUNX1T1/CBFB-MYH11 transcripts.20–22 Currently available evidence suggests that optimal outcomes are achieved when patients with CBF-AML obtain either a molecular remission by RT-qPCR or very significant reductions in RUNX1-RUNX1T1/CBFB-MYH11 transcripts with induction and postremission therapy (defined as at least a > 3 log reduction of transcript levels from baseline after consolidation therapy);20,22,39 higher intensity regimens may lead to deeper log reductions after chemotherapy.20,22,40 Moreover, emerging evidence from a study by Jourdan et al39 suggests that information from post-treatment RUNX1-RUNX1T1 transcript levels may be preferable over high WBC or KIT/FLT3 mutational status to identify patients with high-risk t(8;21)(q22;q22) AML, as only MRD but not the other factors had a significant prognostic impact in multivariate analyses.
3 |. ACUTE PROMYELOCYTIC LEUKEMIA
Acute promyelocytic leukemia (APL) with the balanced translocation t(15;17)(q22;q12) involving PML-RARA accounts for 5–8% of AML cases with a lower incidence in elderly patients and is considered as AML regardless of bone marrow blast cell counts at diagnosis.6 In APL, molecular assessment of disease response has become standard practice, and MRD-directed therapy quite plausibly improves outcome, particularly in patients with high-risk disease.41,42 The chemotherapy-free regimen with arsenic trioxide and all-trans retinoic acid has proven to be highly effective in de novo APL and has become standard first-line therapy in younger adult, non-high-risk (pretreatment WBC ≤ 10 × 109/L) patients, providing the first paradigm of a molecularly targeted treatment.43 Nevertheless, early death is still a major issue in APL, particularly in older patients, emphasizing the need of rapid diagnostics and supportive care together with immediate access to ATRA-based therapy. To date, data on the impact of additional chromosomal or genetic aberrations including FLT3 mutations on outcome in APL patients are still conflicting.43–52 In addition, there seems to be an association of higher WBC at diagnosis and FLT3-ITD mutations in patients with APL as compared to FLT3 wild-type patients,48–52 which may affect outcome. However, recently published data by Cicconi et al49 indicate that FLT3-ITD mutations have no prognostic impact in APL patients treated with ATO/ATRA. Taken together, due to the inconsistencies in the available data and due to its high cure rate, there is currently no convincing evidence that FLT3 mutations should currently be used to guide treatment decisions in APL.
4 |. AML WITH t(6;9)(p23;q34.1)/DEK-NUP214
The translocation t(6;9)(p23;q34.1), which results in formation of the DEK-NUP214 chimeric fusion gene, was first described in AML in 197653 and acts as an aberrant transcription factor.54 It alters the nuclear transport by binding soluble transport factors55 and has been reported to enhance protein synthesis in myeloid cells.56,57 In AML, t(6;9) accounts for only a very small subgroup (1–2%) and has been associated with a poor prognosis with 5-year OS rates of 28% reported in children and only 9% in adults.58 Overall, outcome in both pediatric and adult AML with t(6;9) is poor58–62 and adult patients with this translocation are assigned to the adverse-risk group.13 Of note, up to 90% of AML patients with t(6;9) are described to harbor a concomitant FLT3-ITD,58–60 whereas secondary cytogenetic abnormalities occur in roughly 20%.58,61 Currently, the impact of a concurrent FLT3-ITD is controversial. While results of a meta-analysis of 50 adult AML patients indicated an association between FLT3-ITD and an inferior outcome in t(6;9) AML,63 others were inconclusive due to the low number of FLT3-ITD-unmutated patients,58 or have not found a significant adverse impact.60,61
However, allo-SCT may improve survival if applied early during first CR.58,64 In a matched-pair analysis of de novo AML using data from the Japanese allo-SCT data registry, the outcome of 57 patients with t(6;9) was compared to that of 171 patients with normal karyotype.64 All patients received an allo-SCT between 1996 and 2007, either in first or second CR (CR1 and CR2, n = 116), or as salvage therapy (n = 112). In this matched-pair analysis in patients with t(6;9), the 5-year OS (45% vs 40%), disease-free survival (42% vs 33%), cumulative incidence of relapse (42 vs 45%), and non-relapse mortality (16% vs 22%) were not different as compared to normal karyotype AML.64 In addition, we have recently evaluated the impact of allo-SCT on outcome in n = 123 adult patients with t(6;9) in a large, international collaborative analysis.65 In our cohort, we observed a high CR rate of 79% including sixteen patients, who required an intensive salvage treatment cycle with HiDAC. Five-year OS rates after allo-SCT (n = 51) were very encouraging (55% vs 18%; P < 0.001) as compared to patients who received consolidation chemotherapy (n = 44) regardless of FLT3-ITD status, additional cytogenetic abnormalities, or timing of transplant. These results suggest that allo-SCT may overcome the adverse impact of t(6;9) in AML patients, rendering outcome comparable to patients with normal cytogenetics.
5 |. AML WITH inv(3)(q21.3q26.2) OR t(3;3) (q21.3;q26.2); GATA2, MECOM
AML with inv(3) or t(3;3) accounts for 1–2% of all AML and occurs most commonly in adults.6 AML with inv(3)/t(3;3) is most frequently associated with monosomy 7 or complex karyotype (≥3 chromosomal abnormalities in the absence of one of the WHO designated recurring translocations or inversions) and confers an unfavorable outcome.66 Genetically, inv(3)/t(3;3) involves MECOM (also termed EVI1) at 3q26.2 and repositions the GATA2 enhancer to activate MECOM expression, thereby inducing GATA2 haploinsufficiency.67 Gröschel et al68 showed that 98% of inv(3)/t(3;3) myeloid malignancies harbor mutations in genes activating RAS/receptor tyrosine kinase (RTK) signaling pathways. This high incidence of RAS/RTK signaling pathways may provide a target for a rational treatment strategy in this high-risk patient group. In addition, the authors could show that hemizygous mutations in GATA2, as well as heterozygous alterations in RUNX1, SF3B1, and genes encoding epigenetic modifiers, frequently co-occur with inv(3)/t(3;3) aberrations.68 Within their analysis, neither mutational patterns nor gene expression profiles differed across inv(3)/t(3;3) AML, CML, or myelodysplastic syndrome cases. The authors therefore suggested that inv(3)/t(3;3) myeloid malignancies should be regarded as a single disease entity irrespective of blast count.68 From a clinical aspect, however, there is currently no specific treatment available that has shown convincing improvement of outcome of this high-risk disease with only about one-third of patients with inv(3)(t(3;3) achieving a CR despite intensive induction therapy and few, if any, long-term survivors.66 Thus, investigational therapy within a clinical trial should be considered in these patients, whenever possible.
6 |. AML (MEGAKARYOBLASTIC) WITH t(1;22)(p13.3;q13.1)/RBM15-MKL1
Acute megakaryoblastic leukemia (AMKL) with t(1;22)(pp13.3;q13.1) is a subtype of AML in which the cells morphologically resemble abnormal megakaryoblasts. While AMKL is very rare in adults (~1%),69 it accounts for 4–15% of newly diagnosed childhood AML.69,70 In addition, congenital cases have been described71 as well as the occurrence in patients with Down syndrome.6 In most cases, t(1;22) (p13.3;q13.1) is the sole abnormality, leading to the fusion gene RBM15-MKL1.6 This fusion gene seems to be involved in modulation of chromatin organization, HOX-induced differentiation, and extra-cellular signaling pathways.72
Regarding outcome, data are contradictory with some reports indicating that childhood patients with t(1;22)(p13.3;q13.1) may respond well to intensive chemotherapy,73–77 but other studies indicated that this entity seems to be associated with a high-risk and inferior survival as compared to patients without t(1;22) (p13.3;q13.1).78,79 This might be attributable to the high rate of early deaths in patients with t(1;22)(p13.3;q13.1), particularly due to their very young age at diagnosis.78 Interestingly, in the publication by Schweitzer et al79 allo-SCT in first CR did not provide a significant survival benefit. Recently, de Rooij et al80 have evaluated cooperating mutations in 75 pediatric and 24 adult patients with non-Down syndrome AMKL by RNA and exome sequencing analysis to gain further insights into the genomic alterations that lead to non-Down syndrome AMKL. They identified chimeric oncogenes in a substantial number, including CBFA2T3-GLIS2 (18.6%), KMT2A gene rearrangements (17.4%), NUP98-KDM5A (11.6%), and RBM15-MKL1 (10.5%). They could show that outcome was based on chimeric oncogenes and cooperating mutations in epigenetic and kinase signaling genes, such as GATA1 (13.3%), Cohesin or CTCF genes (18.1%), JAK/STAT genes (16.9%), and RAS pathway genes (15.7%). Of note, all GATA1-mutated cases which lacked a fusion gene were cured, mimicking the excellent outcomes observed in Down syndrome AMKL.81 In contrast, patients with CBFA2T3-GLIS2, KMT2A gene rearrangements, and NUP98-KDM5A were associated with a poor prognosis and should therefore be candidates for allo-SCT.80
7 |. AML WITH BCR-ABL1
BCR-ABL1-mutated AML again is a rare subtype (<1%) that is now included as a provisional entity in the current WHO classification.6 Since a clear distinction between de novo BCR-ABL-mutated AML and CML in blast crisis is challenging in many cases, the existence of de novo BCR-ABL-mutated AML has been a matter of debate for a long time. However, increasing evidence suggests that BCR-ABL-mutated AML is in fact a distinct subgroup of AML.82,83 Preliminary data suggest that deletion of antigen receptor genes (immunoglobulin heavy chain and T-cell receptor), IKZF1, and/or CDKN2A may support a diagnosis of AML rather than CML blast phase.84
The prognosis of BCR-ABL-mutated AML seems to depend on the cytogenetic and/or molecular background rather than on BCR-ABL itself.84 A therapy with tyrosine kinase inhibitors (TKIs) such as imatinib, dasatinib, or nilotinib is reasonable,85 but their use cannot be routinely recommended in first-line therapy due to a lack of systematic clinical data. However, intensive chemotherapy in combination with TKI86,87 and/or as a bridge to allo-SCT seems to be a feasible approach.
8 |. AML WITH GENE MUTATIONS
8.1 |. AML with mutated NPM1
Frameshift mutations of the NPM1 gene are one of the most frequent molecular abnormalities in AML, particularly in patients with a normal karyotype88 with a high incidence in both young and older AML patients.89–91 NPM1 mutations result in cytoplasmic accumulation, regulating the ARF-p53 tumor suppressor pathway, thus controlling proliferation and apoptosis.92 In contrast to other molecular aberrations (eg, FLT3), NPM1 mutations are typically stable during the course of the disease, which supports the notion that they are an early pathogenetic lesion in AML.93 To date, more than 50 different NPM1 mutations have been reported; however, the subtypes A, B, and D comprise 90% of all variants.94 These three mutation subtypes have been shown to be reliable markers for MRD detection with high sensitivity.7,95 The same assay can be adapted for cases with rare NPM1 mutation variants by replacing mutation-specific primers, but case-specific RT-qPCRs need to be carefully established to avoid non-specific background amplification from the wild-type NPM1 allele.96
Concurrent mutations occur typically in FLT3, DNMT3A, IDH1/2, or TET2.88 In several studies, it has been shown that the prognostic impact of NPM1 should be interpreted in the context of a cooperating FLT3-ITD mutation, which is present in approximately 45% of this patient population with normal karyotype.97–99 In particular, in younger adult NPM1-mutated patients with high FLT3-ITD allelic ratio (≥0.5)13,100–102 the favorable prognostic effect of NPM1 is mitigated or even abolished as compared to patients with a low allelic ratio.21,101,102 In comparison, patients with mutated NPM1 without FLT3-ITD or FLT3-ITD with a low allelic ratio (< 0.5) are associated with a somewhat better outcome.13,102 These data have recently been confirmed in a large cohort of intensively treated adult AML patients.103 However, in patients with mutated NPM1 without FLT3-ITD two reports from cooperative study groups showed a negative impact of cooperating IDH1/2 mutations on RFS and OS.104,105 In contrast, Patel et al2 reported on a favorable impact of mutated NPM1 without FLT3-ITD only if cooperating IDH1/2 mutations were present. Such opposed effects of genotypes on outcome highlight statistical shortcomings of retrospective molecular studies.
Similar to the findings in CBF-AML, RT-qPCR assessment of MRD can distinguish patients at high risk of relapse: In a study on 245 adult patients with NPM1-mutated AML, relevant MRD check-points could be defined.106 Achievement of RT-qPCR negativity after two courses of induction therapy identified patients with a low CIR (6.5% after 4 years) as compared to RT-qPCR-positive patients (53% after 4 years; P < 0.001), translating into significant differences in OS (90% vs 51%, respectively; P = 0.001). After completion of therapy, CIR was 15.7% in RT-qPCR-negative patients as compared to 66.5% in RT-qPCR-positive patients (P < 0.001).106 These data are extended by the study of Hubmann et al107 in whom a NPM1 mutation cutoff level of 0.01 after induction therapy as measured by RT-qPCR (with a sensitivity of 10−6) was associated with a CIR after 2 years of 77.8% for patients with ratios above as compared to 26.4% for those with ratios below the cutoff. Within the ALFA-0701 trial, NPM1 MRD positivity as measured by RT-qPCR (quantitative detection limit of 0.1%) after one course of standard induction chemotherapy according to the 7 + 3 scheme with or without GO was associated with an increased cause-specific hazard of relapse of 3.66 (P = 0.035). Of note, NPM1-MRD was predictive for response to therapy since more MRD-negative results were obtained in patients treated in the GO arm as compared to those treated in the control arm after induction therapy (39% vs 7%; P = 0.006) as well as at the end of treatment (91% vs 61%; P = 0.028).108 This is one of the first randomized studies indicating that MRD assessment may serve as a surrogate for survival endpoints for the treatment under investigation. Additionally, in a retrospective analysis performed by the German Study Alliance Leukemia, increasing levels of NPM1 MRD were predictive of an impending relapse after chemotherapy (MRD increase >1% NPM1mut/ABL1) or allo-SCT (MRD increase >10% NPM1mut/ABL1).109 Importantly, MRD status has been found to be a better predictor of the relapse risk than FLT3-ITD in NPM1-mutated AML.110 Besides, different studies have convincingly demonstrated that MRD positivity by RT-qPCR before allo-SCT is independently associated with a significantly increased risk of relapse and inferior survival.111,112 Assuming that a further reduction of MRD levels optimizes outcome after allo-SCT, this relationship would justify risk-stratified treatment allocation, including the use of additional pretransplant chemotherapy. However, as MRD might simply reflect reduced sensitivity of leukemia cells to chemotherapy, the presence of residual disease might only mark those patients who are unlikely to be cured with subsequent similar-type therapies, even if disease levels are brought temporarily below the level of detection. Therefore, another approach could be pre-emptive immune113 or antibody therapy (ClinicalTrials.gov Identifier: NCT02789254) in MRD-positive patients, which has successfully been demonstrated in childhood AML with mixed chimerism after allo-SCT,114 or by post-transplant application of demethylating agents, such as azacitidine, to prevent imminent relapse in MRD-positive patients.115
8.2 |. AML with biallelic mutation of CEBPA
The CCAAT/enhancer binding protein alpha (CEBPA) gene encodes a transcription factor for granulocyte differentiation. CEBPA mutations prevent DNA binding, resulting in a lack of granulocyte differentiation.116 CEBPA mutations have been reported in roughly 10% of AML patients and half of them comprise biallelic mutations, generally involving both the N- and C-terminal domains.117 The frequency of biallelic CEBPA mutations seems to be age-dependent and decreases with increasing age.118–121 Several studies have shown convincingly that AML with biallelic mutation of CEBPA can be distinguished from AML with single mutated CEBPA with respect to biological and prognostic features and that the favorable prognostic impact of mutated CEBPA can be attributed to the subtype of AML with biallelic CEBPA mutations.118–121 In this subgroup of patients, CR rates of 85% to 92% have been reported; however, these results are mainly based on younger adults.118–121 Therefore, the current WHO classification has included AML with biallelic mutated CEBPA as a separate entity.6 Concurrent mutations most frequently include TET2 (34%), followed by GATA2 (21%), WT1 (13.7%), DNMT3A (9.6%), ASXL1 (9.5%), RAS mutations (NRAS, 8.4%; KRAS, 3.2%), IDH1/2, and FLT3-ITD (6.3%, each).122 Regarding the prognostic impact, concurrent TET2 mutations were associated with an unfavorable impact on OS as compared to TET2 wild-type patients (P = 0.035), whereas GATA2 mutations had a favorable impact (P = 0.032). The favorable prognostic impact of biallelic mutated CEBPA gets lost in the presence of FLT3-ITD.119,120,122 Given the high CR rate after reinduction therapy in younger adult relapsed AML patients with biallelic mutated CEBPA of 83% (n = 35/42) and favorable outcome after allo-SCT in CR2,123 we recommend HiDAC-based consolidation chemotherapy in patients with biallelic mutated CEBPA in CR1.
8.3 |. AML with mutated RUNX1
The Runt-related transcription factor 1 (RUNX1) gene encodes a transcription factor critical for hematopoiesis and was associated with embryonic lethality in a murine model.124 In AML, RUNX1 mutations have been reported to occur with an incidence of 5.6%15 to 13.2%,125 predominantly in patients with intermediate-risk cytogenetics. In patients with cytogenetically normal AML, the incidence seems to increase with higher age, with an incidence of 8% in younger patients compared with 16% in older patients.126 Interestingly, RUNX1 mutations are almost mutually exclusive of other disease-defining genetic aberrations such as NPM1, biallelic mutated CEBPA, CBFB-MYH11, RUNX1-RUNX1T1, and PML-RARA.126–128 In addition, RUNX1 mutations are characterized by a distinct gene expression pattern,125,127 and monoallelic germline mutations have been reported in rare cases of familial platelet disorder with predisposition to AML,128 further supporting the idea of a separate disease entity. Therefore, the new provisional entity “AML with mutated RUNX1” (excluding cases with myelodysplasia-related changes) was added to the current WHO classification.6 Approximately two-thirds of RUNX1 mutations are found in cytogenetically normal AML and have been associated with a very unfavorable prognosis in both young and elderly patients.126,127 Thus, RUNX1 was added to the adverse-risk group.13
8.4 |. AML with TP53
The tumor protein p53 (TP53) encodes a transcription factor, which is involved in cell cycle arrest and apoptosis.129 TP53 mutations occur in roughly 12% of AML patients,130 predominantly in therapy-related or secondary AML as well as in elderly patients.131 Moreover, TP53 alterations are found in roughly 70% of AML patients with a complex karyotype.132 TP53 alterations predict for very low CR rates (less than 30%) and were shown to be an independent poor prognostic factor among the subgroup of AML with complex karyotype.132 Interestingly, TP53 could be identified in hematopoietic stem and progenitor cells (HSPC) in chemotherapy-naïve controls and in therapy-related or secondary AML patients years prior to development of overt disease, suggesting that HSPC carrying TP53 may be chemotherapy-resistant and expand after treatment.133 Individuals with clonal hematopoiesis with indeterminate potential have a 13-fold increased risk of developing a hematologic malignancy, and the data by Wong et al133 suggest that this risk is increased in the context of cytotoxic therapy, at least if a TP53 mutation is present. Recently published data suggest that treatment with decitabine at a dose of 20 mg/m2 per day for 10 consecutive days in monthly cycles may improve the dismal outcome of AML with TP53 alterations.134 Although these responses were not durable, they resulted in OS rates that were similar to those among patients with AML who had an intermediate-risk cytogenetic profile and who also received serial 10-day courses of decitabine.134 Recent data from 2 phase I trials suggest a high response rate after the combination therapy of venetoclax, a BCL2 inhibitor, with either decitabine, azacitidine (ClinicalTrials.gov identifier: NCT02203773),135 or low-dose cytarabine136 in newly diagnosed elderly (≥60 years) AML patients not eligible for intensive chemotherapy, a group in whom a high incidence of TP53 mutations would be suspected. After venetoclax and low-dose cytarabine, the overall response rate was 61% with 54% in patients achieving CR/CRi.136 Venetoclax and low-dose cytarabine were shown to be active across a wide range of cytogenetic mutations and patient profiles (overall response rate: 70% in patients ≥75 years; 52% in secondary AML; 47% in patients with adverse karyotypes; 53% in patients previously treated with hypomethylating agents). The OS was estimated to be 79% at 6 months and 70% at 12 months, with the median not reached.135 Based on these encouraging data, venetoclax received breakthrough designation by the Food & Drug Administration (FDA) in combination with cytarabine for the treatment of elderly patients with treatment-naïve AML not eligible for intensive chemotherapy and a large, international randomized phase III study evaluating azacitidine with or without venetoclax (planned inclusion number: n = 400; ClinicalTrials.gov identifier: NCT02993523) is currently recruiting patients. In addition, a large, international phase III randomized trial of venetoclax in combination with low-dose cytarabine vs low-dose cytarabine alone in treatment-naïve AML patients ineligible for intensive chemotherapy has started recruiting (ClinicalTrials.gov Identifier: NCT03069352). Moreover, venetoclax is currently evaluated at the MD Anderson in a phase Ib/II study in combination with intensive chemotherapy with FLAG-IDA in patients with newly diagnosed or relapsed/refractory AML (ClinicalTrials.gov Identifier: NCT03214562).
8.5 |. FLT3-mutated AML
Activating FLT3 mutations are one of the most frequently affected genetic abnormalities in AML and are present in about 30% of newly diagnosed patients.3 As a member of the type III receptor tyrosine kinase subfamily, including c-KIT, c-FMS, and PDGFR-α/β, it is involved in proliferation and differentiation of myeloid progenitor cells.137 Mutations of the FLT3 gene lead to ligand-independent activation and dysregulation of downstream pathways such as PI3K/AKT, MAPK/ERK, and STAT5.138–140 Clinically, FLT3 mutations are associated with high WBC count and myeloid blast cells in bone marrow and peripheral blood and a more frequent diagnosis of de novo rather than secondary AML.141 In cytogenetically normal AML, FLT3-ITD confers an unfavorable prognosis due to a high relapse rate and very limited options after relapse with conventional salvage regimens, whereas the prognostic impact of point mutations within the tyrosine kinase domain (FLT3-TKDs) remains controversial.141 Nevertheless, FLT3-TKDs can occur after treatment with FLT3 TKIs as a mechanism of resistance, thus implicating an adverse prognosis.142 Besides cytogenetically normal AML, FLT3-ITD is frequently associated with t(6;9)(p23;q34)58–60 as well as with t(15;17)(q22;q12) in APL.3
The incidence of FLT3-ITD is associated with age: Whereas it can only rarely be found in children, its incidence is highest in young adults up to the age of 60 years and declines in the elderly.143 Regarding specific ITD characteristics, the size of these duplications varies widely, typically ranging from 3 to over 100 base pairs (bps) with a median of 48 bps.141 In addition, size and ITD insertion site in the FLT3 gene seem to be correlated in that the more 3’ the insertion site within the FLT3 gene, the longer the ITD.144 The impact of the size on outcome is still unclear with some publications stating no impact on outcome,145,146 whereas one publication found that short ITDs may impart an unfavorable outcome.147 Nevertheless, most publications stated that longer ITDs correlate with lower CR rates and shorter OS and EFS.148–150 In addition, the ITD insertion site within the FLT3 gene has been shown to be an important prognostic factor and was associated with an inferior prognosis.144 About one-third of all FLT3-ITDs occur within the tyrosine kinase domain 1 (TKD1) of the FLT3 gene, in particular in the beta1-sheet.144,151 In cell culture analyses, a prototypic FLT3-ITD with insertion site in the β2-sheet of the TKD1 (FLT3-ITD627E) mediated phosphorylation of FLT3 and STAT5, suggesting that non-JMD FLT3-ITD mutations confer constitutive activation of the receptor.151 Additionally, FLT3-ITD627E induced transformation of hematopoietic 32D cells and led to a lethal myeloproliferative disease in a syngeneic mouse model. Insertions in the beta 1-sheet of TKD1 may introduce a greater instability into the protein structure and may therefore be associated with a pronounced adverse prognosis.151 Besides the insertion site, further prognostic and predictive impact has been shown for the allelic ratio,100,102 which is quantified by GeneScan analysis using DNA fragment analysis. A high allelic ratio is associated with an unfavorable impact on OS and EFS.100,102,144 The prognostic impact of FLT3-ITD is also affected by concurrent mutations, such as NPM1 and DNMT3A. In normal karyotype AML with NPM1 mutation, FLT3-ITDs are present in about 45% of patients.97,99 Recent recommendations from the ELN include a revised version of the risk stratification according to genetics including the FLT3-ITD allelic ratio (Table 1).13 It should be noted, however, that no internationally standardized method of determining the allelic ratio has been established, and it is not routinely reported by many or even most commercial diagnostic laboratories. This is problematic for a practitioner attempting to managing these patients according to these ELN guidelines.
In addition, NUP98/NSD1 has been described to have a further independent prognostic impact in FLT3-ITD-mutated AML.152,153 In 1421 patients from six Children Oncology Group/Southwest Oncology Group trials, NUP98/NSD1 was detected in 15% (37 of 253) of FLT3-ITD and 7% (26 of 367) of cytogenetically normal AMLs. Patients with FLT3-ITD and NUP98/NSD1 (82% of NUP98/NSD1 patients) had a CR rate of 27% as compared to 69% in FLT3-ITD without NUP98/NSD1 (P < 0.001). The corresponding 3-year OS rate was 31% as compared to 48% (P = 0.011), respectively.154 Moreover, WT1 mutations were enriched in patients with FLT3-ITD and NUP98/NSD1, with a prevalence of 31% as compared to 17% in those with FLT3-ITD only (P = 0.047). The authors suggest that additional genetic lesions (ie, WT1+) might further impact response to therapy and outcome of patients harboring NUP98/NSD1 and FLT3-ITD. However, this finding needs to be confirmed in an independent cohort.
Pretherapeutic molecular testing for NPM1 and FLT3 is considered standard of care to determine the best treatment option. Whereas NPM1 has been shown to be a reliable marker for MRD detection with high sensitivity,94–97 the suitability of FLT3-ITD for MRD detection has been questioned. First, FLT3-ITD mutations display substantial heterogeneity in terms of size, number of clones per patient, allelic ratio, and insertion site within the FLT3 gene and second, its proposed instability (reported on about 25% of paired diagnosis-relapse samples) during the course of treatment.
Current methods used to determine FLT3-ITD mutations have limited sensitivity and are not suitable for MRD detection. Newer techniques, such as RT-qPCR with patient-specific primers, aim to improve the sensitivity of FLT3-ITD.155 Nevertheless, this approach has limitations, since each FLT3-ITD mutation needs a clone-specific primer/probe set, which is time-consuming and may not be possible in every case. In addition, direct sequencing may be hampered due to low allelic ratio since the wild-type sequence is competitively amplified. Recently, another PCR-based assay for FLT3-ITD MRD was reported.156,157 This assay employed primers oriented in the opposite direction; hence, amplification occurred only if a FLT3-ITD was present. Again, this approach has limitations since short FLT3-ITDs (less than 30–40 bases) are not detected due to insufficient primer annealing space, which may apply to roughly 25% of all FLT3-ITD cases. Both approaches are therefore not ready to be implemented in clinical routine care. Next-generation sequencing (NGS) is potentially useful158,159 since it can identify clonal populations but generates complex data, which is still expensive and requires considerable expertise to interpret. Additionally, NGS gives only an estimate on the allelic ratio. In patients with a concurrent NPM1 mutation, however, MRD can be assessed by analysis of NPM1-mutated transcripts. In summary, FLT3 mutational testing should be mandatory in all AML patients at diagnosis as well as at relapse for prognostic purposes and for guiding therapeutic decisions. At present, it has little utility for MRD monitoring until different methodologies can be standardized.
Regarding postremission therapy in FLT3-ITD AML, allo-SCT has been shown to improve outcomes particular in patients with a high allelic ratio.160–162 Nevertheless, recent studies indicate that AML patients with NPM1 mutation and low FLT3-ITD allelic ratio may have a more favorable prognosis and should therefore not routinely be assigned to allo-SCT.103,163,164 In contrast, an ITD insertion site in the TKD1 remained an unfavorable prognostic factor regardless of the applied therapy.102
Currently, midostaurin (Rydapt®, Novartis Pharmaceuticals, Inc.) is the first approved TKI in combination with standard intensive chemotherapy for adult patients without age restriction with newly diagnosed FLT3-mutated AML in the United States and Europe. The approval of midostaurin was based on the positive results from the large, international randomized phase III trial.163 The combination of midostaurin with intensive chemotherapy significantly improved OS in younger adults with FLT3-mutated AML translating into a median OS of 74.7 months for the midostaurin arm (range, 31.5 months-not reached) as compared to 25.6 months for the placebo arm (range, 18.6–42.9 months), respectively.163 Interestingly, this improvement was regardless of the FLT3 mutational status (either ITD or TKD) or the FLT3-ITD allelic ratio. Furthermore, patients receiving an allo-SCT in first CR had better outcome if they were treated with midostaurin during induction therapy (P = 0.08), suggesting that the optimal treatment strategy in FLT3-mutated AML would be to move on to allo-SCT early in first CR. Given the remarkable difference in survival after allo-SCT early in first CR in patients treated with midostaurin as compared to those treated with placebo, it is tempting to speculate that the combination of midostaurin with intensive chemotherapy results in deeper remissions.
Nevertheless, a significant proportion of patients within the CALGB 10603/RATIFY trial still relapsed within the first two years, even in the midostaurin arm,163 raising the question as to whether or not TKIs with higher FLT3 selectivity would be more efficient. Currently, various other, more selective FLT3 inhibitors, such as quizartinib,164,165 crenolanib,166 and gilteritinib,167 are in clinical evaluation. Overall, these second-generation inhibitors are significantly more potent and selective with respect to FLT3 inhibition as compared to midostaurin. Quizartinib is an oral selective FLT3 inhibitor currently in phase III development for relapsed/refractory (QuANTUM-R; ClinicalTrials.gov identifier: NCT02668653) and newly diagnosed (QuANTUM-First; ClinicalTrials.gov identifier: NCT02668653) FLT3-ITD AML in the United States, EU, and Japan, and phase II development for relapsed/refractory FLT3-ITD AML in Japan. Recently, the large, randomized international phase III trial QuANTUM-R evaluating quizartinib monotherapy vs salvage chemotherapy in relapsed/refractory FLT3-ITD-positive AML patients (age >18 years) has reached its planned inclusion number of 367 patients.168 Allowed salvage chemotherapy regimens were low-dose cytarabine; mitoxantrone, etoposide, and intermediate-dose cytarabine (MEC); or FLAG-IDA. Up to 2 cycles of MEC or FLAG-IDA were permitted; quizartinib and low-dose cytarabine were given until lack of benefit, unacceptable toxicity, or allo-SCT. Quizartinib significantly prolonged OS as a single agent compared to salvage chemotherapy in patients with relapsed/refractory FLT3-ITD AML (27 weeks vs 20.4 weeks).168 Based on these results, quizartinib was granted breakthrough therapy designation by the FDA on 1 August 2018. Quizartinib has also been granted fast track designation by the FDA for the treatment of relapsed/refractory AML. Additionally, a large, randomized, international phase III trial (QuANTUM-First) tests quizartinib as compared to placebo for newly diagnosed FLT3-ITD AML patients in the United States and Europe (age range: 18–75 years, planned inclusion number: n = 536). The trial design is similar to the CALGB 10603/RATIFY trial: Quizartinib or placebo is administered in combination with standard intensive induction chemotherapy and up to four cycles of consolidation with cytarabine and/or allo-SCT including up to 12 months of maintenance therapy. Recruitment was initiated in 09/2016 and is ongoing.
In addition, gilteritinib has been granted fast track designation by the FDA for the treatment of adult patients with relapsed/refractory FLT3-mutated AML as detected by an FDA-approved test based on the randomized phase III ADMIRAL trial (ClinicalTrials.gov identifier: NCT02421939). The trial design is comparable to the QuANTUM-R trial; currently, the trial has reached its planned inclusion number (n = 371). Both quizartinib and gilteritinib are under FDA review and likely will become available for therapy in relapsed/refractory AML.
9 |. ISOCITRATE DEHYDROGENASE (IDH)
Mutations in IDH1 and IDH2 are detected in about 8% and 12% of patients with AML, respectively.88 IDH1 mutations almost exclusively occur at R132 while IDH2 involve substitutions at R140 or R172.169 These mutations tend to occur early in disease pathogenesis170 and affect the active site of the IDH enzymes that exist in the cytoplasm (IDH1) and the mitochondria (IDH2). Functionally, IDH mutations result in arrest of hematopoietic differentiation due to increased levels of the oncometabolite 2-hydroxyglutarate leading to DNA hypermethylation via inhibition of histone demethylation.169,171,172 IDH mutations are associated with higher platelet counts,173,174 lower median WBC counts,104 intermediate-risk, or normal karyotype cytogenetics, and less frequently occur in therapy-related AML.173,174 Genetically, IDH mutations are associated with NPM1 mutations,105 but less frequently co-occur with TET2 or WT1 mutations, presumably because all three classes of mutations affect DNA methylation.2,175
Regarding outcome in IDH-mutated AML, data are conflicting. For instance, in AML exhibiting the genotype mutated NPM1 with unmutated FLT3-ITD two reports from cooperative study groups showed a negative impact of cooperating IDH1/2 mutations on relapse-free survival and OS.104,105 In contrast, Patel et al2 reported on a favorable impact of the genotype mutated NPM1 with unmutated FLT3-ITD only if cooperating IDH1/2 mutations were present. The effects on survival are likely distinct for each of the IDH mutations, with the presence or absence of other mutations also affecting outcomes. Additionally, such opposed effects of genotypes on outcome highlight statistical shortcomings of retrospective molecular studies.
The prognosis and treatment of IDH-mutated AML may also be changed by IDH inhibitors, which have been associated with responses in the relapsed setting and are now being added to induction chemotherapy in upcoming trails. Enasidenib, a potent IDH2 inhibitor (formerly known as AG-221/CC-90007), received regulatory approval in the United States on 1 August 2017 for the treatment of relapsed/refractory AML with an IDH2 mutation as detected by an FDA-approved test. The recommended dose of enasidenib is 100 mg orally once daily until disease progression or unacceptable toxicity. Analysis of paired diagnosis/relapse samples did not identify second-site mutations in IDH2 at relapse.176 Instead, relapse arose by clonal evolution or selection of terminal or ancestral clones, thus highlighting multiple bypass pathways that could potentially be targeted to restore differentiation arrest.
In addition, ivosidenib (Tibsovo®; formerly known as AG120-C-001) has received FDA approval on 20 July 2018 for the treatment of adult patients with relapsed/refractory IDH1-mutated AML. The approval was based on an open-label, single-arm, multicenter clinical trial (ClinicalTrials.gov identifier: NCT02074839) that included 179 adult patients with relapsed or refractory IDH1-mutated AML.177 Ivosidenib was given orally at a starting dose of 500 mg daily until disease progression, unacceptable toxicity, or allo-SCT. The rate of CR or CR with incomplete hematologic recovery was 30.4%, and the overall response rate was 41.6%. The median treatment and response duration were 4.1 months and 8.2 months, respectively.177 Both inhibitors are currently evaluated in several clinical trials in newly diagnosed as well as relapsed/refractory AML, as single agent or in combination with chemotherapy (ClinicalTrials.gov identifier: NCT02074839, NCT02073994, NCT02632708, NCT02677922). Other IDH1178 and combined IDH1/2 inhibitors (such as AG-881, ClinicalTrials.gov identifier: NCT02492737) have also entered clinical development. Finally, Debarri et al179 have evaluated MRD monitoring of IDH1/2 mutations by NGS in 31 AML patients. Within their analysis, IDH1/2 mutations were reliable MRD markers that allowed the prediction of relapse in the majority of patients.
10 |. LYSINE-SPECIFIC METHYLTRANSFERASE 2A (KMT2A)
Chromosomal rearrangement of the KMT2A (formerly known as mixed lineage leukemia, MLL) gene at 11q23 has been reported in approximately 10% of acute leukemias.180 This rearrangement results in AMLs with predominantly monocytic or myelomonocytic phenotypes. KMT2A rearrangement is also associated with therapy-related myeloid neoplasm, specifically following topoisomerase II inhibitors.181 KMT2A encodes a histone methyltransferase, which regulates homeobox genes affecting hematopoiesis.182 A subtype is characterized by internal partial tandem duplication in the KMT2A gene (KMT2A-PTD). KMT2A-PTD often occurs in elderly patients and consists of 3%-5% of de novo AML.183,184 KMT2A-PTD is generally believed to act as an oncogenic driver by modulating expression of HOX genes. However, mice carrying KMT2A-PTD alone do not develop spontaneous leukemia,185 unless they are crossed with those harboring another major leukemogenic driver (eg, FLT3-ITD),186 suggesting that by itself KMT2A-PTD is not sufficient to transform hematopoietic cells. Clonality analysis suggested that KMT2A-PTD is acquired after initiating mutations (ie, IDH1/2, DNMT3A, TET2, and U2AF1), but prior to proliferation mutations (such as FLT3 and RAS).187 KMT2A-PTD seems to be mutually exclusive with DNMT3A2 and NPM1 mutations.187 However, 25% of patients harbor a FLT3-ITD, which may contribute to the poor prognosis.188 Outcome of patients with acute leukemia and a translocation or PTD of the KMT2A gene is poor. In a study of 1897 patients with AML treated within German AML Cooperative Group trials between 1992 and 1999, 2.8% of patients were found to have a rearrangement involving 11q23 and OS rate at 3 years was 12.5%.189
Elegant in vitro and in vivo experiments demonstrated that a key mediator of KMT2A-rearranged leukemia is the histone methyltransferase DOT1L.190–194 Similarly, preclinical studies of DOT1L inhibition in acute leukemia associated with translocations involving the KMT2A gene have shown remarkable effectiveness.195,196 Translating these results to patients, however, has been more difficult. Inhibition of DOT1L with the small molecule inhibitor EPZ-5676 resulted in a CR in only two of 34 patients with a KMT2A rearrangement or partial tandem duplication.197 In cell culture analysis using a DNMT3A-mutated cell line, inhibition of DOT1L resulted in inhibition of proliferation, induction of apoptosis, and terminal differentiation.198 Again, the DOT1L inhibitor EPZ5676 was effective in a xenograft model as well as primary patient samples with DNMT3A-mutated AML, suggesting that DOT1L could be a therapeutic target.198
11 |. DNMT3A MUTATIONS
Awareness of the complexity of the leukemic genome has further been highlighted by the discovery of mutations in genes important for epigenetic regulation of gene transcription. DNA methyltransferase 3 alpha (DNMT3A) belongs to the family of DNA methyltransferase enzymes that catalyzes the addition of a methyl group to the cytosine of CpG dinucleotides, thereby regulating gene transcription. DNMT3A mutations can be found in 15%-25% of AML patients, particularly in AML with normal cytogenetics,5,199–201 and are thought to be a “founder” mutation since they are present in early preleukemic hematopoietic stem cells.202 Approximately 60% of all DNMT3A mutations affect residue R882,5,199 and seem to be age-dependent with a higher incidence in elderly patients.203 Several studies evaluated the stability of DNMT3A mutations in paired diagnosis and relapse material.93 In the largest analyses, Hou et al204 studied sequentially 316 samples from 138 patients, including 35 patients with distinct DNMT3A mutations and 103 patients without mutations at diagnosis. At relapse, all DNMT3A-mutated patients who had available samples for serial study regained the same mutations, whereas all 103 patients without DNMT3A mutation at diagnosis remained DNMT3A negative at relapse.204 Due to its high stability, the authors claimed DNMT3A mutations to be a potential marker for MRD monitoring. Ploen et al201 who developed a multiplex allele-specific quantitative PCR assay for the sensitive detection of DNMT3A mutations affecting residue R882 questioned the suitability as a MRD marker. Analysis of DNA from 298 diagnostic AML samples revealed DNMT3A mutations in 45 AML patients (15%); the mutation was stable in 12 of 13 patients presenting with relapse or secondary myelodysplastic syndrome, but was also found in remission samples from 14 patients (at allele frequencies of <1–50%) up to 8 years after initial AML diagnosis, despite the loss of all other molecular AML markers.201 Nine of the 14 patients relapsed within a median time of 9.5 months (range 4–104 months); the five remaining patients were reported to be in continuous CR and had a long-term survival at a median time of 53 months (range 34–100 months). Due to its long-term persistence without relapse, the authors question DNMT3A mutations as a suitable biomarker for AML patient management.201
12 |. CONCLUSIONS
Progress in deciphering the molecular pathogenesis of pediatric and adult AML205 and the identification of the genetic determinants of response to treatment have been impressive, and translation of these findings into the clinical decision making has been increasing in recent years. The availability of the molecular profile enables a targeted-based treatment. Thus, evaluation of the genetic profile at diagnosis, but also at relapse, is of utmost importance. Besides the achievement of a morphological remission as a prerequisite of cure, the MRD level gives further insights into the remission status and determines kinetics of disease response as well as enables to detect an impending hematologic relapse. However, non-uniform definitions of MRD-positive/negative results, interobserver variability using flow cytometric MRD detection, sampling error from marrow to blood, and lack of uniformly agreed standardization upon molecular targets, methods, and sampling timing have hampered the transition into routine clinical practice. Thus, there is a high need for dedicated diagnostic standards to avoid wrong, ineffective, and expensive targeted treatment approaches. Therefore, a common international attempt to move forward standardization of genetic diagnostics including cryptic fusions (eg, CBF-GLIS2, NUP98/NSD1) as well as immunophenotypic abnormalities (eg, RAM phenotype) leading to the recommendation of standardized assays is mandatory for future use of targeted treatment approaches.
ACKNOWLEDGEMENTS
S.K. gratefully acknowledges to be supported by the Olympia-Morata program from the Medical Faculty of the Heidelberg University. M.J.L. is supported by a grant from the NCI (NCI Leukemia SPORE P50 CA100632).
Footnotes
CONFLICT OF INTEREST
S.K. has served as a consultant for Novartis. M.J.L. receives research funding from Novartis and Astellas. M.J.L. serves as a consultant for Novartis, Daiichi-Sankyo, Astellas, and Arog.
REFERENCES
- 1.Döhner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid leukemia. N Engl J Med. 2015;373(12):1136–1152. [DOI] [PubMed] [Google Scholar]
- 2.Patel JP, Gönen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Cancer Genome Atlas Research Network, Ley TJ, Miller C, Ding L, et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Mardis ER, Ding L, Dooling DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009;361(11):1058–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–2433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Swerdlow SH, Campo E, Harris NL, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, revised 4th edn Geneva, Switzerland: WHO Press; 2017. [Google Scholar]
- 7.Grimwade D, Freeman SD. Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for “prime time”? Blood. 2014;124(23):3345–3355. [DOI] [PubMed] [Google Scholar]
- 8.Baccarani M, Deininger MW, Rosti G, et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122(6):872–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Vora A, Goulden N, Wade R, et al. Treatment reduction for children and young adults with low-risk acute lymphoblastic leukaemia defined by minimal residual disease (UKALL 2003): a randomised controlled trial. Lancet Oncol. 2013;14(3):199–209. [DOI] [PubMed] [Google Scholar]
- 10.Vora A, Goulden N, Mitchell C, et al. Augmented post-remission therapy for a minimal residual disease-defined high-risk subgroup of children and young people with clinical standard-risk and intermediate-risk acute lymphoblastic leukaemia (UKALL 2003): a randomised controlled trial. Lancet Oncol. 2014;15(8):809–818. [DOI] [PubMed] [Google Scholar]
- 11.Brüggemann M, Raff T, Kneba M. Has MRD monitoring su-perseded other prognostic factors in adult ALL? Blood. 2012;120(23):4470–4481. [DOI] [PubMed] [Google Scholar]
- 12.Brüggemann M, Raff T, Flohr T, et al. Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood. 2006;107(3):1116–1123. [DOI] [PubMed] [Google Scholar]
- 13.Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Löwenberg B, Pabst T, Vellenga E, et al. Cytarabine dose for acute myeloid leukemia. N Engl J Med. 2011;364(11):1027–1036. [DOI] [PubMed] [Google Scholar]
- 15.Schlenk RF, Benner A, Krauter J, et al. Individual patient databased meta-analysis of patients aged 16 to 60 years with core binding factor acute myeloid leukemia: a survey of the German Acute Myeloid Leukemia Intergroup. J Clin Oncol. 2004;22(18):3741–3750. [DOI] [PubMed] [Google Scholar]
- 16.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(18):4173–4179. [PubMed] [Google Scholar]
- 17.Byrd JC, Dodge RK, Carroll A, et al. Patients with t(8;21)(q22;q22) and acute myeloid leukemia have superior failure-free and overall survival when repetitive cycles of high-dose cytarabine are administered. J Clin Oncol. 1999;17:3767–3775. [DOI] [PubMed] [Google Scholar]
- 18.Byrd JC, Ruppert AS, Mrozek K, et al. Repetitive cycles of high-dose cytarabine benefit patients with acute myeloid leukemia and inv(16)(p13q22) or t(16;16)(p13;q22): results from CALGB 8461. J Clin Oncol. 2004;22:1087–1094. [DOI] [PubMed] [Google Scholar]
- 19.Cornelissen JJ, van Putten WL, Verdonck LF, 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. 2007;109(9):3658–3666. [DOI] [PubMed] [Google Scholar]
- 20.Yin JA, O’Brien MA, Hills RK, Daly SB, Wheatley K, Burnett AK. 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(14):2826–2835. [DOI] [PubMed] [Google Scholar]
- 21.Hoyos M, Nomdedeu JF, Esteve J, et al. Core binding factor acute myeloid leukemia: the impact of age, leukocyte count, molecular findings, and minimal residual disease. Eur J Haematol. 2013;91(3):209–218. [DOI] [PubMed] [Google Scholar]
- 22.Zhu HH, Zhang XH, Qin YZ, et al. MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: results from the AML05 multicenter trial. Blood. 2013;121(20):4056–4062. [DOI] [PubMed] [Google Scholar]
- 23.Koreth J, Schlenk R, Kopecky KJ, et al. Allogeneic stem cell transplantation for acute myeloid leukemia in first complete remission: systematic review and meta-analysis of prospective clinical trials. JAMA. 2009;301(22):2349–2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cornelissen JJ, Gratwohl A, Schlenk RF, et al. The European LeukemiaNet AML Working Party consensus statement on allogeneic HSCT for patients with AML in remission: an integrated-risk adapted approach. Nat Rev Clin Oncol. 2012;9(10):579–590. [DOI] [PubMed] [Google Scholar]
- 25.Burnett AK, Russell NH, Hills RK, et al. Optimization of chemotherapy for younger patients with acute myeloid leukemia: results of the medical research council AML15 trial. J Clin Oncol. 2013;31(27):3360–3368. [DOI] [PubMed] [Google Scholar]
- 26.Petersdorf SH, Kopecky KJ, Slovak M, et al. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. Blood. 2013;121:4854–4860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Burnett AK, Russell NH, Hills RK, et al. Addition of gemtuzumab ozogamicin to induction chemotherapy improves survival in older patients with acute myeloid leukemia. J Clin Oncol. 2012;30(32):3924–3931. [DOI] [PubMed] [Google Scholar]
- 28.Castaigne S, Pautas C, Terré C, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. The Lancet. 2012;379(9825):1508–1516. [DOI] [PubMed] [Google Scholar]
- 29.Prebet T, Bertoli S, Delaunay J, et al. Anthracycline dose intensification improves molecular response and outcome of patients treated for core binding factor acute myeloid leukemia. Haematologica. 2014;99(10):e185–e187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Borthakur G, Cortes JE, Estey EE, et al. Gemtuzumab ozogamicin with fludarabine, cytarabine, and granulocyte colony stimulating factor (FLAG-GO) as front-line regimen in patients with core binding factor acute myelogenous leukemia. Am J Hematol. 2014;89(10):964–968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Schlenk RF, Frech P, Weber D, et al. Impact of pretreatment characteristics and salvage strategy on outcome in patients with relapsed acute myeloid leukemia. Leukemia. 2017;31(5):1217–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Nguyen S, Leblanc T, Fenaux P, et al. A white blood cell index as the main prognostic factor in t(8;21) acute myeloid leukemia (AML): a survey of 161 cases from the French AML Intergroup. Blood. 2002;99(10):3517–3523. [DOI] [PubMed] [Google Scholar]
- 33.Schnittger S, Kohl TM, Haferlach T, et al. KITD816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006;107(5):1791–1799. [DOI] [PubMed] [Google Scholar]
- 34.Cairoli R, Beghini A, Grillo G, et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study. Blood. 2006;107(9):3463–3468. [DOI] [PubMed] [Google Scholar]
- 35.Boissel N, Leroy H, Brethon B, et al. Acute Leukemia French Association (ALFA); Leucemies Aiguës Myeloblastiques de l’Enfant (LAME) Cooperative Groups. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML). Leukemia. 2006;20(6):965–970. [DOI] [PubMed] [Google Scholar]
- 36.Paschka P, Marcucci G, Ruppert AS, et al. Cancer and Leukemia Group B. 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(24):3904–3911. [DOI] [PubMed] [Google Scholar]
- 37.Paschka P, Du J, Schlenk RF, et al. Secondary genetic lesions in acute myeloid leukemia with inv(16) or t(16;16): a study of the German-Austrian AML Study Group (AMLSG). Blood. 2013;121(1): 170–177. [DOI] [PubMed] [Google Scholar]
- 38.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(9):1891–1901. [DOI] [PubMed] [Google Scholar]
- 39.Jourdan E, Boissel N, Chevret S, et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood. 2013;121(12):2213–2223. [DOI] [PubMed] [Google Scholar]
- 40.Boddu P, Gurguis C, Cortes JE, et al. Response kinetics and factors predicting survival among patients with core binding factor (CBF) acute myeloid leukemia (AML). Blood. 2017;130:27. [Google Scholar]
- 41.Grimwade D, Jovanovic JV, Hills RK. Can we say farewell to monitoring minimal residual disease in acute promyelocytic leukaemia? Best Pract Res Clin Haematol. 2014;27(1):53–61. [DOI] [PubMed] [Google Scholar]
- 42.National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Acute Myeloid Leukemia. Version 2.2014. http://www.nccn.org
- 43.Kayser S, Schlenk RF, Platzbecker U. Management of patients with acute promyelocytic leukemia. Leukemia. 2018;32(6):1277–1294. [DOI] [PubMed] [Google Scholar]
- 44.De Botton S, Chevret S, Sanz M, et al. Additional chromosomal abnormalities in patients with acute promyelocytic leukaemia (APL) do not confer poor prognosis: results of APL 93 trial. Br J Haematol. 2000;111(3):801–806. [DOI] [PubMed] [Google Scholar]
- 45.Lou Y, Suo S, Tong H, et al. Characteristics and prognosis analysis of additional chromosome abnormalities in newly diagnosed acute promyelocytic leukemia treated with arsenic trioxide as the front-line therapy. Leuk Res. 2013;37(11):1451–1456. [DOI] [PubMed] [Google Scholar]
- 46.Cervera J, Montesinos P, Hernández-Rivas JM, et al. Additional chromosome abnormalities in patients with acute promyelocytic leukemia treated with all-trans retinoic acid and chemotherapy. Haematologica. 2010;95(3):424–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pantic M, Novak A, Marisavljevic D, et al. Additional chromosome aberrations in acute promyelocytic leukemia: characteristics and prognostic influence. Med Oncol. 2000;17(4):307–313. [DOI] [PubMed] [Google Scholar]
- 48.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(7):1523–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cicconi L, Divona M, Ciardi C, et al. PML-RARα kinetics and impact of FLT3-ITD mutations in newly diagnosed acute promyelocytic leukaemia treated with ATRA and ATO or ATRA and chemotherapy. Leukemia. 2016;30(10):1987–1992. [DOI] [PubMed] [Google Scholar]
- 50.Kiyoi H, Naoe T, Yokota S, et al. Internal tandem duplication of FLT3 associated with leukocytosis in acute promyelocytic leukemia. Leukemia Study Group of the Ministry of Health and Welfare (Kohseisho). Leukemia. 1997;11:1447–1452. [DOI] [PubMed] [Google Scholar]
- 51.Noguera NI, Breccia M, Divona M, et al. Alterations of the FLT3 gene in acute promyelocytic leukemia: association with diagnostic characteristics and analysis of clinical outcome in patients treated with the Italian AIDA protocol. Leukemia. 2002;16:2185–2189. [DOI] [PubMed] [Google Scholar]
- 52.Shih LY, Kuo MC, Liang DC, et al. Internal tandem duplication and Asp835 mutations of the FMS-like tyrosine kinase 3 (FLT3) gene in acute promyelocytic leukemia. Cancer. 2003;98:1206–1216. [DOI] [PubMed] [Google Scholar]
- 53.Rowley JD, Potter D. Chromosomal banding patterns in acute non-lymphocytic leukemia. Blood. 1976;47(5):705–721. [PubMed] [Google Scholar]
- 54.von Lindern M, Fornerod M, van Baal S, et al. The translocation (6;9), associated with a specific subtype of acute myeloid leukemia, results in the fusion of two genes, dek and can, and the expression of a chimeric, leukemia-specific dek-can mRNA. Mol Cell Biol. 1992;12(4):1687–1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Scandura JM, Boccuni P, Cammenga J, Nimer SD. Transcription factor fusions in acute leukemia: variations on a theme. Oncogene. 2002;21(21):3422–3444. [DOI] [PubMed] [Google Scholar]
- 56.Ageberg M, Drott K, Olofsson T, Gullberg U, Lindmark A. Identification of a novel and myeloid specific role of the leukemia-associated fusion protein DEK-NUP214 leading to increased protein synthesis. Genes Chromosom Cancer. 2008;47(4):276–287. [DOI] [PubMed] [Google Scholar]
- 57.Boer J, Bonten-Surtel J, Grosveld G. Overexpression of the nucleoporin CAN/NUP214 induces growth arrest, nucleocytoplasmic transport defects, and apoptosis. Mol Cell Biol. 1998;18(3):1236–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Slovak ML, Gundacker H, Bloomfield CD, et al. A retrospective study of 69 patients with t(6;9)(p23;q34) AML emphasizes the need for a prospective, multicenter initiative for rare ‘poor prognosis’ myeloid malignancies. Leukemia. 2006;20(7):1295–1297. [DOI] [PubMed] [Google Scholar]
- 59.Gupta M, Ashok Kumar J, Sitaram U, et al. The t(6;9)(p22;q34) in myeloid neoplasms: a retrospective study of 16 cases. Cancer Genet Cytogenet. 2010;203(2):297–302. [DOI] [PubMed] [Google Scholar]
- 60.Sandahl JD, Coenen EA, Forestier E, et al. t(6;9)(p22;q34)/DEK-NUP214-rearranged pediatric myeloid leukemia: an international study of 62 patients. Haematologica. 2014;99(5):865–872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Tarlock K, Alonzo TA, Moraleda PP, et al. Acute myeloid leukaemia (AML) with t(6;9)(p23;q34) is associated with poor outcome in childhood AML regardless of FLT3-ITD status: a report from the Children’s Oncology Group. Br J Haematol. 2014;166(2):254–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Grimwade D, Hills RK, Moorman AV, et al. 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(3):354–365. [DOI] [PubMed] [Google Scholar]
- 63.Thiede C, Bloomfield CD, Lo Coco F, et al. The high prevalence of FLT3-ITD mutations is associated with the poor outcome in adult patients with t(6;9)(p23;q34) positive AML - results of an international meta-analysis. Blood. 2007;110:761. [Google Scholar]
- 64.Ishiyama K, Takami A, Kanda Y, et al. Allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia with t(6;9) (p23;q34) dramatically improves the patient prognosis: a matched-pair analysis. Leukemia. 2012;26(3):461–464. [DOI] [PubMed] [Google Scholar]
- 65.Kayser S, Hills RK, Luskin MR, et al. Characteristics and Outcome of Patients with Acute Myeloid Leukemia and t(6;9)(p22;q34). Blood. 2017;130:3906. [Google Scholar]
- 66.Lugthart S, Gröschel S, Beverloo HB, et al. Clinical, molecular, and prognostic significance of WHO type inv(3)(q21q26.2)/t(3;3) (q21;q26.2) and various other 3q abnormalities in acute myeloid leukemia. J Clin Oncol. 2010;28(24):3890–3898. [DOI] [PubMed] [Google Scholar]
- 67.Gröschel S, Sanders MA, Hoogenboezem R, et al. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell. 2014;157(2):369–381. [DOI] [PubMed] [Google Scholar]
- 68.Gröschel S, Sanders MA, Hoogenboezem R, et al. Mutational spectrum of myeloid malignancies with inv(3)/t(3;3) reveals a predominant involvement of RAS/RTK signaling pathways. Blood. 2015;125(1):133–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Pagano L, Pulsoni A, Vignetti M, et al. Acute megakaryoblastic leukemia: experience of GIMEMA trials. Leukemia. 2002;16(9):1622–1626. [DOI] [PubMed] [Google Scholar]
- 70.Athale UH, Razzouk BI, Raimondi SC, et al. Biology and outcome of childhood acute megakaryoblastic leukemia: a single institution’s experience. Blood. 2001;97(12):3727–3732. [DOI] [PubMed] [Google Scholar]
- 71.Bain BJ, Chakravorty S, Ancliff P. Congenital acute megakaryoblastic leukemia. Am J Hematol. 2015;90(10):963. [DOI] [PubMed] [Google Scholar]
- 72.Mercher T, Coniat MB, Monni R, et al. Involvement of a human gene related to the Drosophila spen gene in the recurrent t(1;22) translocation of acute megakaryocytic leukemia. Proc Natl Acad Sci USA. 2001;98(10):5776–5779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bernstein J, Dastugue N, Haas OA, et al. Nineteen cases of the t(1;22)(p13;q13) acute megakaryblastic leukaemia of infants/children and a review of 39 cases: report from a t(1;22) study group. Leukemia. 2000;14(1):216–218. [DOI] [PubMed] [Google Scholar]
- 74.Duchayne E, Fenneteau O, Pages MP, et al. Acute megakaryoblastic leukaemia: a national clinical and biological study of 53 adult and childhood cases by the Groupe Français d’Hématologie Cellulaire (GFHC). Leuk Lymphoma. 2003;44(1):49–58. [DOI] [PubMed] [Google Scholar]
- 75.Carroll A, Civin C, Schneider N, et al. The t(1;22) (p13;q13) is nonrandom and restricted to infants with acute megakaryoblastic leukemia: a Pediatric Oncology Group Study. Blood. 1991;78(3):748–752. [PubMed] [Google Scholar]
- 76.O’Brien MM, Cao X, Pounds S, et al. Prognostic features in acute megakaryoblastic leukemia in children without Down syndrome: a report from the AML02 multicenter trial and the Children’s Oncology Group Study POG 9421. Leukemia. 2013;27(3):731–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.de Rooij JD, Hollink IH, Arentsen-Peters ST, et al. NUP98/JARID1A is a novel recurrent abnormality in pediatric acute megakaryoblastic leukemia with a distinct HOX gene expression pattern. Leukemia. 2013;27(12):2280–2288. [DOI] [PubMed] [Google Scholar]
- 78.Inaba H, Zhou Y, Abla O, et al. Heterogeneous cytogenetic subgroups and outcomes in childhood acute megakaryoblastic leukemia: a retrospective international study. Blood. 2015;126(13):1575–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Schweitzer J, Zimmermann M, Rasche M, et al. Improved outcome of pediatric patients with acute megakaryoblastic leukemia in the AML-BFM 04 trial. Ann Hematol. 2015;94(8):1327–1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.de Rooij JD, Branstetter C, Ma J, et al. Pediatric non-Down syndrome acute megakaryoblastic leukemia is characterized by distinct genomic subsets with varying outcomes. Nat Genet. 2017;49(3):451–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Creutzig U, Reinhardt D, Diekamp S, Dworzak M, Stary J, Zimmermann M. AML patients with Down syndrome have a high cure rate with AML-BFM therapy with reduced dose intensity. Leukemia. 2005;19(8):1355–1360. [DOI] [PubMed] [Google Scholar]
- 82.Konoplev S, Yin CC, Kornblau SM, et al. Molecular characterization of de novo Philadelphia chromosome-positive acute myeloid leukemia. Leuk Lymphoma. 2013;54(1):138–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Paietta E, Racevskis J, Bennett JM, et al. Biologic heterogeneity in Philadelphia chromosome-positive acute leukemia with myeloid morphology: the Eastern Cooperative Oncology Group experience. Leukemia. 1998;12(12):1881–1885. [DOI] [PubMed] [Google Scholar]
- 84.Neuendorff NR, Burmeister T, Dörken B, Westermann J. BCR-ABL-positive acute myeloid leukemia: a new entity? Analysis of clinical and molecular features. Ann Hematol. 2016;95(8):1211–1221. [DOI] [PubMed] [Google Scholar]
- 85.Pompetti F, Spadano A, Sau A, et al. Long-term remission in BCR/ABL-positive AML-M6 patient treated with Imatinib Mesylate. Leuk Res. 2007;31(4):563–567. [DOI] [PubMed] [Google Scholar]
- 86.Lazarevic V, Golovleva I, Nygren I, Wahlin A. Induction chemotherapy and post-remission imatinib therapy for de Novo BCR-ABL-positive AML. Am J Hematol. 2006;81(6):470–471. [DOI] [PubMed] [Google Scholar]
- 87.Kondo T, Tasaka T, Sano F, et al. Philadelphia chromosome-positive acute myeloid leukemia (Ph + AML) treated with imatinib mesylate (IM): a report with IM plasma concentration and bcr-abl transcripts. Leuk Res. 2009;33(9):e137–e138. [DOI] [PubMed] [Google Scholar]
- 88.Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Mrozek 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(36):4515–4523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Schlenk RF, Döhner K, Kneba M, 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. 2009;94(1):54–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.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(4):596–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005;352(3):254–266. [DOI] [PubMed] [Google Scholar]
- 93.Krönke J, Bullinger L, Teleanu V, et al. Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia. Blood. 2013;122(1):100–108. [DOI] [PubMed] [Google Scholar]
- 94.Falini B, Sportoletti P, Martelli MP. Acute myeloid leukemia with mutated NPM1: diagnosis, prognosis and therapeutic perspectives. Curr Opin Oncol. 2009;21(6):573–581. [DOI] [PubMed] [Google Scholar]
- 95.Gorello P, Cazzaniga G, Alberti F, et al. Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations. Leukemia. 2006;20(6):1103–1108. [DOI] [PubMed] [Google Scholar]
- 96.Schnittger S, Kern W, Tschulik C, et al. Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML. Blood. 2009;114(11):2220–2231. [DOI] [PubMed] [Google Scholar]
- 97.Thiede C, Koch S, Creutzig E, et al. Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML). Blood. 2006;107:4011–4020. [DOI] [PubMed] [Google Scholar]
- 98.Döhner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood. 2005;106:3740–3746. [DOI] [PubMed] [Google Scholar]
- 99.Schlenk RF, Döhner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358(18):1909–1918. [DOI] [PubMed] [Google Scholar]
- 100.Pratcorona M, Brunet S, Nomdedéu J, et al. Favorable outcome of patients with acute myeloid leukemia harboring a lowallelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy. Blood. 2013;121:2734–2738. [DOI] [PubMed] [Google Scholar]
- 101.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;111:2776–2784. [DOI] [PubMed] [Google Scholar]
- 102.Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124:3441–3449. [DOI] [PubMed] [Google Scholar]
- 103.Schetelig J, Röllig C, Kayser S, et al. Validation of the ELN 2017 classification for AML with intermediate risk cytogenetics with or without NPM1-mutations and high or low ratio FLT3-ITD s. Blood. 2017;130:2694. [Google Scholar]
- 104.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(22):3636–3643. [DOI] [PubMed] [Google Scholar]
- 105.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(14):2348–2355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.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(19):2709–2716. [DOI] [PubMed] [Google Scholar]
- 107.Hubmann M, Köhnke T, Hoster E, et al. Molecular response assessment by quantitative real-time polymerase chain reaction after induction therapy in NPM1-mutated patients identifies those at high risk of relapse. Haematologica. 2014;99(8):1317–1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Lambert J, Lambert J, Nibourel O, et al. MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin. Oncotarget. 2014;5(15):6280–6288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Shayegi N, Kramer M, Bornhäuser M, et al. The level of residual disease based on mutant NPM1 is an independent prognostic factor for relapse and survival in AML. Blood. 2013;122(1):83–92. [DOI] [PubMed] [Google Scholar]
- 110.Ivey A, Hills RK, Simpson MA, et al. Assessment of minimal residual disease in standard-risk AML. N Engl J Med. 2016;374(5):422–433. [DOI] [PubMed] [Google Scholar]
- 111.Kayser S, Benner A, Thiede C, et al. Pretransplant NPM1 MRD levels predict outcome after allogeneic hematopoietic stem cell transplantation in patients with acute myeloid leukemia. Blood Cancer J. 2016;6(7):e449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Balsat M, Renneville A, Thomas X, et al. Postinduction minimal residual disease predicts outcome and benefit from allogeneic stem cell transplantation in acute myeloid leukemia with NPM1 mutation: a study by the Acute Leukemia French Association Group. J Clin Oncol. 2017;35(2):185–193. [DOI] [PubMed] [Google Scholar]
- 113.Hofmann S, Götz M, Schneider V, et al. Donor lymphocyte infusion induces polyspecific CD8(+) T-cell responses with concurrent molecular remission in acute myeloid leukemia with NPM1 mutation. J Clin Oncol. 2013;31:e44–e47. [DOI] [PubMed] [Google Scholar]
- 114.Rettinger E, Willasch AM, Kreyenberg H, et al. Preemptive immunotherapy in childhood acute myeloid leukemia for patients showing evidence of mixed chimerism after allogeneic stem cell transplantation. Blood. 2011;118:5681–5688. [DOI] [PubMed] [Google Scholar]
- 115.Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia. 2012;26:381–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Pabst T, Mueller BU, Zhang P, et al. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (C/EBPalpha), in acute myeloid leukemia. Nat Genet. 2001;27(3):263–270. [DOI] [PubMed] [Google Scholar]
- 117.Fröhling S, Schlenk RF, Stolze I, et al. CEBPA mutations in younger adults with acute myeloid leukemia and normal cytogenetics: prognostic relevance and analysis of cooperating mutations. J Clin Oncol. 2004;22(4):624–633. [DOI] [PubMed] [Google Scholar]
- 118.Wouters BJ, Lowenberg B, Erpelinck-Verschueren CA, van Putten WL, Valk PJ, Delwel R. 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(13):3088–3091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Dufour A, Schneider F, Metzeler KH, 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. 2010;28(4):570–577. [DOI] [PubMed] [Google Scholar]
- 120.Green CL, Koo KK, Hills RK, Burnett AK, Linch DC, Gale RE. Prognostic significance of CEBPA mutations in a large cohort of younger adult patients with acute myeloid leukemia: impact of double CEBPA mutations and the interaction with FLT3 and NPM1 mutations. J Clin Oncol. 2010;28(16):2739–2747. [DOI] [PubMed] [Google Scholar]
- 121.Taskesen E, Bullinger L, Corbacioglu A, 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. 2011;117(8):2469–2475. [DOI] [PubMed] [Google Scholar]
- 122.Grossmann V, Haferlach C, Nadarajah N, et al. CEBPA double-mutated acute myeloid leukaemia harbours concomitant molecular mutations in 76.8% of cases with TET2 and GATA2 alterations impacting prognosis. Br J Haematol. 2013;161(5):649–658. [DOI] [PubMed] [Google Scholar]
- 123.Schlenk RF, Taskesen E, van Norden Y, et al. The value of allogeneic and autologous hematopoietic stem cell transplantation in prognostically favorable acute myeloid leukemia with double mutant CEBPA. Blood. 2013;122(9):1576–1582. [DOI] [PubMed] [Google Scholar]
- 124.Okuda T, van Deursen J, Hiebert SW, Grosveld G, Downing JR. AML1, the target of multiple chromosomal translocations in human leukemia, is essential for normal fetal liver hematopoiesis. Cell. 1996;84(2):321–330. [DOI] [PubMed] [Google Scholar]
- 125.Tang JL, Hou HA, Chen CY, et al. AML1/RUNX1 mutations in 470 adult patients with de novo acute myeloid leukemia: prognostic implication and interaction with other gene alterations. Blood. 2009;114(26):5352–5361. [DOI] [PubMed] [Google Scholar]
- 126.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(25):3109–3118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Gaidzik VI, Bullinger L, Schlenk RF, et al. RUNX1 mutations in acute myeloid leukemia: results from a comprehensive genetic and clinical analysis from the AML study group. J Clin Oncol. 2011;29(10):1364–1372. [DOI] [PubMed] [Google Scholar]
- 128.Michaud J, Wu F, Osato M, et al. In vitro analyses of known and novel RUNX1/AML1 mutations in dominant familial platelet disorder with predisposition to acute myelogenous leukemia: implications for mechanisms of pathogenesis. Blood. 2002;99:1364–1372. [DOI] [PubMed] [Google Scholar]
- 129.Rotter V, Aloni-Grinstein R, Schwartz D, et al. Does wild-type p53 play a role in normal cell differentiation? Semin Cancer Biol. 1994;5(3):229–236. [PubMed] [Google Scholar]
- 130.Grossmann V, Schnittger S, Kohlmann A, et al. A novel hierarchical prognostic model of AML solely based on molecular mutations. Blood. 2012;120(15):2963–2972. [DOI] [PubMed] [Google Scholar]
- 131.Fenaux P, Preudhomme C, Quiquandon I, et al. Mutations of the P53 gene in acute myeloid leukaemia. Br J Haematol. 1992;80(2):178–183. [DOI] [PubMed] [Google Scholar]
- 132.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(9):2114–2121. [DOI] [PubMed] [Google Scholar]
- 133.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(7540):552–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Welch JS, Petti AA, Miller CA, et al. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med. 2016;375(21):2023–2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Pratz K, Pollyea DA, Jonas BA, et al. Safety and efficacy of venetoclax (ven) in combination with decitabine or azacitidine in treatment-naive, elderly patients (≥65 years) with acute myeloid leukemia (AML). Haematologica. 2017;S472. [Google Scholar]
- 136.Wei AH, Strickland SA, Roboz GJ, et al. Updated safety and efficacy results of phase 1/2 study of venetoclax plus low-dose cytarabine in treatment-naïve acute myeloid leukemia patients aged ≥65 years and unfit for standard induction therapy. Haematologica. 2017;S473. [Google Scholar]
- 137.Brandts CH, Sargin B, Rode M, et al. Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation. Cancer Res. 2005;65(21):9643–9650. [DOI] [PubMed] [Google Scholar]
- 138.Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood. 2002;100:1532–1542. [DOI] [PubMed] [Google Scholar]
- 139.Rosnet O, Marchetto S, deLapeyriere O, et al. Murine Flt3, a gene encoding a novel tyrosine kinase receptor of the PDGFR/CSF1R family. Oncogene. 1991;6:1641–1650. [PubMed] [Google Scholar]
- 140.Meshinchi S, Appelbaum FR. Structural and functional alterations of FLT3 in acute myeloid leukemia. Clin Cancer Res. 2009;15:4263–4269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Kayser S, Levis MJ. FLT3 tyrosine kinase inhibitors in acute myeloid leukemia: clinical implications and limitations. Leuk Lymphoma. 2014;55(2):243–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Levis M. FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? Hematol Am Soc Hematol Educ Program. 2013;2013:220–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Schneider F, Hoster E, Schneider S, et al. Age-dependent frequencies of NPM1 mutations and FLT3-ITD in patients with normal karyotype AML (NK-AML). Ann Hematol. 2012;91:9–18. [DOI] [PubMed] [Google Scholar]
- 144.Kayser S, Schlenk RF, Londono MC, et al. Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood. 2009;114:2386–2392. [DOI] [PubMed] [Google Scholar]
- 145.Schnittger S, Bacher U, Haferlach C, et al. Diversity of the juxtamembrane and TKD1 mutations (exons 13–15) in the FLT3 gene with regards to mutant load, sequence, length, localization, and correlation with biological data. Genes Chromosom Cancer. 2012;51:910–924. [DOI] [PubMed] [Google Scholar]
- 146.Ponziani V, Gianfaldoni G, Mannelli F, et al. The size of duplication does not add to the prognostic significance of FLT3 internal tandem duplication in acute myeloid leukemia patients. Leukemia. 2006;20:2074–2076. [DOI] [PubMed] [Google Scholar]
- 147.Kusec R, Jaksic O, Ostojic S, et al. More on prognostic significance of FLT3/ITD size in acute myeloid leukemia (AML). Blood. 2006;108:405–406. [DOI] [PubMed] [Google Scholar]
- 148.Kim Y, Lee GD, Park J, et al. Quantitative fragment analysis of FLT3-ITD efficiently identifying poor prognostic group with high mutant allele burden or long ITD length. Blood Cancer J. 2015;5:e336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Stirewalt DL, Kopecky KJ, Meshinchi S, et al. Size of FLT3 internal tandem duplication has prognostic significance in patients with acute myeloid leukemia. Blood. 2006;107:3724–3726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Blau O, Berenstein R, Sindram A, et al. Molecular analysis of different FLT3-ITD mutations in acute myeloid leukemia. Leuk Lymphoma. 2013;54:145–152. [DOI] [PubMed] [Google Scholar]
- 151.Breitenbuecher F, Schnittger S, Grundler R, et al. Identification of a novel type of ITD mutations located in nonjuxtamembrane domains of the FLT3 tyrosine kinase receptor. Blood. 2009;113:4074–4077. [DOI] [PubMed] [Google Scholar]
- 152.Hollink IH, van den Heuvel-Eibrink MM, Arentsen-Peters ST, et al. NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern. Blood. 2011;118(13):3645–3656. [DOI] [PubMed] [Google Scholar]
- 153.Akiki S, Dyer SA, Grimwade D, et al. NUP98-NSD1 fusion in association with FLT3-ITD mutation identifies a prognostically relevant subgroup of pediatric acute myeloid leukemia patients suitable for monitoring by real time quantitative PCR. Genes Chromosom Cancer. 2013;52(11):1053–1064. [DOI] [PubMed] [Google Scholar]
- 154.Ostronoff F, Othus M, Gerbing RB, et al. NUP98/NSD1 and FLT3/ITD coexpression is more prevalent in younger AML patients and leads to induction failure: a COG and SWOG report. Blood. 2014;124(15):2400–2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Schiller J, Praulich I, Krings Rocha C, et al. Patient-specific analysis of FLT3 internal tandem duplications for the prognostication and monitoring of acute myeloid leukemia. Eur J Haematol. 2012;89:53–62. [DOI] [PubMed] [Google Scholar]
- 156.Grunwald MR, Tseng LH, Lin MT, et al. Improved FLT3 internal tandem duplication PCR assay predicts outcome after allogeneic transplant for acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20:1989–1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Lin MT, Tseng LH, Beierl K, et al. Tandem duplication PCR: an ultrasensitive assay for the detection of internal tandem duplications of the FLT3 gene. Diagn Mol Pathol. 2013;22:149–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Thol F, Kolking B, Damm F, et al. Next-generation sequencing for minimal residual disease monitoring in acute myeloid leukemia patients with FLT3-ITD or NPM1 mutations. Genes Chromosom Cancer. 2012;51:689–695. [DOI] [PubMed] [Google Scholar]
- 159.Levis MJ, Perl AE, Altman JK, et al. A next-generation sequencing-based assay for minimal residual disease assessment in AML patients with FLT3-ITD mutations. Blood Adv. 2018;2(8):825–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.DeZern AE, Sung A, Kim S, et al. Role of allogeneic transplantation for FLT3/ITD acute myeloid leukemia: outcomes from 133 consecutive newly diagnosed patients from a single institution. Biol Blood Marrow Transplant. 2011;17:1404–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Brunet S, Labopin M, Esteve J, 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. 2012;30:735–741. [DOI] [PubMed] [Google Scholar]
- 162.Lin PH, Lin CC, Yang HI, et al. Prognostic impact of allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia patients with internal tandem duplication of FLT3. Leuk Res. 2013;37:287–292. [DOI] [PubMed] [Google Scholar]
- 163.Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377(5):454–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Cortes JE, Tallman MS, Schiller GJ, et al. Phase 2b study of two dosing regimens of quizartinib monotherapy in FLT3-ITD mutated, relapsed or refractory AML. Blood. 2018;132(6):598–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Cortes J, Perl AE, Döhner H, et al. Quizartinib, an FLT3 inhibitor, as monotherapy in patients with relapsed or refractory acute myeloid leukaemia: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2018;19(7):889–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Wang ES, Stone RM, Tallman MS, Walter RB, Eckardt JR, Collins R. Crenolanib, a type I FLT3 TKI, can be safely combined with cytarabine and anthracycline induction chemotherapy and results in high response rates in patients with newly diagnosed FLT3 mutant acute myeloid leukemia. Blood. 2016;128(22). [Google Scholar]
- 167.Perl AE, Altman JK, Cortes J, et al. Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1–2 study. Lancet Oncol. 2017;18(8):1061–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Cortes J, Khaled S, Martinelli G, et al. Quizartinib significantly prolongs overall survival in patients with FLT3-internal tandem duplication–mutated (mut) relapsed/refractory aml in the phase 3, randomized, controlled QUANTUM-R trial. Haematologica. 2018;LBA2600. [Google Scholar]
- 169.Stein EM. IDH2 inhibition in AML: finally progress? Best Pract Res Clin Haematol. 2015;28(2–3):112–115. [DOI] [PubMed] [Google Scholar]
- 170.Corces-Zimmerman MR, Majeti R. Pre-leukemic evolution of hematopoietic stem cells: the importance of early mutations in leukemogenesis. Leukemia. 2014;28:2276–2282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Reitman ZJ, Yan H. Isocitrate dehydrogenase 1 and 2 mutations in cancer: alterations at a crossroads of cellular metabolism. J Natl Cancer Inst. 2010;102:932–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Lu C, Ward PS, Kapoor GS, et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature. 2012;483:474–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Abbas S, Lugthart S, Kavelaars FG, et al. Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood. 2010;116:2122–2126. [DOI] [PubMed] [Google Scholar]
- 174.DiNardo CD, Ravandi F, Agresta S, et al. Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML. Am J Hematol. 2015;90:732–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Rampal R, Alkalin A, Madzo J, et al. DNA hydroxymethylation profiling reveals that WT1 mutations result in loss of TET2 function in acute myeloid leukemia. Cell Rep. 2014;9:1841–1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Quek L, David MD, Kennedy A, et al. Clonal heterogeneity of acute myeloid leukemia treated with the IDH2 inhibitor enasidenib. Nat Med. 2018. August;24(8):1167–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.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(25):2386–2398. [DOI] [PubMed] [Google Scholar]
- 178.Chaturvedi A, Herbst L, Pusch S, et al. Pan-mutant-IDH1 inhibitor BAY1436032 is highly effective against human IDH1 mutant acute myeloid leukemia in vivo. Leukemia. 2017;31(10):2020–2028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Debarri H, Lebon D, Roumier C, et al. IDH1/2 but not DNMT3A mutations are suitable targets for minimal residual disease monitoring in acute myeloid leukemia patients: a study by the Acute Leukemia French Association. Oncotarget. 2015;6:42345–42353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Marschalek R. Systematic classification of mixed-lineage leukemia fusion partners predicts additional cancer pathways. Ann Lab Med. 2016;36(2):85–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Kayser S, Döhner 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;117:2137–2145. [DOI] [PubMed] [Google Scholar]
- 182.Yu BD, Hess JL, Horning SE, Brown GA, Korsmeyer SJ. Altered Hox expression and segmental identity in Mll-mutant mice. Nature. 1995;378:505–508. [DOI] [PubMed] [Google Scholar]
- 183.Dicker F, Haferlach C, Sundermann J, et al. Mutation analysis for RUNX1, MLL-PTD, FLT3-ITD, NPM1 and NRAS in 269 patients with MDS or secondary AML. Leukemia. 2010;24:1528–1532. [DOI] [PubMed] [Google Scholar]
- 184.Bacher U, Kern W, Schnittger S, Hiddemann W, Haferlach T, Schoch C. Population based age-specific incidences of cytogenetic subgroups of acute myeloid leukemia. Haematologica. 2005;90:1502–1510. [PubMed] [Google Scholar]
- 185.Dorrance AM, Liu S, Yuan W, et al. Mll partial tandem duplication induces aberrant Hox expression in vivo via specific epigenetic alterations. J Clin Invest. 2006;116:2707–2716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zorko NA, Bernot KM, Whitman SP, et al. Mll partial tandem duplication and Flt3 internal tandem duplication in a double knock-in mouse recapitulates features of counterpart human acute myeloid leukemias. Blood. 2012;120:1130–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Sun QY, Ding LW, Tan KT, et al. Ordering of mutations in acute myeloid leukemia with partial tandem duplication of MLL (MLL-PTD). Leukemia. 2017;31(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Whitman SP, Ruppert AS, Marcucci G, et al. Longterm disease-free survivors with cytogenetically normal acute myeloid leukemia and MLL partial tandem duplication: a Cancer and Leukemia Group B study. Blood. 2007;109:5164–5167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Schoch C, Schnittger S, Klaus M, Kern W, Hiddemann W, Haferlach T. AML with 11q23/MLL abnormalities as defined by the WHO classification: incidence, partner chromosomes, FAB subtype, age distribution, and prognostic impact in an unselected series of 1897 cytogenetically analyzed AML cases. Blood. 2003;102(7):2395–2402. [DOI] [PubMed] [Google Scholar]
- 190.Bernt KM, Zhu N, Sinha AU, et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell. 2011;20(1):66–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Deshpande AJ, Chen L, Fazio M, et al. Leukemic transformation by the MLL-AF6 fusion oncogene requires the H3K79 methyltransferase Dot1 l. Blood. 2013;121(13):2533–2541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Chen L, Deshpande AJ, Banka D, et al. Abrogation of MLL-AF10 and CALM-AF10-mediated transformation through genetic inactivation or pharmacological inhibition of the H3K79 methyltransferase Dot1 l. Leukemia. 2013;27(4):813–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Chen CW, Koche RP, Sinha AU, et al. DOT1L inhibits SIRT1-mediated epigenetic silencing to maintain leukemic gene expression in MLL-rearranged leukemia. Nat Med. 2015;21(4):335–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Kühn MW, Hadler MJ, Daigle SR, et al. MLL partial tandem duplication leukemia cells are sensitive to small molecule DOT1L inhibition. Haematologica. 2015;100(5):e190–e193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Daigle SR, Olhava EJ, Therkelsen CA, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20(1):53–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Daigle SR, Olhava EJ, Therkelsen CA, et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood. 2013;122(6):1017–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood. 2018;131(24):2661–2669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Rau RE, Rodriguez BA, Luo M, et al. DOT1L as a therapeutic target for the treatment of DNMT3A-mutant acute myeloid leukemia. Blood. 2016;128(7):971–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Yan XJ, Xu J, Gu ZH, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat Genet. 2011;43(4):309–315. [DOI] [PubMed] [Google Scholar]
- 200.Shen Y, Zhu YM, Fan X, et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood. 2011;118(20):5593–5603. [DOI] [PubMed] [Google Scholar]
- 201.Pløen GG, Nederby L, Guldberg P, et al. Persistence of DNMT3A mutations at long-term remission in adult patients with AML. Br J Haematol. 2014;167(4):478–486. [DOI] [PubMed] [Google Scholar]
- 202.Shlush LI, Zandi S, Mitchell A, et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 2014;506(7488):328–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Hou HA, Kuo YY, Liu CY, et al. DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood. 2012;119(2):559–568. [DOI] [PubMed] [Google Scholar]
- 205.Bolouri H, Farrar JE, Triche T Jr, et al. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat Med. 2018;24(1):103–112. [DOI] [PMC free article] [PubMed] [Google Scholar]