• FLT3-ITD MRD identifies patients at high relapse risk after intensive chemotherapy with midostaurin.

  • Conversion from FLT3-ITD MRDneg to FLT3-ITD MRDpos during follow-up was associated with a high relapse rate and inferior outcome.

Abstract

Measurable residual disease (MRD) monitoring in acute myeloid leukemia (AML) with an FLT3 internal tandem duplication (FLT3-ITDpos) has been hampered by the broad heterogeneity of ITD mutations. Using our recently developed FLT3-ITD paired-end next-generation sequencing (NGS)–based MRD assay (limit of detection 10−4 to 10−5), we evaluated the prognostic impact of MRD at different time points in 157 patients with FLT3-ITDpos AML who were enrolled in the German-Austrian Acute Myeloid Leukemia Study Group 16-10 trial and who were treated with a combination of intensive chemotherapy and midostaurin, followed by midostaurin maintenance. MRD negativity (MRDneg) after 2 cycles of chemotherapy (Cy2), which was observed in 111 of 142 (78%) patients, was predictive of superior 4-year rates of cumulative incidence of relapse (CIR) (4y-CIR; 26% vs 46%; P = .001) and overall survival (OS) (4y-OS; 70% vs 44%; P = .012). This survival advantage was also seen among patients who underwent allogeneic hematopoietic-cell transplantation during first complete remission (4y-CIR, 14% vs 39%; P = .001; 4y-OS, 71% vs 49%; P = .029). Multivariate models for CIR and OS after Cy2 revealed FLT3-ITD MRDneg as the only consistent favorable variable for CIR (hazard ratio [HR], 0.29; P = .006) and OS (HR, 0.39; P = .018). During follow-up, conversion from MRDneg to MRD positivity (MRDpos) was a strong, independent factor for inferior CIR (HR, 16.64; P < .001) and OS (HR, 4.05; P < .001). NGS-based FLT3-ITD MRD monitoring identifies patients at high risk for relapse and death following treatment with intensive chemotherapy and midostaurin. Using NGS-based technology.

Internal tandem duplications of the FLT3 gene (FLT3-ITD) are found in ∼10% to 15% of adult patients with newly diagnosed acute myeloid leukemia (AML).1-4 ITD mutations have been shown to be associated with poor prognosis because of a high relapse rate, in particular in cases with a high mutant to wild-type allelic ratio (AR; ≥0.5),5-9 an insertion site located in the beta-1 sheet of tyrosine kinase domain-1,9-11 and in patients without concomitant mutations in NPM1.7-9,12,13 

In the 2022 European LeukemiaNet (ELN) risk classification, AML with FLT3-ITD (without adverse-risk genetic lesions) is now categorized as intermediate risk, irrespective of the AR or concurrent presence of NPM1 mutation (NPM1mut).14 This revision to the 2017 ELN classification was based on methodologic issues with standardizing the assays for measurement of the FLT3-ITD AR, the modifying impact of midostaurin-based therapy on FLT3-ITD,11,13,15 and the increasing role of measurable residual disease (MRD) in treatment decisions.14,16 MRD monitoring allows response assessment and the early detection of relapse and therefore can be used for treatment decision-making and early intervention. In addition, MRD data have contributed significantly to refining relapse risk. Moreover, MRD monitoring is a powerful tool to assess kinetics and the depth of response during therapy, which is particularly informative for the evaluation of treatment effects within clinical trials that are investigating novel therapies.14,16,17 Based on these many clinical implications, MRD is currently being considered to serve as a surrogate end point in clinical trials, which may accelerate the approval of new drugs.

The currently most widely used methods for MRD assessment are multiparameter flow cytometry and quantitative polymerase chain reaction (qPCR).18 When compared with other molecular targets in AML, such as the recurrent gene fusions RUNX1::RUNX1T1, CBFB::MYH11, and PML::RARA, as well as NPM1mut, FLT3-ITD MRD monitoring using qPCR has been hampered by the heterogeneity of the ITD mutation types that are determined by the broad variety of ITD lengths and insertion sites. Recent advances in next-generation sequencing (NGS) have been shown to overcome these limitations and now offers the opportunity for MRD monitoring in FLT3-ITD–positive (FLT3-ITDpos) AML.19-23 The 2021 update consensus document set forth by the ELN MRD Working Party acknowledged these efforts and included technical specifications for NGS-based MRD testing and integrative assessment of MRD, irrespective of technique.16 

The objective of our study was to prospectively evaluate the prognostic impact of NGS-based MRD monitoring of FLT3-ITD in a cohort of 157 patients with FLT3-ITDpos AML who received intensive chemotherapy in combination with midostaurin, followed by midostaurin maintenance within the German-Austrian Acute Myeloid Leukemia Study Group 16-10 (AMLSG16-10) treatment trial (ClinicalTrials.gov identifier: NCT01477606).24,25 

Patient selection

All patients were enrolled in the AMLSG16-10 treatment trial.25 Patients were selected based on the following criteria: (1) achievement of complete remission (CR) or CR with incomplete blood count recovery (CRi) after 2 cycles of intensive chemotherapy (Cy2) combined with midostaurin, (2) availability of a diagnostic bone marrow (BM) or peripheral blood (PB) sample, and (3) ≥1 subsequent sample (BM after Cy2 and/or at end of treatment [EOT] and/or BM or PB during 3 to 12 months follow-up [FU]). Following these criteria, 157 patients were evaluated at diagnosis, 142 after Cy2, 116 at EOT, and 148 during the defined FU period (supplemental Figure 1). All patients gave informed consent according to the Declaration of Helsinki. Approval was obtained from the institutional review boards of the participating AMLSG institutions.

Molecular analyses

At the time of diagnosis, mutation analysis for FLT3-ITD and NPM1, as well as NPM1mut MRD assessment, was performed as previously described.15,17,26 

FLT3-ITD detection by targeted NGS

The NGS libraries were paired-end sequenced on an Illumina NGS platform according to the manufacturer’s recommendation (Illumina, San Diego, CA) with minor modifications to our previously described assay that exhibited a variant allele frequency (VAF) sensitivity of 10−4 to 10−5.20 Details on the experimental procedures are available in the supplemental Data. The raw sequencing data were analyzed using the bioinformatics program getITD.20 

Statistical analyses

CR/CRi, partial remission, overall survival (OS), relapse-free survival, and cumulative incidence of relapse (CIR) were defined according to standard criteria.14 Using landmark analyses, survival times for the time point after Cy2 were calculated from the date of first CR (CR1) or from the date of allogeneic hematopoietic-cell transplantation (HCT) in CR1 and were determined from the date the MRD sample was obtained for the EOT and FU time points. Patients who did not experience the event of interest at the end of FU were censored at the date of last contact. CIR was computed according to the method described by Gray.27 The median FU for survival was calculated using the reverse Kaplan-Meier estimate.28 Logistic regression and Cox proportional hazards models were used to identify prognostic variables for CR/CRi and OS.29 CIR was analyzed using cause-specific Cox models in which death during CR was considered a competing event. Additional covariates in the multivariate analysis were sex, FLT3-ITD AR, and NPM1 mutation status as dichotomous variables and BM blast count, white blood cell (WBC) count (log10 transformed), and age (10-year increase) as continuous variables; HCT during CR1 and MRD status over time were included as time-dependent variables. Missing values for covariates (WBC and BM blasts) were addressed by multiple imputation using chained equations. Mann-Whitney U tests were used to compare quantitative variables between patient subgroups; categorical variables were compared by means of Fisher exact tests. Associations between continuous variables were analyzed using the Spearman rank correlation coefficient. Survival distributions were estimated using the Kaplan-Meier method, and differences between groups were analyzed using 2-sided log-rank tests. An effect was considered significant if its P value was <5%. The analyses were not adjusted for multiple testing. All statistical analyses were performed using IBM SPSS Statistics 28.0.1.0, statistical software R (version 4.2.2), using the packages survival (version 3.5-0) and cmprsk (version 2.2-11), and/or GraphPad Prism7.

Patient and disease characteristics

Table 1 summarizes the baseline characteristics of the 157 patients with FLT3-ITDpos AML and of the 142 patients according to the FLT3-ITD MRD status after Cy2.

Table 1.

Characteristics of the 157 patients with FLT3-ITD-positive AML according to MRD status after Cy2 (n = 142)

Clinical and genetic variablesAll patients (N = 157)MRDneg Cy2 (n = 111)MRDpos Cy2 (n = 31)P value
Age (y), median (range) 54 (20-70) 54 (20-70) 51 (25-70) .229 
Sex, n (%)     
Male 69 (44) 45 (40) 17 (55) .219 
Female 88 (56) 66 (60) 14 (45)  
WBC (109/L)     
Median (range) 51.8 (0.5-356.4) 46.3 (0.5-356.4) 67.8 (1.1-279.6) .056 
Missing  
Hemoglobin (g/dL)     
Median (range) 9.3 (4.1-15.0) 9.2 (4.1-15.0) 9.6 (5.6-13.8) .553 
Missing  
Platelets (109/L)     
Median (range) 60 (5-352) 56 (5-352) 59 (13-148) .547 
Missing  
BM blasts (%)     
Median (range) 81 (0-100) 85 (0-100) 83 (20-100) .564 
Missing 18 13  
PB blasts (%)     
Median (range) 46 (0-99) 42 (0-99) 53 (0-98) .364 
Missing 10  
AML type, n (%)     
De novo 142 (90) 100 (90) 29 (94) .388 
Secondary 8 (5) 6 (5)  
Therapy-related 7 (5) 5 (5) 2 (6)  
ELN 2017 risk classification, n (%)     
Favorable 31 (20) 22 (20) 4 (13) <.001 
Intermediate 100 (64) 79 (72) 15 (48)  
Adverse 25 (16) 9 (8) 12 (39)  
Missing  
FLT3-ITD AR, n (%)     
Low (<0.5) 67 (43) 47 (42) 12 (39) .837 
High (≥0.5) 90 (57) 64 (58) 19 (61)  
FLT3-TKD, n (%)     
Yes 6 (4) 2 (2) 3 (10) .069 
No 151 (96) 109 (98) 28 (90)  
Mutated NPM1, n (%)     
Yes 111 (71) 90 (81) 12 (39) <.001 
No 46 (29) 21 (19) 19 (61)  
Clinical and genetic variablesAll patients (N = 157)MRDneg Cy2 (n = 111)MRDpos Cy2 (n = 31)P value
Age (y), median (range) 54 (20-70) 54 (20-70) 51 (25-70) .229 
Sex, n (%)     
Male 69 (44) 45 (40) 17 (55) .219 
Female 88 (56) 66 (60) 14 (45)  
WBC (109/L)     
Median (range) 51.8 (0.5-356.4) 46.3 (0.5-356.4) 67.8 (1.1-279.6) .056 
Missing  
Hemoglobin (g/dL)     
Median (range) 9.3 (4.1-15.0) 9.2 (4.1-15.0) 9.6 (5.6-13.8) .553 
Missing  
Platelets (109/L)     
Median (range) 60 (5-352) 56 (5-352) 59 (13-148) .547 
Missing  
BM blasts (%)     
Median (range) 81 (0-100) 85 (0-100) 83 (20-100) .564 
Missing 18 13  
PB blasts (%)     
Median (range) 46 (0-99) 42 (0-99) 53 (0-98) .364 
Missing 10  
AML type, n (%)     
De novo 142 (90) 100 (90) 29 (94) .388 
Secondary 8 (5) 6 (5)  
Therapy-related 7 (5) 5 (5) 2 (6)  
ELN 2017 risk classification, n (%)     
Favorable 31 (20) 22 (20) 4 (13) <.001 
Intermediate 100 (64) 79 (72) 15 (48)  
Adverse 25 (16) 9 (8) 12 (39)  
Missing  
FLT3-ITD AR, n (%)     
Low (<0.5) 67 (43) 47 (42) 12 (39) .837 
High (≥0.5) 90 (57) 64 (58) 19 (61)  
FLT3-TKD, n (%)     
Yes 6 (4) 2 (2) 3 (10) .069 
No 151 (96) 109 (98) 28 (90)  
Mutated NPM1, n (%)     
Yes 111 (71) 90 (81) 12 (39) <.001 
No 46 (29) 21 (19) 19 (61)  

TKD, tyrosine kinase domain.

Paired BM and PB analysis

To determine tissue-dependency on VAF, we compared 29 paired PB and BM samples at diagnosis (n = 10) and after Cy2 (n = 19). Although the median FLT3-ITD VAF was slightly higher in BM than in PB (29.24% vs 23.59%; P = .065) at diagnosis, the median VAF (BM, 0.11% vs PB, 0.03%; P < .001) and MRD negativity (MRDneg; BM, 0/19 [0%] vs PB, 7/19 [37%]; P = .008) differed significantly after Cy2 (supplemental Figure 2), clearly indicating the higher sensitivity in BM. Therefore, subsequently, only BM samples were selected for MRD assessment after Cy2 and at EOT. PB samples were analyzed only at diagnosis (n = 37) and during FU (n = 29).

NGS-based assessment of FLT3-ITD at diagnosis

A total of 465 ITDs were identified in 157 patients with FLT3-ITDpos AML at diagnosis. Of the 465 ITDs, the median ITD length was 51 nucleotides (range, 9-285) and the median ITD VAF was 0.312% (0.006-92.26) (supplemental Figure 3). In total, 108 patients (69%) exhibited >1 ITD (median, 2; range, 1-16). The median total ITD VAF per patient (determined as sum of individual ITD VAFs) was 31.54% (0.46-92.26). Total ITD VAF per patient was correlated positively with higher WBC count (Rho, 0.287; P < .001), BM blast count (Rho, 0.196; P = .020), PB blast count (Rho, 0.264; P = .001), and lactate dehydrogenase level (Rho = 0.316; P < .001) and correlated inversely with the number of ITD clones (Rho, −0.226; P = .004). There was no correlation with age, sex, or NPM1 mutation status. The NGS-based calculated ITD AR (∑VAF/[100 − ∑VAF]) correlated positively with AR as determined by a GeneScan analysis (Rho, 0.855; P < .001; supplemental Figure 4).

At diagnosis, FLT3-ITD VAF a as log2 transformed continuous variable did not have an impact on OS (hazard ratio [HR], 1.15; 95% confidence interval [CI], 0.92-1.42; P = .218) or CIR (HR, 0.95; 95% CI, 0.78-1.17; P = .646).

Prognostic impact of FLT3-ITD MRD during therapy

The median FU time of the 157 patients with FLT3-ITDpos AML was 47.4 months (95% CI, 40.9-53.9); 59 of the 157 (37.6%) patients died. The median OS and 2-year OS rate were 68.9 months (95% CI, 51.5% to not applicable) and 0.72 (95% CI, 0.64-0.79), respectively. Allogeneic HCT during CR1 was performed in 122 of 157 (78%) patients and was performed at any time during the disease course in 135 of 157 (86%) patients.

Impact of FLT3-ITD MRD after Cy2

All FLT3-ITDs identified after Cy2 were already detectable at diagnosis. In relation to diagnosis, the median log10 reduction in the total FLT3-ITD VAF was 4.6 (range, 0.42-5.17; P < .001); FLT3-ITD MRDneg, defined as undetectable FLT3-ITD, was achieved in 111 of 142 (78%) patients (Figure 1A). Patients with FLT3-ITD MRDneg and MRD positivity (MRDpos) differed significantly in terms of concurrent NPM1mut and the ELN 2017 risk classification (Figure 1B; Table 1). The only favorable factor for achievement of FLT3-ITD MRDneg after Cy2 was concurrent NPM1mut (odds ratio [OR], 10.45; 95% CI, 3.40-32.07; P < .001); adverse factors were WBC count (OR for 10-fold increase, 0.31; 95% CI, 0.10-0.92; P = .035) and the administration of a second induction cycle (administered in 37 patients who achieved partial remission only after the first induction; OR, 0.19; 95% CI, 0.06-0.58; P = .004; supplemental Table 1).

Figure 1.

Kinetics of FLT3-ITD VAF during treatment and early FU. (A) For the entire cohort of 157 FLT3-ITDpos, ITD VAF significantly decreased during therapy and during early FU. FLT3-ITD MRDneg was achieved in 78% after Cy2, 94% at EOT, and 85% during FU. (B) Patients separated according to NPM1mut status, demonstrating a significantly higher VAF reduction after Cy2 and higher frequencies of MRDneg for concurrent NPM1mut patients. Dx, diagnosis; wt, wild-type.

Figure 1.

Kinetics of FLT3-ITD VAF during treatment and early FU. (A) For the entire cohort of 157 FLT3-ITDpos, ITD VAF significantly decreased during therapy and during early FU. FLT3-ITD MRDneg was achieved in 78% after Cy2, 94% at EOT, and 85% during FU. (B) Patients separated according to NPM1mut status, demonstrating a significantly higher VAF reduction after Cy2 and higher frequencies of MRDneg for concurrent NPM1mut patients. Dx, diagnosis; wt, wild-type.

Close modal

A higher log10 reduction in FLT3-ITD VAF was significantly associated with a lower CIR rate (HR for 10-fold better VAF reduction, 0.56; 95% CI, 0.43-0.71; P < .001) and improved OS (HR, 0.75; 95% CI, 0.60-0.93; P = .010; supplemental Table 2).

We next examined the prognostic impact of MRDneg, which was achieved in 111 of 142 (78%) patients. In the univariate analysis, achieving MRDneg was predictive of a superior 4-year CIR rate (4y-CIR; 26% vs 46% for MRDpos; HR, 0.33; 95% CI, 0.17-0.64; P = .001) and 4-year OS (4y-OS; 70% vs 44%; HR, 0.47; 95% CI, 0.26-0.85; P = .012) (Figure 2A-B; supplemental Table 2). Figure 3 illustrates the course of events for every individual patient according to FLT3-ITD MRD status after Cy2. Of note, for patients with FLT3-ITD MRDpos status, the risk for relapse correlated with the MRD burden, in particular ≥0.1%, the threshold provisionally used to define NGS-MRD test positivity according to the 2021 ELN MRD Working Party (Figure 4; supplemental Table 2). We additionally evaluated the impact of a ≥3 log10 reduction in FLT3-ITD VAF (MR3.0). Achieving MR3.0 after Cy2 was associated with a lower CIR rate (4y-CIR, 30% vs 54%; HR, 0.31; 95% CI, 0.16-0.62; P < .001) but not with improved OS (4y-OS, 68% vs 45%; HR, 0.58; 95% CI, 0.30-1.10; P = .097) (supplemental Figure 5; supplemental Table 2).

Figure 2.

Prognostic impact of FLT3-ITD MRDneg after Cy2. (A) Marginal Aalen-Johansen plot of CIR and (B) marginal Kaplan-Meier plot of OS for the 142 FLT3-ITDpos patients during CR according to FLT3-ITD MRD status. (C) CIR and (D) OS according to FLT3-ITD MRD status before HCT in CR1. HRs and 95% CIs are given for FLT3-ITD MRDneg status.

Figure 2.

Prognostic impact of FLT3-ITD MRDneg after Cy2. (A) Marginal Aalen-Johansen plot of CIR and (B) marginal Kaplan-Meier plot of OS for the 142 FLT3-ITDpos patients during CR according to FLT3-ITD MRD status. (C) CIR and (D) OS according to FLT3-ITD MRD status before HCT in CR1. HRs and 95% CIs are given for FLT3-ITD MRDneg status.

Close modal
Figure 3.

Individual disease and treatment course after Cy2 according to FLT3-ITD MRD status. (A) Course of events for the 111 patients who were FLT3-ITD MRD negative after Cy2 and (B) for the 31 patients who were FLT3-ITD MRD positive after Cy2. (C) Course of events for the 22 patients who were FLT3-ITD MRD positive during FU.

Figure 3.

Individual disease and treatment course after Cy2 according to FLT3-ITD MRD status. (A) Course of events for the 111 patients who were FLT3-ITD MRD negative after Cy2 and (B) for the 31 patients who were FLT3-ITD MRD positive after Cy2. (C) Course of events for the 22 patients who were FLT3-ITD MRD positive during FU.

Close modal
Figure 4.

Outcome according to FLT3-ITD MRD cutoffs after Cy2. CIR (A) and OS (B) according to various FLT3-ITD MRD cutoffs after Cy2. Results of pairwise comparisons are provided below the x-axis.

Figure 4.

Outcome according to FLT3-ITD MRD cutoffs after Cy2. CIR (A) and OS (B) according to various FLT3-ITD MRD cutoffs after Cy2. Results of pairwise comparisons are provided below the x-axis.

Close modal

Next, we performed a multivariate Cox regression analysis in different models that included the FLT3-ITD MRD log10 reduction, an FLT3-ITD VAF <0.1%, achievement of MR3.0, or FLT3-ITD MRDneg status. In all models, a higher log10 reduction, FLT3-ITD VAF <0.1%, achievement of MR3.0, and MRDneg were the only consistent favorable variables for risk of relapse and OS with the exception of MR3.0 for OS. NPM1mut and HCT during CR1 were favorable factors only for CIR (Table 2; supplemental Table 3).

Table 2.

Multivariate analyses used to determine the prognostic significance of FLT3-ITD MRDneg at various landmarks

Clinical and genetic variablesCIROS
HR (95% CI)PHR (95% CI)P
Landmark Cy2 (n = 142) 
Age (10 y-increase) 0.95 (0.71-1.27) .727 1.42 (1.07-1.89) .017 
Female 0.61 (0.32-1.15) .123 0.63 (0.36-1.09) .097 
WBC (log101.18 (0.69-2.05) .534 0.68 (0.38-1.23) .200 
BM blasts 1.00 (0.98-1.02) .890 1.00 (0.99-1.02) .684 
NPM1mut 0.31 (0.14-0.68) .005 0.85 (0.45-1.59) .599 
FLT3-ITDhigh 1.02 (0.52-2.01) .948 1.12 (0.61-2.07) .711 
HCT in CR1  0.13 (0.05-0.30) <.001 0.64 (0.37-1.12) .117 
FLT3-ITD MRDneg 0.29 (0.13-0.69) .006 0.39 (0.18-0.85) .018 
Landmark EOT (n = 116) 
Age (10 y-increase) 0.98 (0.71-1.36) .900 1.73 (1.27-2.35) <.001 
Female 0.61 (0.30-1.25) .175 0.64 (0.36-1.14) .127 
WBC (log101.10 (0.61-2.00) .743 0.82 (0.49-1.37) .448 
BM blasts 1.00 (0.98-1.02) .955 0.99 (0.98-1.01) .354 
NPM1mut 0.32 (0.14-0.76) .009 0.82 (0.42-1.60) .568 
FLT3-ITDhigh 1.03 (0.45-2.36) .948 0.96 (0.50-1.85) .901 
HCT in CR1  0.19 (0.08-0.44) <.001 0.59 (0.30-1.17) .130 
FLT3-ITD MRDneg 0.34 (0.09-1.20) .093 0.30 (0.09-0.98) .046 
Landmark FU (3-12 mo) (n = 148) 
Age (10 y-increase) 1.03 (0.80-1.35) .826 1.24 (0-95-1.629) .109 
Female 0.48 (0.24-0.95) .035 0.62 (0.35-1.11) .107 
WBC (log100.74 (0.40-1.354) .321 0.69 (0.40-1.19) .180 
BM blasts 1.00 (0.99-1.02) .749 1.00 (0.99-1.02) .626 
NPM1mut 0.53 (0.25-1.12) .094 0.95 (0.47-1.90) .879 
FLT3-ITDhigh 1.12 (0.52-2.39) .774 0.99 (0.51-1.93) .9477 
HCT in CR1 0.12 (0.05-0.32) <.001 0.81 (0.40-1.62) .541 
FLT3-ITD MRDneg   
FLT3-ITD MRDconv_FU 16.64 (6.52-42.48) <.001 4.05 (1.78-9.18) <.001 
FLT3-ITD MRDpers 51.98 (14.75-183.15) <.001 3.69 (1.26-10.82) .017 
Clinical and genetic variablesCIROS
HR (95% CI)PHR (95% CI)P
Landmark Cy2 (n = 142) 
Age (10 y-increase) 0.95 (0.71-1.27) .727 1.42 (1.07-1.89) .017 
Female 0.61 (0.32-1.15) .123 0.63 (0.36-1.09) .097 
WBC (log101.18 (0.69-2.05) .534 0.68 (0.38-1.23) .200 
BM blasts 1.00 (0.98-1.02) .890 1.00 (0.99-1.02) .684 
NPM1mut 0.31 (0.14-0.68) .005 0.85 (0.45-1.59) .599 
FLT3-ITDhigh 1.02 (0.52-2.01) .948 1.12 (0.61-2.07) .711 
HCT in CR1  0.13 (0.05-0.30) <.001 0.64 (0.37-1.12) .117 
FLT3-ITD MRDneg 0.29 (0.13-0.69) .006 0.39 (0.18-0.85) .018 
Landmark EOT (n = 116) 
Age (10 y-increase) 0.98 (0.71-1.36) .900 1.73 (1.27-2.35) <.001 
Female 0.61 (0.30-1.25) .175 0.64 (0.36-1.14) .127 
WBC (log101.10 (0.61-2.00) .743 0.82 (0.49-1.37) .448 
BM blasts 1.00 (0.98-1.02) .955 0.99 (0.98-1.01) .354 
NPM1mut 0.32 (0.14-0.76) .009 0.82 (0.42-1.60) .568 
FLT3-ITDhigh 1.03 (0.45-2.36) .948 0.96 (0.50-1.85) .901 
HCT in CR1  0.19 (0.08-0.44) <.001 0.59 (0.30-1.17) .130 
FLT3-ITD MRDneg 0.34 (0.09-1.20) .093 0.30 (0.09-0.98) .046 
Landmark FU (3-12 mo) (n = 148) 
Age (10 y-increase) 1.03 (0.80-1.35) .826 1.24 (0-95-1.629) .109 
Female 0.48 (0.24-0.95) .035 0.62 (0.35-1.11) .107 
WBC (log100.74 (0.40-1.354) .321 0.69 (0.40-1.19) .180 
BM blasts 1.00 (0.99-1.02) .749 1.00 (0.99-1.02) .626 
NPM1mut 0.53 (0.25-1.12) .094 0.95 (0.47-1.90) .879 
FLT3-ITDhigh 1.12 (0.52-2.39) .774 0.99 (0.51-1.93) .9477 
HCT in CR1 0.12 (0.05-0.32) <.001 0.81 (0.40-1.62) .541 
FLT3-ITD MRDneg   
FLT3-ITD MRDconv_FU 16.64 (6.52-42.48) <.001 4.05 (1.78-9.18) <.001 
FLT3-ITD MRDpers 51.98 (14.75-183.15) <.001 3.69 (1.26-10.82) .017 

FLT3-ITDhigh, FLT3-internal tandem duplication with AR ≥0.5; MRDconv_FU, conversion from MRD negative at the last previously assessed time point to MRD positive at the time point FU; MRDpers, persistent MRDpos.

As time-dependent variable.

Impact of FLT3-ITD MRD before allogeneic HCT

Of the 142 patients assessed for FLT3-ITD MRD after Cy2, 107 patients underwent HCT during CR1 with 84 patients undergoing HCT immediately after Cy2 and 23 patients after receiving additional consolidation therapy with high-dose cytarabine. In total, 81 of 107 patients (76%) were FLT3-ITD MRDneg before HCT; the median time from MRD assessment after Cy2 and HCT was 30 days (range, 7-88). FLT3-ITD MRDpos before HCT was associated with an increased risk for relapse (4y-CIR, 39% vs 14%; HR, 4.90; 95% CI, 1.87-12.83; P = .001) and inferior outcome (4y-OS, 49% vs 71%; HR, 2.17; 95% CI, 1.06-4.43; P = .029) (Figure 2C-D). As shown in supplemental Figure 6, patients with FLT3-ITD MRDpos status before HCT had a comparable outcome as those who were FLT3-ITD MRDneg after Cy2 and were treated with conventional consolidation. Of the 26 patients with FLT3-ITD MRDpos status before HCT, 21 patients were also eligible at EOT, and 15 (71%) patients achieved FLT3-ITD MRDneg after HCT.

Impact of FLT3-ITD MRD at EOT

All FLT3-ITDs detected at EOT were already detectable at diagnosis. When compared with diagnosis, the median log10 reduction in the total FLT3-ITD VAF was 4.7 (range, 1.55-5.19; P < .001); FLT3-ITD MRDneg status was achieved in 109 of 116 (94%) patients (Figure 1A). In the Cox regression analysis, MRDneg status at EOT was significantly associated with improved OS and trended toward a reduced risk for relapse, likely because of the high rate of patients with FLT3-ITD MRDneg statusat EOT (Table 2).

FLT3-ITD MRD monitoring during FU

To assess the risk for relapse after completion of intensive therapy, we analyzed MRD at least at 1 time point between 3 and 12 months after EOT during FU in 148 patients (BM, n = 119; PB, n = 29). Of these 148 patients, 117 (79%) had started maintenance with midostaurin with a median of 9 cycles (range, 1-12). Overall, 22 (15%) patients were FLT3-ITD MRDpos during FU (all FLT3-ITDs were detected at diagnosis; Figure 3C). FLT3-ITD MRD persistence, defined as MRDpos at all time points (range, 3-4) assessed, was detected in 6 patients and all relapsed; 16 patients (including 9 under maintenance) converted from FLT3-ITD MRDneg to MRDpos (median VAF, 0.058%; range, 0.006%-91.965%) within 5.78 months (median time from last MRDneg to first MRDpos sample; range, 2.07-10.0 months). Of the 16 patients, 13 (81%) (including 7 after HCT) relapsed within a median time of 7 days (measured from time point of MRD conversion in FU [MRDconv_FU] to hematologic relapse; range, 0-197 days), translating into a significantly increased relapse risk (2y-CIR, 81% vs 16%; HR, 11.00; 95% CI, 5.44-22.28; P < .001) and inferior OS (2y-OS, 31% vs 80%; HR, 4.31; 95% CI, 2.22-8.38; P < .001) (supplemental Figure 7). Three of the 16 converted patients remained in continuous remission after allogeneic HCT, and all 3 exhibited low FLT3-ITD MRD levels (<0.008%); 2 of them received maintenance with midostaurin, and all 3 became MRD negative later on.

To analyze the impact of the FLT3-ITD MRD status over time, we performed 2 Cox regression models with MRD status as a time-dependent covariate. One model considered the impact of MRDpos at any time point (after Cy2, at EOT, and/or during FU) and the second addressed MRD conversion (MRDconv, MRDneg to MRDpos) after achievement of MRDneg after Cy2. In both models, MRDpos was the only consistent unfavorable variable for risk of relapse and OS (Table 3).

Table 3.

Multivariate analyses used to determine the prognostic significance of FLT3-ITD MRD status over time

Clinical and genetic variablesCIROS
HR (95% CI)PHR (95% CI)P
Patients N = 157 
Events n = 35 n = 59 
Age (10 y-increase) 0.99 (0.96-1.02) .644 1.03 (1.01-1.06) .016 
Female 0.36 (0.17-0.76) .007 0.57 (0.33-0.97) .038 
WBC (log100.91 (0.48-1.74) .779 0.67 (0.40-1.12) .129 
BM blasts 1.00 (0.98-1.01) .769 1.00 (0.99-1.01) .882 
NPM1mut 0.34 (0.16-0.71) .004 0.90 (0.49-1.65) .727 
FLT3-ITDhigh 1.16 (0.51-2.64) .715 1.28 (0.69-2.36) .437 
HCT in CR1 0.17 (0.08-0.38) <.001 0.95 (0.50-1.82) .885 
FLT3-ITD MRDpos  6.83 (2.72-17.13) <.001 3.32 (1.69-6.54) <.001 
Patients n = 110 
Events n = 21 n = 36 
Age (10 y-increase) 0.99 (0.95-1.03) .597 1.03 (0.99-1.07) .106 
Female 0.34 (0.13-0.89) .028 0.48 (0.24-0.95) .034 
WBC (log100.82 (0.36-1.87) .646 0.56 (0.29-1.09) .087 
BM blasts 0.99 (0.97-1.02) .603 1.00 (0.99-1.02) .630 
NPM1mut 0.42 (0.13-1.28) .126 1.00 (0.39-2.58) .998 
FLT3-ITDhigh 1.52 (0.50-4.66) .464 1.30 (0.57-2.93) .534 
HCT in CR1 0.09 (0.03-0.27) <.001 1.30 (0.59-2.86) .508 
FLT3-ITD MRDconv  9.94 (2.24-44.22) .003 2.83 (0.98-8.18) .055 
Clinical and genetic variablesCIROS
HR (95% CI)PHR (95% CI)P
Patients N = 157 
Events n = 35 n = 59 
Age (10 y-increase) 0.99 (0.96-1.02) .644 1.03 (1.01-1.06) .016 
Female 0.36 (0.17-0.76) .007 0.57 (0.33-0.97) .038 
WBC (log100.91 (0.48-1.74) .779 0.67 (0.40-1.12) .129 
BM blasts 1.00 (0.98-1.01) .769 1.00 (0.99-1.01) .882 
NPM1mut 0.34 (0.16-0.71) .004 0.90 (0.49-1.65) .727 
FLT3-ITDhigh 1.16 (0.51-2.64) .715 1.28 (0.69-2.36) .437 
HCT in CR1 0.17 (0.08-0.38) <.001 0.95 (0.50-1.82) .885 
FLT3-ITD MRDpos  6.83 (2.72-17.13) <.001 3.32 (1.69-6.54) <.001 
Patients n = 110 
Events n = 21 n = 36 
Age (10 y-increase) 0.99 (0.95-1.03) .597 1.03 (0.99-1.07) .106 
Female 0.34 (0.13-0.89) .028 0.48 (0.24-0.95) .034 
WBC (log100.82 (0.36-1.87) .646 0.56 (0.29-1.09) .087 
BM blasts 0.99 (0.97-1.02) .603 1.00 (0.99-1.02) .630 
NPM1mut 0.42 (0.13-1.28) .126 1.00 (0.39-2.58) .998 
FLT3-ITDhigh 1.52 (0.50-4.66) .464 1.30 (0.57-2.93) .534 
HCT in CR1 0.09 (0.03-0.27) <.001 1.30 (0.59-2.86) .508 
FLT3-ITD MRDconv  9.94 (2.24-44.22) .003 2.83 (0.98-8.18) .055 

FLT3-ITDhigh, FLT3-internal tandem duplication with AR ≥0.5; MRDconv, conversion from MRD negative after Cy2 of intensive chemotherapy to MRD positive over time; MRDpos, MRD positive at any time point.

As time-dependent variable.

Impact of concurrent NPM1 mutation on FLT3-ITD MRD

In total, 111 of the 157 (71%) patients had concomitant NPM1mut. Concurrent NPM1mut favorably impacted the log10 VAF reduction (median, 4.7 vs 3.7 for NPM1wt; P < .001) and the achievement of FLT3-ITD MRDneg status (88% vs 53%; P < .001) after Cy2 (Figure 1B). This translated into a lower CIR rate (4y-CIR FLT3-ITD MRDneg/NPM1mut vs FLT3-ITD MRDneg/NPM1WT, 19% vs 68%; HR, 0.23; 95% CI, 0.10-0.50; P < .001) and a trend toward improved OS (4y-OS, 72% vs 59%; HR, 0.52; 95% CI, 0.25-1.09; P = .071; supplemental Figure 8). No additional benefit was observed for NPM1mut at EOT, but NPM1mut was associated with FLT3-ITD MRDneg during FU (91% vs 70%; P = .002; Figure 1B).

Comparative analysis of FLT3-ITD and NPM1mut MRD assessment

According to the ELN MRD Working Party, in NPM1mut AML, MRD should be assessed, preferentially in PB, after Cy2 and in BM at EOT and during FU. In line with this recommendation, we correlated NGS-based FLT3-ITD MRD assessed in BM with qPCR-based NPM1mut MRD assessed in PB after Cy2 (Figure 5A). Of the 82 eligible patients, 41 (50%) were FLT3-ITD MRDneg/NPM1mut MRDneg, 33 (40%) were FLT3-ITD MRDneg/NPM1mut MRDpos, 7 (9%) were FLT3-ITD MRDpos/NPM1mut MRDpos, and 1 (1%) was FLT3-ITD MRDpos/NPM1mut MRDneg. It should be noted that an FLT3-ITD MRDneg status was associated with a lower relapse risk and improved outcome irrespective of NPM1mut MRD status (Figure 5B-C). Because of the small number of events in this subgroup (relapses, n = 13; deaths, n = 14) Cox regression analysis on relapse-free survival was performed and confirmed the beneficial impact of FLT3-ITD MRDneg status regardless of the NPM1mut MRD status (supplemental Table 4).

Figure 5.

Paired FLT3-ITD and NPM1mut MRD status. (A) MRD kinetics of FLT3-ITD and NPM1mut and corresponding frequencies of MRDneg status of respective pts during therapy and early FU. (B) CIR and (C) OS according to FLT3-ITD MRD and NPM1mut MRD status after Cy2 (1 pt with FLT3-ITD MRDpos/NPM1mut MRDneg was excluded). Dx, diagnosis; pts, patients.

Figure 5.

Paired FLT3-ITD and NPM1mut MRD status. (A) MRD kinetics of FLT3-ITD and NPM1mut and corresponding frequencies of MRDneg status of respective pts during therapy and early FU. (B) CIR and (C) OS according to FLT3-ITD MRD and NPM1mut MRD status after Cy2 (1 pt with FLT3-ITD MRDpos/NPM1mut MRDneg was excluded). Dx, diagnosis; pts, patients.

Close modal

At EOT and during FU, an FLT3-ITD MRDneg status was more frequent than an NPM1mut MRDneg status. In addition, all 3 patients with FLT3-ITD MRDpos statuswere also NPM1mut MRDpos at EOT. Furthermore, of the 30 patients with NPM1mut MRDpos, 26 exhibited NPM1mut MRD at low level provisionally defined as <2%. Similar findings were also observed during FU. The proportion of FLT3-ITD MRDneg/NPM1mut MRDneg increased from 50% after Cy2 to 68% during FU (Figure 5).

In this study, we performed FLT3-ITD MRD monitoring in 157 adult patients with FLT3-ITDpos AML using a highly sensitive NGS-based assay as previously reported by us.20 The data revealed that FLT3-ITD MRD was a highly significant risk factor for relapse and OS and even outperformed known risk factors, such as NPM1 mutational status and FLT3-ITD AR.

The particular strengths of our study are twofold. First, sequential biosampling was done prospectively within a controlled clinical trial at defined time points, that is, after Cy2, at the EOT, and during FU. Second, all patients received intensive chemotherapy in combination with the FLT3 inhibitor midostaurin, which has improved outcomes and is now considered standard of care in patients with FLT3-ITDpos AML.15,25 Recently, addition of the second-generation FLT3 inhibitor quizartinib to intensive chemotherapy has also been approved for the treatment of patients with newly diagnosed FLT3-ITDpos AML.

Three recently published studies reported on the clinically relevant impact of FLT3-ITD MRD monitoring using NGS-based assays.21-23 In contrast with our study, these analyses were restricted to a single time point (after 1 or 2 induction cycles) and were performed in heterogeneous patient cohorts, particularly with respect to treatment with an additional FLT3 inhibitor.21-23 Furthermore, 2 of these studies evaluated the impact of FLT3-ITD MRD specifically before HCT; the Pre-MEASURE study analyzed the PB of 608 patients with FLT3-ITDpos AML before HCT during CR123 and the study by Loo et al and Dillon et al analyzed samples from 104 patients with FLT3-ITDpos AML, irrespective of CR1, CR2, or molecular relapse.22 In the HOVON study, 93 of 161 (58%) intensively treated patients underwent HCT during CR1.21 Regarding concurrent NPM1mut, the prevalence of 75% in the study by Loo et al and Dillon et al was comparable with ours (71%) and lower than that in the HOVON (57%) and Pre-MEASURE studies (58%).

After 2 cycles of intensive chemotherapy is the first time point at which MRD assessment in the BM is considered to be clinically relevant.16 In our study, a higher reduction in FLT3-ITD VAF and achieving FLT3-ITD MRDneg after Cy2 in the BM were statistically significant favorable prognostic factors in terms of both relapse risk and OS and therefore enable a refined risk assessment of patients in hematologic response. A concurrent NPM1 mutation and HCT during CR1 were additional significant favorable factors for CIR, whereas FLT3-ITD MRDneg was the only significant factor for OS. Concurrent NPM1mut correlated with a deeper molecular response, as reflected in a better FLT3-ITD VAF reduction and a significantly higher rate of FLT3-ITD MRDneg after Cy2.

Our observation that FLT3-ITD MRDneg status after Cy2 is of high prognostic relevance is in line with the data recently published by the HOVON group.21 In their study, achievement of MRDneg after 2 induction cycles identified patients with a significantly lower 4y-CIR rate (33% vs 75%; P < .001) and improved OS (4y-OS, 57% vs 31%; P < .001). Similar results were reported for the QuANTUM-first study that showed that patients who achieved CR/composite CR after Cy2 and MRD levels <10−4 had a significantly improved OS when compared with patients above that MRD cutoff.30 The current ELN Working Party on MRD proposed a cutoff of 0.1% for NGS-based MRD assessment but also stated that this cutoff is provisional and not based on robust data. As shown in the 3 published studies and this study, the evaluation of different cutoffs is limited by the small patient numbers that underline the current uncertainty regarding thresholds and highlight the need for future studies (eg, meta-analysis) to define this.16,21-23 For MRD-based prognostication and prediction and for comparability between different study populations and techniques, the value of cutoffs beyond MRDneg, such as the log reduction in the transcript level between diagnosis and after induction, are under evaluation in clinical trials, as are other well-established MRD targets.16 

In FLT3-ITDpos AML, concurrent NPM1mut has been shown to be a favorable prognostic factor for all survival endpoints.7,8,12,13,22 Consistent with the HOVON data, concurrent NPM1mut was associated with a significantly higher percentage of FLT3-ITD MRDneg after Cy2. Although, in the HOVON study, identical relapse rates were observed for the entire FLT3-ITD MRDneg cohort and the FLT3-ITD MRDneg/NPM1mut cohort (4y-CIR 33% each), we found a lower CIR rate for FLT3-ITD MRDneg/NPM1mut patients than for the entire cohort of patients with FLT3-ITD MRDneg (4y-CIR, 19% vs 29%), underlining the favorable effect of a concurrent NPM1mut in terms of achieving a deeper molecular response.

Allogeneic HCT has been shown to improve the outcome of patients with FLT3-ITDpos AML.8,9,13,14 In our study, HCT during CR1 was an independent favorable factor for CIR at all time points, demonstrating that HCT during CR1 is an important pillar for treatment of FLT3-ITDpos AML. Of note, FLT3-ITD MRD status before HCT provided an additional prognostic impact. An FLT3-ITD MRDneg status before HCT was associated with a lower risk for relapse and superior outcome (Figure 2). In line with the recently published studies that demonstrated the prognostic impact of FLT3-ITD MRDneg status before HCT and considering previous studies on various molecular MRD markers, these data underline the prognostic value of molecular remission before HCT.21-23,31-34 Moreover, recent results from the MORPHO study demonstrated that FLT3-ITD MRDneg before and after HCT was associated with improved outcomes and only patients who were MRD positive significantly benefited from maintenance treatment with gilteritinib.35 

The proportion of patients who achieved FLT3-ITD MRDneg status after Cy2/before HCT varied slightly across the published cohorts (63%,22 71%,21 and 86%23) and our cohorts (78%). Differences might be explained by patient characteristics and the tissues assessed, the various prevalence rates of concurrent NPM1mut, and/or the additional treatment with midostaurin in our cohort.

To our knowledge, this was the first study to evaluate the impact of FLT3-ITD MRD status over time and MRD conversion during early FU. In patients with FLT3-ITD MRD conversion (MRDneg to MRDpos) during FU, we observed subsequent relapses in 13 of 16 (81%) patients, which translated into a significantly increased relapse risk. However, the FLT3-ITD MRD status was not systematically evaluated during the FU period, and FLT3-ITD MRD monitoring for relapse surveillance needs to be confirmed in additional studies. The 3 patients who converted to MRDpos during FU but who remained in continuous remission exhibited very low VAF levels. This reflects the clinical challenge to interpret MRDpos status during FU and underscores the indispensability to discriminate molecular persistence at (very) low VAF level from molecular progression and molecular relapse by analyzing a second sample as recommended by the ELN MRD Working Party.16 

In line with the 2 previous studies, combined FLT3-ITD and NPM1mut MRD positivity was associated with a poor outcome.21,22 Favorable outcomes were observed for patients with FLT3-ITD MRDneg status irrespective of the NPM1mut MRD status. This implies that within FLT3-ITDpos/NPM1mut AML, the FLT3-ITD MRD status may further refine the individual prognosis by better discrimination of patients at high and low risk for relapse than when using NPM1mut MRD detection alone.

Considering the different assays used for FLT3-ITD MRD monitoring, an international consensus on standardizing the approach is needed, which is also currently discussed in the ELN MRD Working Party.

In summary, beyond the known risk factors, NGS-based FLT3-ITD MRD monitoring allows for the identification of patients at high risk for relapse and death. Currently, randomized studies that are comparing midostaurin and second-generation FLT3 inhibitors are ongoing (eg, NCT04027309). FLT3-ITD MRD analysis will be informative in assessing whether these more selective inhibitors will increase the depth of molecular remission and if these deeper responses are associated with an improved outcome.

The authors acknowledge Julia K. Herzig, Susanne Lux, and Laura K. Schmalbrock for technical support during next-generation sequencing data analysis and the members of the German-Austrian AML Study Group for providing patient samples and clinical information.

This work was supported, in part, by the Collaborative Research Center (SFB 1074), projects B3 (L.B. and K.D.), B12 (K.D.), and Z1 (H.D.).

Contribution: F.G.R., L.B., H.D., and K.D. designed the study; S.C., S.S., T.J.L., A.C., V.I.G., A.M., S.A., and F. Theis performed the experiments and validated the data; F.G.R., J.K., E.P., and A.B. performed the statistical analyses; F.G.R., D.W., F.S., A.S., W.F., H.R.S., G.W., H.S., T.S., K.S.G., M.W.M.K., M.L., R.F.S., F. Thol, M.H., A.G., H.D., and K.D. collected, assembled, analyzed and interpreted data; F.G.R., H.D., and K.D. wrote the first draft of the manuscript; and all authors undertook manuscript writing, editing and approval, revised the manuscript, and reviewed and approved the final version.

Conflict-of-interest disclosure: F.G.R. reports receiving honoraria from and serving as a consultant for Jazz Pharmaceuticals, Novartis, and Bristol Myers Squibb (BMS)/Celgene, and receiving travel support from Jazz Pharmaceuticals. L.B. reports receiving honoraria from AbbVie, Amgen, Astellas, BMS/Celgene, Daiichi Sankyo, Gilead, Janssen, Jazz Pharmaceuticals, Menarini, Novartis, Pfizer, Roche, and Sanofi, and receiving research support from Bayer and Jazz Pharmaceuticals. V.I.G. reports serving in an advisory role for Jazz Pharmaceuticals, AbbVie, and Boehringer Ingelheim; serving on the speakers' bureau of Pfizer, Janssen, and AbbVie; and receiving travel support from AbbVie. F.S. reports receiving honoraria from and serving as a consultant for AOP Orphan Pharmaceuticals, MorphoSys, BMS/Celgene, Incyte, Novartis, and Pfizer. W.F. reports receiving personal fees and nonfinancial support from AbbVie; receiving grants, personal fees, and nonfinancial support from Amgen and Pfizer; receiving personal fees from Jazz Pharmaceuticals, Celgene, MorphoSys, Incyte, Stemline Therapeutics, Clinigen, Daiichi Sankyo, Otsuka, and Servier outside the submitted work; receiving research support from Apis; filing a patent with Amgen; and receiving support for medical writing for Amgen, Pfizer, and AbbVie. H.S. reports receiving honoraria from AbbVie, Amgen, AstraZeneca, BMS/Celgene, Genzyme, GlaxoSmithKline, Janssen, Oncopeptides, Pfizer, Roche, Sanofi, Stemline Therapeutics, and Takeda, and travel expenses from Amgen, BMS/Celgene, Janssen, and Sanofi. K.S.G. reports serving in an advisory role for BMS, Jazz Pharmaceuticals, Pfizer, and AbbVie. M.W.M.K. reports receiving honoraria from and serving as a consultant for Pfizer, Kura Oncology, Jazz Pharmaceuticals, BMS/Celgene, and AbbVie, and serving on the speakers bureau of Gilead. M.L. reports serving in an advisory role for AbbVie, Astex Pharmaceuticals, Imago BioSciences, Janssen, Otsuka, and Syros, and receiving research support from Janssen and Cheplapharm. R.F.S. reports serving in an advisory role or as a consultant for Daiichi Sankyo, Pfizer, Astellas, and Novartis; receiving research funding from PharmaMar, AstraZeneca, Pfizer, Roche, Boehringer Ingelheim, Daiichi Sankyo, and Recordati; and receiving funding for travel, accommodation, and expenses from Daiichi Sankyo. F. Thol reports serving in an advisory role for Novartis, BMS, AbbVie, Menarini, and Rigel. M.H. reports receiving honoraria from Certara, Jazz Pharmaceuticals, Janssen, Novartis, and Sobi; serving as a paid consultant for AbbVie, Amgen, BMS/Celgene, Glycostem, Delbert Lab, Pfizer, Pinotbio, and Servier; and receiving research funding to his institution from AbbVie, Agios, Astellas, BMS/Celgene, Glycostem, Jazz Pharmaceuticals, Karyopharm, Loxo Oncology, and Pinotbio. H.D. reports serving in an advisory role for AbbVie, Agios, Amgen, Astellas, AstraZeneca, Berlin-Chemie, BMS/Celgene, Daiichi Sankyo, GEMoaB, Gilead, Janssen, Jazz Pharmaceuticals, Novartis, Servier, Stemline Therapeutics, and Syndax, and receiving research funding from AbbVie, Agios, Amgen, Astellas, BMS/Celgene, Jazz Pharmaceuticals, Kronos Bio, Novartis, and Pfizer. K.D. reports serving in an advisory role for Amgen, BMS/Celgene, Daiichi Sankyo, Janssen, Jazz Pharmaceuticals, Novartis, and Roche, and receiving research funding from Agios, Astex, Astellas, BMS/Celgene, and Novartis. The remaining authors declare no competing financial interests.

Correspondence: Konstanze Döhner, Department of Internal Medicine III, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89081 Ulm, Germany; email: konstanze.doehner@uniklinik-ulm.de.

1.
Marcucci
G
,
Haferlach
T
,
Döhner
H
.
Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications [published correction appears in J Clin Oncol. 2011;29(13):1798]
.
J Clin Oncol
.
2011
;
29
(
5
):
475
-
486
.
2.
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
.
3.
Grimwade
D
,
Ivey
A
,
Huntly
BJ
.
Molecular landscape of acute myeloid leukemia in younger adults and its clinical relevance
.
Blood
.
2016
;
127
(
1
):
29
-
41
.
4.
Bullinger
L
,
Döhner
K
,
Döhner
H
.
Genomics of acute myeloid leukemia diagnosis and pathways
.
J Clin Oncol
.
2017
;
35
(
9
):
934
-
946
.
5.
Kottaridis
PD
,
Gale
RE
,
Frew
ME
, et al
.
The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials
.
Blood
.
2001
;
98
(
6
):
1752
-
1759
.
6.
Thiede
C
,
Steudel
C
,
Mohr
B
, et al
.
Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis
.
Blood
.
2002
;
99
(
12
):
4326
-
4335
.
7.
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
(
5
):
2776
-
2784
.
8.
Pratcorona
M
,
Brunet
S
,
Nomdedéu
J
, et al
.
Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy
.
Blood
.
2013
;
121
(
14
):
2734
-
2738
.
9.
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
(
23
):
3441
-
3449
.
10.
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
(
12
):
2386
-
2392
.
11.
Rücker
FG
,
Du
L
,
Luck
TJ
, et al
.
Molecular landscape and prognostic impact of FLT3-ITD insertion site in acute myeloid leukemia: RATIFY study results
.
Leukemia
.
2022
;
36
(
1
):
90
-
99
.
12.
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
.
13.
Döhner
K
,
Thiede
C
,
Jahn
N
, et al
.
Impact of NPM1/FLT3-ITD genotypes defined by the 2017 European LeukemiaNet in patients with acute myeloid leukemia
.
Blood
.
2020
;
135
(
5
):
371
-
380
.
14.
Döhner
H
,
Wei
AH
,
Appelbaum
FR
, et al
.
Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN
.
Blood
.
2022
;
140
(
12
):
1345
-
1377
.
15.
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
.
16.
Heuser
M
,
Freeman
SD
,
Ossenkoppele
GJ
, et al
.
2021 update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party
.
Blood
.
2021
;
138
(
26
):
2753
-
2767
.
17.
Kapp-Schwoerer
S
,
Weber
D
,
Corbacioglu
A
, et al
.
Impact of gemtuzumab ozogamicin on MRD and relapse risk in patients with NPM1-mutated AML: results from the AMLSG 09-09 trial
.
Blood
.
2020
;
136
(
26
):
3041
-
3050
.
18.
Dillon
R
,
Potter
N
,
Freeman
S
,
Russell
N
.
How we use molecular minimal residual disease (MRD) testing in acute myeloid leukaemia (AML)
.
Br J Haematol
.
2021
;
193
(
2
):
231
-
244
.
19.
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
.
20.
Blätte
TJ
,
Schmalbrock
LK
,
Skambraks
S
, et al
.
getITD for FLT3-ITD-based MRD monitoring in AML
.
Leukemia
.
2019
;
33
(
10
):
2535
-
2539
.
21.
Grob
T
,
Sanders
MA
,
Vonk
CM
, et al
.
Prognostic value of FLT3-internal tandem duplication residual disease in acute myeloid leukemia
.
J Clin Oncol
.
2023
;
41
(
4
):
756
-
765
.
22.
Loo
S
,
Dillon
R
,
Ivey
A
, et al
.
Pretransplant FLT3-ITD MRD assessed by high-sensitivity PCR-NGS determines posttransplant clinical outcome
.
Blood
.
2022
;
140
(
22
):
2407
-
2411
.
23.
Dillon
LW
,
Gui
G
,
Page
KM
, et al
.
DNA sequencing to detect residual disease in adults with acute myeloid leukemia prior to hematopoietic cell transplant
.
JAMA
.
2023
;
329
(
9
):
745
-
755
.
24.
Schlenk
RF
,
Weber
D
,
Fiedler
W
, et al;
German-Austrian AML Study Group
.
Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD
.
Blood
.
2019
;
133
(
8
):
840
-
851
.
25.
Döhner
H
,
Weber
D
,
Krzykalla
J
, et al
.
Midostaurin plus intensive chemotherapy for younger and older patients with AML and FLT3 internal tandem duplications
.
Blood Adv
.
2022
;
6
(
18
):
5345
-
5355
.
26.
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
(
12
):
3740
-
3746
.
27.
Gray
RJ
.
A class of k-sample tests for comparing the cumulative incidence of a competing risk
.
Ann Stat
.
1988
;
16
(
3
):
1141
-
1154
.
28.
Schemper
M
,
Smith
TL
.
A note on quantifying follow-up in studies of failure time
.
Control Clin Trials
.
1996
;
17
(
4
):
343
-
346
.
29.
Cox
DR
.
Regression models and life-tables
.
J R Stat Soc Series B Stat Methodol
.
1972
;
34
(
2
):
187
-
202
.
30.
Perl
A
,
Erba
HP
,
Montesinos
P
, et al
.
QuANTUM-first trial: FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD)-specific measurable residual disease (MRD) clearance assessed through induction (IND) and consolidation (CONS) is associated with improved overall survival (OS) in newly diagnosed (nd) FLT3-ITD+ AML patients (pts)
.
Blood
.
2023
;
142
(
suppl 1
):
832
.
31.
Gaballa
S
,
Saliba
R
,
Oran
B
, et al
.
Relapse risk and survival in patients with FLT3 mutated acute myeloid leukemia undergoing stem cell transplantation
.
Am J Hematol
.
2017
;
92
(
4
):
331
-
337
.
32.
Thol
F
,
Gabdoulline
R
,
Liebich
A
, et al
.
Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML
.
Blood
.
2018
;
132
(
16
):
1703
-
1713
.
33.
Dillon
R
,
Hills
R
,
Freeman
S
, et al
.
Molecular MRD status and outcome after transplantation in NPM1-mutated AML
.
Blood
.
2020
;
135
(
9
):
680
-
688
.
34.
Helbig
G
,
Koclęga
A
,
Wieczorkiewicz-Kabut
A
, et al
.
Pre-transplant FLT3/ITD status predicts outcome in FLT3-mutated acute myeloid leukemia following allogeneic stem cell transplantation
.
Ann Hematol
.
2020
;
99
(
8
):
1845
-
1853
.
35.
Levis
MJ
,
Hamadani
M
,
Logan
B
, et al
.
Gilteritinib as post-transplant maintenance for AML with internal tandem duplication mutation of FLT3
.
J Clin Oncol
.
2024
;
42
(
15
):
1766
-
1775
.

Author notes

Presented in part in abstract form and as a poster presentation at the virtual edition of the 25th annual congress of the European Hematology Association, 11 to 21 June 2020.

Presented in part in abstract form and as an oral presentation at the virtual edition of the 62nd annual meeting of the American Society of Hematology, 5 to 8 December 2020.

Presented in part in abstract form and as a poster presentation at the virtual edition of the 26th annual congress of the European Hematology Association, 9 to 17 June 2021.

Presented in part in abstract form and as an oral presentation at the virtual edition of the 28th annual congress of the European Hematology Association, 8 to 11 June 2023.

Original data are available from the corresponding author, Konstanze Döhner (konstanze.doehner@uniklinik-ulm.de). Individual participant data will not be shared.

The full-text version of this article contains a data supplement.