TO THE EDITOR:

The presence of measurable residual disease (MRD) in patients with acute myeloid leukemia (AML) is an important prognostic factor in all stages of the disease.1-5  In particular, patients in a morphologic remission who still have detectable MRD prior to allogeneic transplant have an outcome similar to patients with morphologically persistent disease pretransplant, at least when conventional polymerase chain reaction (PCR)- and flow cytometry–based techniques are used to identify MRD. However, the molecular heterogeneity of AML has rendered it challenging to develop any assay that can consistently detect residual disease, and most methods available today lack sensitivity beyond 1 cell in 100.

In the randomized trial of chemotherapy plus or minus the FLT3 inhibitor midostaurin for FLT3-mutant AML (CALGB10603/RATIFY), patients who underwent allogeneic transplant in remission after treatment with chemotherapy plus midostaurin had a markedly better outcome posttransplant compared with patients on the control arm who were transplanted after achieving remission with chemotherapy alone.6  The assumption has been that patients on the midostaurin arm had a lower level of MRD, although the means to determine this have thus far not been available. We have developed a highly sensitive combination PCR–next-generation sequencing (NGS) MRD assay for FLT–internal tandem duplication (ITD) mutations and validated this assay using paired diagnosis and remission samples from FLT3-ITD AML patients. Our results offer clear support for the aforementioned assumptions regarding the RATIFY trial.

The assay is depicted in supplemental Figure 1A, available on the Blood Web site. PCR (10 cycles) was performed using primers flanking exons 13 and 15 to amplify the FLT3 gene. Highly diverse NGS libraries were generated using additional PCR cycles and sequenced using Illumina’s sequencing-by-synthesis method. Using a custom bioinformatics approach, unique FLT3-ITD mutations of varying lengths were identified, and mutant allelic frequencies were calculated. DNA from FLT3-ITD mutant clinical samples and cell lines (MV4-11, PL-21, and MOLM-13) was either tested neat or after being spiked into DNA derived from healthy blood donors to evaluate the assay’s sensitivity and linearity (supplemental Figure 1B).

We validated the assay using clinical samples from a series of 17 patients with FLT3-ITD AML (clinical features summarized in Table 1). All 17 patients had an FLT3-ITD mutation, a canonical C-terminal frameshift NPM1 mutation, and an intermediate risk karyotype. All patients received a single course of infusional cytarabine and anthracycline–based (eg, intensive) chemotherapy, and all achieved a complete remission conforming to international working group criteria.7  In all 17 samples, multiparameter flow cytometry detected no leukemia by immunophenotype in the remission samples. Although 7 patients received time-sequential induction therapy (AcIVP16; Table 1), previous studies have shown a comparable remission rate between 7 + 3 and time-sequential therapy for FLT3-ITD-mutated AML.8,9  The capillary electrophoresis (CE) PCR assay to detect FLT3-ITD mutations was negative in the remission sample for all 17 cases. In each case, this was after a single course of treatment, and the remission biopsy was performed a mean of 40 days (range 29 to -58 days) after the start of induction. Unfortunately, because these were banked DNA samples, an RNA-based quantitative PCR assay for NPM1 mutations could not be performed. All 17 patients underwent allogeneic transplant in first remission, and all had diagnostic and remission DNA samples banked in accordance with an institutional review board–approved protocol conforming to the Declaration of Helsinki. One specific criterion that we did not select for, but realized only in retrospect, was that 9 of these patients received chemotherapy alone, whereas 8 received chemotherapy plus an FLT3 tyrosine kinase inhibitor (gilteritinib-4; midostaurin-3; sorafenib-1). The investigators performing the MRD assay were blinded to the clinical data.

Table 1.

Patient characteristics and FLT3-ITD results

Patient numberAge/sexInduction therapyITD mutation by CE-PCR at diagnosisITD mutation by MRD assay at diagnosisITD mutation by MRD assay at remissionClinical outcome
Length, bpVAF, %Length, bpVAFLength, bpVAF
59/F AcIVP16 51 51 7.7 51 0.0017 Alive, disease free 5 y after diagnosis 
30 30 1.42 30 (ND) 
56/F AcIVP16 51 37 51 47.48 51 0.0027 Alive, disease free 5 y after diagnosis 
64/M AcIVP16 81 46 81 75.69 81 0.020 Alive, disease free 3 y after diagnosis 
67/M 7 + 3 + gilteritinib 21 21 3.93, 0.16 None detected Alive, disease free 1 y after diagnosis 
60 
67/F AcIVP16 18 18 11.09 18 0.0077 Alive, disease free 4 y after diagnosis 
30 0.10 30 (ND) 
39 0.35 39 (ND) 
56/M 7 + 3 + gilteritinib 51 81 51 85.77 51 0.06 Alive, disease free 2.5 y after diagnosis 
52/M AcIVP16 27 24 0.57 24 0.2 Relapsed with the 42-bp ITD, died of relapsed disease 2 y after diagnosis 
42 <5 27 5.68 27 0.099 
93 <5 42 0.75 42 0.1 
96 <5 63 0.20 63 0.22 
  93 7.02 93 (ND) 
  96 11.45 96 0.013 
31/F 7 + 3 + midostaurin 15 45 15 45.27 15 0.004 Alive, disease free 2 y after diagnosis 
72/F 7 + 3 + midostaurin 33 33 6.48, 0.32 33 0.005 Alive, disease free 2 y after diagnosis 
69 69 (ND) 
10 68/F 7 + 3 24 37 24 39.53 24 0.018 Alive, disease free 4 y after diagnosis 
11 57/F AcIVP16 15 45 15 44.39 15 0.04 Relapsed with the 15-bp ITD and died 1 y after diagnosis 
12 43/M 7 + 3 + midostaurin 60 30 60 38.36 60 0.008 Alive, disease free 2 y after diagnosis 
13 67/F 7 + 3 + gilteritinib 27 10 27 25.52, 1.87 27 0.003 On gilteritinib maintenance after transplant, relapsed with AML lacking ITD 
108 108 (ND) 
14 47/M 7 + 3 + sorafenib 72 72 30.97 72 0.005 Died of GVHD 19 mo after diagnosis 
15 68/F 7 + 3 45 31 45 46.7 45 2.17 Alive, disease free 4 y after diagnosis 
16 60/M 7 + 3 + gilteritinib 24 36 24 37.63 24 0.004 Alive, disease free 2 y after diagnosis 
17 44/F AcIVP16 48 72 48 70.37 48 0.031 Alive, disease free 5 y after diagnosis 
Patient numberAge/sexInduction therapyITD mutation by CE-PCR at diagnosisITD mutation by MRD assay at diagnosisITD mutation by MRD assay at remissionClinical outcome
Length, bpVAF, %Length, bpVAFLength, bpVAF
59/F AcIVP16 51 51 7.7 51 0.0017 Alive, disease free 5 y after diagnosis 
30 30 1.42 30 (ND) 
56/F AcIVP16 51 37 51 47.48 51 0.0027 Alive, disease free 5 y after diagnosis 
64/M AcIVP16 81 46 81 75.69 81 0.020 Alive, disease free 3 y after diagnosis 
67/M 7 + 3 + gilteritinib 21 21 3.93, 0.16 None detected Alive, disease free 1 y after diagnosis 
60 
67/F AcIVP16 18 18 11.09 18 0.0077 Alive, disease free 4 y after diagnosis 
30 0.10 30 (ND) 
39 0.35 39 (ND) 
56/M 7 + 3 + gilteritinib 51 81 51 85.77 51 0.06 Alive, disease free 2.5 y after diagnosis 
52/M AcIVP16 27 24 0.57 24 0.2 Relapsed with the 42-bp ITD, died of relapsed disease 2 y after diagnosis 
42 <5 27 5.68 27 0.099 
93 <5 42 0.75 42 0.1 
96 <5 63 0.20 63 0.22 
  93 7.02 93 (ND) 
  96 11.45 96 0.013 
31/F 7 + 3 + midostaurin 15 45 15 45.27 15 0.004 Alive, disease free 2 y after diagnosis 
72/F 7 + 3 + midostaurin 33 33 6.48, 0.32 33 0.005 Alive, disease free 2 y after diagnosis 
69 69 (ND) 
10 68/F 7 + 3 24 37 24 39.53 24 0.018 Alive, disease free 4 y after diagnosis 
11 57/F AcIVP16 15 45 15 44.39 15 0.04 Relapsed with the 15-bp ITD and died 1 y after diagnosis 
12 43/M 7 + 3 + midostaurin 60 30 60 38.36 60 0.008 Alive, disease free 2 y after diagnosis 
13 67/F 7 + 3 + gilteritinib 27 10 27 25.52, 1.87 27 0.003 On gilteritinib maintenance after transplant, relapsed with AML lacking ITD 
108 108 (ND) 
14 47/M 7 + 3 + sorafenib 72 72 30.97 72 0.005 Died of GVHD 19 mo after diagnosis 
15 68/F 7 + 3 45 31 45 46.7 45 2.17 Alive, disease free 4 y after diagnosis 
16 60/M 7 + 3 + gilteritinib 24 36 24 37.63 24 0.004 Alive, disease free 2 y after diagnosis 
17 44/F AcIVP16 48 72 48 70.37 48 0.031 Alive, disease free 5 y after diagnosis 

All 17 patients had normal/intermediate karyotype and NPM1 mutation at diagnosis. All 17 patients achieved complete remission after a single cycle of induction therapy, and all underwent allogeneic transplant in first remission. Chemotherapy regimens: AcIVP16 = Cytarabine 2000 mg/m2 continuous infusion days 1 to 3, idarubicin 8 mg/m2 per day on days 1 to 3, etoposide 400 mg/m2 per day on days 8 to 10; 7 + 3 = cytarabine 100 to 200 mg/m2 per day on days 1 to 7, idarubicin 12 mg/m2 per day on days 1 to 3.

GVHD, graft-versus-host disease; ND, not detectable; VAF, variant allele frequency.

The MRD assay results were compared with the conventional CE PCR assay results for diagnostic and remission time points. At diagnosis, the MRD assay detected 100% of the ITD mutations detected by the CE assay, and in 6 of the 17 cases, it detected additional (1 to 6) ITD mutations (Table 1). In the remission samples, the MRD assay detected persistence of the diagnostic ITD mutation in 16 out of 17 cases. Although the %ITD levels between diagnosis and remission were statistically significant in all patients (Figure 1; P value 6.315e-06 to 7.78e-05 using Wilcoxon rank sum test), more striking was the fact that in the 8 patients treated with chemotherapy plus an FLT3 inhibitor during induction, the average level of ITD mutation was significantly lower compared with the 9 patients treated with chemotherapy alone (see Figure 1 for %ITD level values; P = .0078 by the nonparametric median test). In addition to the median test, a Wilcoxon rank sum test corroborated the statistical significance between these groups (P = .04563). The 1 patient (Table 1 patient 4) who had no detectable ITD mutation in the remission sample had received 7 + 3 (cytarabine 100 mg/m2 per day for 7 days, idarubicin 12 mg/m2 per day for 3 days) plus 120 mg/d of gilteritinib from days 4 to 17 of induction. The time between the start of induction and remission/MRD assessment was similar between the 2 groups (inhibitor group: mean 40.5 days; no inhibitor group: mean 40 days). Although this data set is small, these findings are consistent with the concept that when FLT3 inhibition is combined with induction chemotherapy, the result is a deeper remission, which may translate into better posttransplant survival.

Figure 1.

FLT3-ITD levels before and after induction. Banked genomic DNA samples from the patients listed in Table 1 were analyzed by MRD assay. (A) One-way analysis of the difference in FLT3-ITD levels at remission, comparing patients treated with chemotherapy only with patients treated with chemotherapy plus an FLT3 inhibitor. (B) Graphic comparison of an FLT3-ITD level decrease from diagnosis to remission. Although the difference in %ITD levels between induction and postinduction is statistically significant in both groups (P value 6.315e-06 for −FLT3, and P value 7.78e-05 for patients with chemotherapy plus FLT3 inhibitor using Wilcoxon rank sum test), the %ITD levels in patients with chemotherapy only are much higher than in the former group, and the difference between such groups at the postinduction level is significant (P value .04563 on a Wilcoxon rank sum test).

Figure 1.

FLT3-ITD levels before and after induction. Banked genomic DNA samples from the patients listed in Table 1 were analyzed by MRD assay. (A) One-way analysis of the difference in FLT3-ITD levels at remission, comparing patients treated with chemotherapy only with patients treated with chemotherapy plus an FLT3 inhibitor. (B) Graphic comparison of an FLT3-ITD level decrease from diagnosis to remission. Although the difference in %ITD levels between induction and postinduction is statistically significant in both groups (P value 6.315e-06 for −FLT3, and P value 7.78e-05 for patients with chemotherapy plus FLT3 inhibitor using Wilcoxon rank sum test), the %ITD levels in patients with chemotherapy only are much higher than in the former group, and the difference between such groups at the postinduction level is significant (P value .04563 on a Wilcoxon rank sum test).

Close modal

FLT3-ITD mutated AML is a common variant of this leukemia with an obvious, trackable genetic lesion, an ITD within the juxtamembrane domain.10  Use of this mutation for monitoring MRD has a disadvantage in that it is not stable: it can occasionally be lost at relapse, or, alternatively, it is detected only at relapse.11,12  However, it has the advantage of specificity: each patient’s mutation tends to be unique in size (ranging from 3 to >200 bp in length) and insertion site. It therefore acts as a mutational signature of the patient’s AML, particularly because these mutations tend to be the final “hit” in leukemogenesis.13  Sensitive detection of FLT3-ITD mutations in remission bone marrow specimens using conventional PCR is hampered by template bias, in which the reaction enzymes preferentially amplify the shorter wild-type sequence.14,15  Conventional NGS approaches are generally designed to detect point mutations or short insertions/deletions and likewise lack sensitivity beyond what flow cytometry can routinely detect (eg, 0.1% to 1%). However, the novel combination assay that we describe here can detect a targetable mutation and potentially influence treatment decisions. Although we have presented evidence that the assay is very sensitive, its positive predictive value is undetermined. Furthermore, we focused on a chemo-responsive group of patients: those with a concomitant NPM1 mutation. The clinical impact of this assay might be different in patients harboring more adverse genotypes. Therefore, the next questions (answered, it is hoped, from ongoing randomized clinical trials) include whether this type of assay can identify patients at risk of relapse, and potentially to validate the utility of maintenance therapy with FLT3 inhibitors. In addition, more refined flow cytometry methods are in development that will no doubt complement this PCR-NGS approach.

This assay will be used to measure MRD levels in subjects enrolled on Quantum-First (#NCT02668653), a phase 3 trial of patients with FLT3-ITD AML randomized to receive chemotherapy plus the FLT3 inhibitor quizartinib or placebo. A similar assay is being used in BMT-CTN1506 (“Morpho”), a randomized trial of gilteritinib vs placebo as posttransplant maintenance therapy for FLT3-ITD AML.16 

Further data are available by e-mailing the corresponding author.

Contribution: M.L. designed the study, analyzed the data, and wrote the manuscript; C.G. and E.A. contributed samples and helped edit the manuscript; W.S., C.L., R.P., and J.L. developed the NGS assay, contributed to study design, analyzed data, and helped edit the manuscript; and K.C., F.B., and A.L. contributed to study design and helped edit the manuscript.

Conflict-of-interest disclosure: M.L. serves as a consultant for Daiichi-Sankyo, Novartis, Astellas, Amgen, Arog, and Agios. W.S., C.L., R.P., and J.L. are employees of Navigate BioPharma Services, Inc. K.C., F.B., and A.L. are employees of Daiichi-Sankyo. The remaining authors declare no competing financial interests.

Correspondence: Mark Levis, Kimmel Cancer Center at Johns Hopkins, 1650 Orleans St, Room 2M44, Baltimore, MD 21287; e-mail: levisma@jhmi.edu.

1.
Walter
RB
,
Buckley
SA
,
Pagel
JM
, et al
.
Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission
.
Blood
.
2013
;
122
(
10
):
1813
-
1821
.
2.
Terwijn
M
,
van Putten
WL
,
Kelder
A
, et al
.
High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia: data from the HOVON/SAKK AML 42A study
.
J Clin Oncol
.
2013
;
31
(
31
):
3889
-
3897
.
3.
Jourdan
E
,
Boissel
N
,
Chevret
S
, et al;
French AML Intergroup
.
Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia
.
Blood
.
2013
;
121
(
12
):
2213
-
2223
.
4.
Ivey
A
,
Hills
RK
,
Simpson
MA
, et al;
UK National Cancer Research Institute AML Working Group
.
Assessment of minimal residual disease in standard-risk AML
.
N Engl J Med
.
2016
;
374
(
5
):
422
-
433
.
5.
Jongen-Lavrencic
M
,
Grob
T
,
Hanekamp
D
, et al
.
Molecular minimal residual disease in acute myeloid leukemia
.
N Engl J Med
.
2018
;
378
(
13
):
1189
-
1199
.
6.
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
.
7.
Cheson
BD
,
Bennett
JM
,
Kopecky
KJ
, et al;
International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia
.
Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia [published correction appears in J Clin Oncol. 2004;22(3):576]
.
J Clin Oncol
.
2003
;
21
(
24
):
4642
-
4649
.
8.
Boissel
N
,
Cayuela
JM
,
Preudhomme
C
, et al
.
Prognostic significance of FLT3 internal tandem repeat in patients with de novo acute myeloid leukemia treated with reinforced courses of chemotherapy
.
Leukemia
.
2002
;
16
(
9
):
1699
-
1704
.
9.
Norsworthy
KJ
,
DeZern
AE
,
Tsai
HL
, et al
.
Timed sequential therapy for acute myelogenous leukemia: Results of a retrospective study of 301 patients and review of the literature
.
Leuk Res
.
2017
;
61
:
25
-
32
.
10.
Daver
N
,
Schlenk
RF
,
Russell
NH
,
Levis
MJ
.
Targeting FLT3 mutations in AML: review of current knowledge and evidence
.
Leukemia
.
2019
;
33
(
2
):
299
-
312
.
11.
Kottaridis
PD
,
Gale
RE
,
Langabeer
SE
,
Frew
ME
,
Bowen
DT
,
Linch
DC
.
Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors
.
Blood
.
2002
;
100
(
7
):
2393
-
2398
.
12.
Shih
LY
,
Huang
CF
,
Wu
JH
, et al
.
Internal tandem duplication of FLT3 in relapsed acute myeloid leukemia: a comparative analysis of bone marrow samples from 108 adult patients at diagnosis and relapse
.
Blood
.
2002
;
100
(
7
):
2387
-
2392
.
13.
Jan
M
,
Snyder
TM
,
Corces-Zimmerman
MR
, et al
.
Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia
.
Sci Transl Med
.
2012
;
4
(
149
):
149ra118
.
14.
Polz
MF
,
Cavanaugh
CM
.
Bias in template-to-product ratios in multitemplate PCR
.
Appl Environ Microbiol
.
1998
;
64
(
10
):
3724
-
3730
.
15.
Murphy
KM
,
Levis
M
,
Hafez
MJ
, et al
.
Detection of FLT3 internal tandem duplication and D835 mutations by a multiplex polymerase chain reaction and capillary electrophoresis assay
.
J Mol Diagn
.
2003
;
5
(
2
):
96
-
102
.
16.
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
.

Author notes

The online version of this article contains a data supplement.

Supplemental data

Sign in via your Institution