Next Article in Journal
Metformin, Empagliflozin, and Their Combination Modulate Ex-Vivo Macrophage Inflammatory Gene Expression
Next Article in Special Issue
Understanding the Continuum between High-Risk Myelodysplastic Syndrome and Acute Myeloid Leukemia
Previous Article in Journal
A Unified Deep Learning Framework for Single-Cell ATAC-Seq Analysis Based on ProdDep Transformer Encoder
Previous Article in Special Issue
Solute Carrier Family 29A1 Mediates In Vitro Resistance to Azacitidine in Acute Myeloid Leukemia Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Measurable Residual Disease (MRD) in Acute Myeloid Leukemia (AML): Moving beyond Prognostication

1
Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
2
The Alfred Hospital, Melbourne, VIC 3004, Australia
3
Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia
4
Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
5
The Northern Hospital, Epping, VIC 3076, Australia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(5), 4790; https://doi.org/10.3390/ijms24054790
Submission received: 8 January 2023 / Revised: 1 February 2023 / Accepted: 2 February 2023 / Published: 1 March 2023

Abstract

:
Measurable residual disease (MRD) assessment in acute myeloid leukemia (AML) has an established role in disease prognostication, particularly in guiding decisions for hematopoietic cell transplantation in first remission. Serial MRD assessment is now routinely recommended in the evaluation of treatment response and monitoring in AML by the European LeukemiaNet. The key question remains, however, if MRD in AML is clinically actionable or “does MRD merely portend fate”? With a series of new drug approvals since 2017, we now have more targeted and less toxic therapeutic options for the potential application of MRD-directed therapy. Recent approval of NPM1 MRD as a regulatory endpoint is also foreseen to drastically transform the clinical trial landscape such as biomarker-driven adaptive design. In this article, we will review (1) the emerging molecular MRD markers (such as non-DTA mutations, IDH1/2, and FLT3-ITD); (2) the impact of novel therapeutics on MRD endpoints; and (3) how MRD might be used as a predictive biomarker to guide therapy in AML beyond its prognostic role, which is the focus of two large collaborative trials: AMLM26 INTERCEPT (ACTRN12621000439842) and MyeloMATCH (NCT05564390).

1. Introduction

Although intensive chemotherapy induces complete remission (CR) in approximately 70% of AML cases, 30–80% relapse within the first 2 years, representing a major barrier to long-term cure in AML [1]. The assessment of AML disease response status has been traditionally based on a 5% threshold after morphologic assessment of ~200–500 cells under the microscope; however, leukemia cells could remain at 1 in >106. The assessment of subclinical levels of leukemia, namely measurable residual disease (MRD), provides an integrated assessment beyond baseline characteristics, such as pharmacokinetic resistance, therapeutic sensitivity, immune microenvironment, and other patient and external factors. MRD assessment is now routinely recommended in the evaluation of treatment response and monitoring in AML by the European LeukemiaNet (ELN), using multiparameter flow cytometry MRD (MFC-MRD) based on the combination of diagnostic leukemia-associated immunophenotype (LAIP) and different from normal (DfN) immunophenotype, and validated molecular markers, namely NPM1, RUNX1::RUNX1T1, CBFB::MYH11, and PML::RARA [2,3,4]. Notably, NPM1 MRD was recently approved as a regulatory approval endpoint in a clinical trial (NCT05020665) [5], paving the way for future MRD biomarker-driven trial designs and accelerated drug approvals.
Persistent somatic mutations in morphologic remission are increasingly used as “MRD” markers but often overlap with those observed in clonal hematopoiesis of indeterminate potential (CHIP) and persist in morphologic remission at high variant allele frequencies (VAF) in the preleukemic clones. Notably, FLT3-ITD is increasingly recognized as an important MRD marker, despite its typical late occurrence during leukemogenesis and presence within the leukemic subclone [6]. Whilst the adverse prognostic impact of MRD after intensive chemotherapy and prior to allogeneic hematopoietic cell transplantation (HCT) is well established [7,8], current MRD timepoints, kinetics, and thresholds require further validation in the setting of novel therapies that have become available since 2017 (see recent reviews) [9,10]. Moreover, the role of MRD-directed therapy (i.e., clinical actionability) in AML remains debatable [11,12,13,14], unlike the use of arsenic trioxide in acute promyelocytic leukemia (APL) and blinatumomab in B-cell acute lymphoblastic leukemia (ALL) [15,16].
In this article, we will review (1) the emerging molecular MRD markers, (2) the impact of novel therapeutics on MRD endpoints, and (3) how MRD might be used as a predictive biomarker to guide therapy in AML beyond its prognostic role.

2. Emerging MRD Markers

2.1. AML-Defining Molecular MRD Markers

Traditionally, MRD assessment aims to measure leukemic blasts below the 5% threshold by the morphologic assessment, detect residual disease, and predict relapse. An ideal molecular MRD marker should be exclusively present within the AML-defining clone, absent (or nearly absent) in the preleukemic state (such as CHIP or myelodysplastic syndrome [MDS]), correlate with therapeutic response, recur at disease relapse, and be technically feasible for molecular tracking. Refer to Table 1 for a summary of the characteristics of AML-defining molecular MRD markers. AML with recurrent genetic abnormalities, as defined by the WHO entities, are the prototypical examples, and some subtypes could define AML regardless of blast count.
The latest WHO classification has expanded the list of recurrent genetic abnormalities that define AML regardless of blast count: five specific fusions (PML::RARA, RUNX1::RUNX1T1, CBFB::MYH11, DEK::NUP214, and RBM15::MRTFA), three groups of gene rearrangement (KMT2A, MECOM, and NUP98) and one gene mutation (NPM1). Whilst these are all potential MRD markers, not all are applied in routine clinical practice due to rare entities, multiple translocation partners (such as KMT2A), and rare transcript isoforms (such as non-type A/B/D NPM1). It is also worth noting that inv(3), whilst a defining genetic abnormality, is not a gene fusion but rather results in the reposition of a distal GATA2 enhancer to activate MECOM expression. MRD monitoring for t(9;11)/KMT2A::MLLT3, the most common fusion among this subgroup, could be particularly helpful to risk further stratification, as this is typically considered intermediate-risk cytogenetics [4,17]. Achievement of MRD negativity for t(9;11) appears to be a pre-requisite for long-term remission and might be useful for early detection of relapse [18]. Data on other KMT2A::X fusions also support the utility of MRD monitoring among patients with KMT2A fusions, now with increasing relevance with the use of menin inhibitors, which have been shown to induce clinical and MRD responses [19]. Additionally, t(6;9)/DEK::NUP214, a high-risk lesion detected in ~1% of patients with AML, is also amenable to serial RT-qPCR to detect persistent or relapsing disease [20].

2.2. Persistent Clonal Hematopoiesis with Oncogenic Potential (CHOP)

AML is characterized by a stepwise acquisition of somatic mutations from clonal hematopoiesis to full transformation [21]. At diagnosis, each patient has a median of four to five mutations occurring in >70 genes [22]. Except for IDH1 Arg132 and IDH2 Arg140/R172 mutations, the heterogenous molecular landscape most typically requires a targeted next-generation sequencing panel approach (NGS-MRD). A common example observed in de novo AML is the successive occurrence of a mutation in epigenetic regulation (such as DNMT3A and TET2), followed by an AML-defining mutation (such as NPM1), and lastly, a mutation involved in signaling pathways (such as FLT3, NRAS, and KRAS) [6]. These early mutations, also known as preleukemic mutations, overlap with age-related clonal hematopoiesis and often persist in morphologic remission [23]. Whilst sometimes termed “MRD”, the persistence of these mutations is more accurately termed clonal hematopoiesis (CH), and the clinical challenge is to identify CH with oncogenic potential (CHOP) and distinguish this from CHIP (refer to Table 1).
Despite the caveat of persistent mutations overlapping with CHIP, two landmark studies have found that the persistence of mutations, particularly those other than DTA, is associated with an increased risk of disease relapse, including those persisting at high VAF consistent with preleukemic origin [24,25]. Other studies evaluating the role of NGS-MRD are summarized in Table 2 [23,24,25,26,27,28,29,30,31,32,33]. In studies comparing both NGS-MRD and MFC-MRD, both modalities are found to be complementary, with decremental outcomes observed for MFCneg/NGSneg > MFCneg/NGSpos ≈ MFCpos/NGSneg > MFCpos/NGSpos [23,25,31,32]. The adverse prognostic impact of detectable mutations by NGS might be abrogated by allogeneic HCT in the first remission [25]. Conditioning intensity has been shown to have an impact on the post-HCT outcome of patients with NGS-MRDpos pre-HCT. In patients with no mutations detected, overall survival (OS) did not differ based on conditioning intensity; however, in those with detectable mutations, survival was significantly worse in those who received reduced-intensity conditioning [29,33].
The above findings have led to the ELN recommendation that DTA mutations “should be excluded from MRD analysis” [2]. This notion, however, was recently challenged by the ALFA0702 study, including 181 patients (out of 616 achieving CR/CR with incomplete hematologic recovery [CRi]) aged 18–60 years with de novo AML, available DNA sample, and at least one mutation detected at diagnosis. Using an UMI-corrected NGS-MRD panel (TWIST) with a sensitivity of 0.1% VAF, the authors found that persistent DTA mutations were associated with a significantly adverse cumulative incidence of relapse (CIR), event-free survival (EFS), and OS. The 4-year CIR in NGS-MRDneg (n = 91, 50%) vs. DTA mutations only (n = 42, 23%) vs. NGS-MRDpos (n = 48, 27%) were 68%, 51%, and 39%, and the 4-year OS were 80%, 59%, and 54%, respectively [34]. The potential reasons for this discrepancy from prior studies (such as Jongen-Lavrencic et al. [HOVON-SAKK]) [23] regarding the DTA mutations might be related to patient selection (median age 46 years in ALFA0702 vs. 51 years in HOVON-SAKK), treatment protocols, NGS strategies (UMI corrected vs. non-UMI), and assessment timepoints (post-course 1 vs. 2).
Recurrent hotspot mutations affecting IDH1 Arg132 and IDH2 Arg140/Arg172 occur in approximately 20% of AML cases, and whilst their relevance as molecular MRD markers remain to be determined, they are of particular importance due to the availability of targeted inhibitors [22,35,36]. Whilst IDH1/2 mutations are early events in AML, they are rare (0.01%) in CHIP [37,38] and, when detected, were associated with a very high risk (15 out of 15) of subsequent progression to AML [39]. Earlier studies demonstrated that persistent IDH1/2 mutations after chemotherapy was significantly associated with inferior disease-free survival (3-year disease-free survival 38% vs. 62%) [40] and an increased risk of relapse (59% vs. 24% at 1 year) [41] with no significant impact on OS in both studies. On the contrary, Cappelli et al. studied 150 patients with NPM1 mutant (mut) AML in remission and suggested that IDH1, IDH2, SRSF2, and DTA mutations could be considered together as CHIP-like mutations and did not adversely affect the prognosis. The numbers were small however; 36 patients had mutated IDH1/2, of whom only eight had persistence in the mutation [42]. Among patients who had an initial clearance of IDH1/2mut clone after therapy, a progressive rise in the IDH1/2 clone size might portend AML relapse [43]. Although the three hotspot mutations in IDH1/2 are often considered together, their prognostic impact might differ. In an analysis of patients who received HCT by Bill et al., IDH2 Arg140 mutations had higher VAF at diagnosis (~50%), lower mutation clearance in morphologic remission, did not have a significant prognostic impact, and thus behaved more like a CHIP-related mutation, while IDH1 Arg132 and IDH2 Arg172 were more CHOP-like, including an association of increased risk of relapse [44].

2.3. Signaling Pathway Mutations, Focusing on FLT3-ITD

Gene mutations in the signaling pathway, such as FLT3 (ITD and TKD), NRAS, KRAS, and KIT, among others, typically occur late in the leukemogenesis and are often subclonal. Hence, whilst detection of these mutations represents residual AML, its absence has a low negative predictive value as AML relapse could occur without these mutations. For example, FLT3-ITD could be either lost or gained at the time of AML relapse in 20–30% and 5–10%, respectively [45,46]. Current ELN guidelines recommend that these signaling pathway mutations “are best used in combination with additional MRD markers” [3]. Despite these limitations, several studies have emerged to demonstrate the adverse prognostic impact of FLT3-ITD using a highly sensitive NGS-MRD assay, which will be summarized in this section. Other signaling pathway mutations are frequently considered together with other persistent mutations (Section 2.2) and will not be reviewed here, but it is worth noting their role in therapeutic resistance among patients treated with an IDH inhibitor [47], FLT3 inhibitor [48], and venetoclax (VEN) [49].
Conventional fragment analysis by capillary electrophoresis has limited sensitivity in the detection of FLT3-ITD at ~1% [50,51]. NGS detection of FLT3-ITD has had variable success due to the heterogeneous nature of FLT3-ITD mutations, including various insertion sites, insertion lengths (15–300 bp), insertion sequences, >1 mutant clone, and clonal evolution [52,53,54,55,56]. Standard bioinformatics algorithms are not optimized for the detection of larger insertions or deletions (indel). Amplicon-based NGS, with its uniform amplicon start-stop positions, can further complicate indel detection, resulting in discarded unaligned reads. Random fragmentation, such as in Illumina protocols, may break the target ITD sequences and render them undetectable [52,53]. More recently, a highly sensitive, specific, and proprietary FLT3-ITD NGS-MRD assay was developed by Invivoscribe® [54]. An open-source bioinformatic pipeline, getITD, has also allowed the assay to be more widely implemented in diagnostic laboratories [57].
Using NGS-MRD, we were among the first to demonstrate the adverse prognostic impact of detectable FLT3-ITD below the sensitivity of conventional capillary electrophoresis. Among 104 patients with FLT3-ITDmut AML undergoing HCT in morphologic CR, 38 (37%) had a detectable FLT3-ITD MRD pre-HCT, of which only seven were detectable by conventional capillary electrophoresis. Patients with any FLT3-ITD detectable ≥ 0.001% had an inferior 4-year CIR (67% vs. 16%, p < 0.001) and OS (≤26% vs. 74%, p < 0.0001) [58]. Similarly, Lee et al. identified that 28 (80%) of 35 patients at pre-HCT had FLT3-ITD detectable by NGS (sensitivity 0.001%) and identified a threshold of >0.1% to be most predictive of relapse in a multivariate analysis (HR 5.1, p = 0.008) [59]. In the pre-MEASURE study (abstract and preprint), using a targeted NGS-based MRD panel (sensitivity 0.01%), FLT3-ITD MRD was detectable in 85 of 608 patients (14%) at pre-HCT, which was again significantly associated with risk of relapse and inferior OS [33,60]. In the setting of post-intensive chemotherapy, Grob et al. found that FLT3-ITD NGS-MRD was detectable in 47 of 161 (29%) patients after two cycles of intensive chemotherapy and associated with an increased 4-year CIR (75% vs. 33%, p < 0.001) and inferior OS (31% vs. 57%, p < 0.001) [61]. FLT3-ITD NGS-MRD also exceeded the prognostic value of NPM1 NGS-MRD or MFC-MRD; the data are limited to a direct comparison with the NPM1 qPCR-MRD assessment [58].
The NGS-based assay also has additional advantages beyond its highly sensitive detection of FLT3-ITD. Small FLT3-ITD subclones and, thus, clonal heterogeneity are better detected by NGS, although its impact on relapse risk or survival is yet to be determined and is an area that requires further clarity [62,63]. The insertion site can also be determined from NGS. A beneficial effect of midostaurin appeared to be restricted to patients with sole juxtamembrane domain (JMD) insertion sites [64].

3. Impact of Novel AML Therapies on MRD Endpoints

After three to four decades of a stagnant therapeutic landscape, we finally saw a series of new drug approvals in AML since 2017 (see recent reviews) [9,10]: FLT3 (midostaurin and gilteritinib) and IDH inhibitors (ivosidenib and enasidenib), VEN, glasdegib, gemtuzumab ozogamicin, CPX-351, and oral azacitidine (AZA). In this section, we will review selected novel therapies with available data on MRD. Table 3 summarizes the impact of novel therapies on MRD endpoints.

3.1. VEN and Hypomethylating Agent (HMA)/Low Dose Cytarabine (LDAC)

The use of BCL-2 inhibitor VEN in combination with AZA or low dose cytarabine (LDAC) has changed the treatment landscape in patients with AML ineligible for intensive chemotherapy [65,66]. Improved response rates and OS (median 14.7 months vs. 9.6 months in VEN-AZA vs. AZA alone) from the pivotal phase 3 VIALE-A study led to the FDA approval of VEN-AZA in this population. Despite this, the duration of remission (DOR) was 18 months, and long-term OS was <20% [67]. In 164 patients treated with upfront VEN-AZA evaluable for MFC-MRD, 41% achieved MFC-MRDneg (<0.1%); the rate of MFC-MRDneg was highest (88%) among patients with an NPM1 mutation [68]. Among patients achieving MFC-MRDneg, approximately 50% achieved this by the end of cycle 4, with an additional 27% by the end of cycle 7 and the remainder beyond that. The median DOR and OS were not reached in patients with MFC-MRDneg < 0.1% (12-month estimates were 81.2% and 94.0%), versus the median DOR at 9.7 months and OS at 18.7 months (12-month estimates at 46.6% and 67.9%) in patients with MFC-MRDpos ≥ 0.1%. Other retrospective studies on the prognostic value of MFC-MRD are also consistent with those from the VIALE-A study [69,70].
Molecular MRD data among VEN-treated patients are comparatively more limited. Among those achieving durable remission of >2 years, sustained molecular negativity in NPM1mut MRD was observed (n = 4/4 evaluable), whereas molecular clearance of other mutations was variable [49]. In a retrospective analysis of 55 patients who received VEN in combination with AZA (n = 28), decitabine (DEC) (n = 1), or LDAC (n = 26) and who achieved CR/CRi and were evaluable for an NPM1 qPCR MRD response within the first 6 months, qPCR-MRDneg was achieved in 46%, ≥4 log10 reduction in 19%, and <4 log10 reduction in 35%. Achievement of qPCR-MRD < 0.005% (per 100 ABL1) was associated with a significantly improved OS (88% vs. 34% at 18 months) at a median follow-up time of 24.3 months, updated from <0.2% in the initially published abstract [71].
Other novel VEN-based combinations outside of AZA, DEC, or LDAC have resulted in high response rates, but longer follow-up is required to ascertain the durability of remission. VEN-cladribine-LDAC alternating with VEN-AZA was assessed in 60 patients with newly diagnosed AML unfit for intensive chemotherapy; the response rate was 93%, and MFC-MRDneg (<0.1%) was achieved in 84% of 51 patients with sample availability, resulting in a significant difference in 2-year OS compared with patients with MFC-MRDpos (80% vs. 45%, HR 3.97, p = 0.016) [72]. In attempts to improve the induction regimen, fludarabine, cytarabine, idarubicin, and granulocyte colony-stimulating factor (FLAG-IDA) combined with VEN in the newly diagnosed setting resulted in an 89% composite CR rate, of which 93% attained MFC-MRDneg (<0.1%) with demonstrable survival benefit (median OS NR vs. 16 months, p = 0.03) [73].
In the future, it is conceivable to apply MRD monitoring in the VEN-based low-intensity cohort to consider cessation of therapy in a select group of patients who achieve MRD negativity [74,75].

3.2. FLT3 Inhibitors

FLT3-ITD MRD response after the use of FLT3 inhibitors is of particular interest. The improved OS, despite similar protocol-specified CR in the RATIFY study, has been attributed to deeper MRD remission from the addition of midostaurin, but this has not been verified [76,77]. Among patients treated with midostaurin and intensive chemotherapy in the single-arm phase 2 AMLSG16-10 trial, significantly lower CIR (HR 0.1, p < 0.001) and favorable OS (HR 0.27, p < 0.03) were observed for 87% of patients who achieved FLT3-ITD NGS-MRDneg at the end of treatment [78].
The phase 3 ADMIRAL study led to the FDA approval of gilteritinib as the treatment for relapsed/refractory (R/R) FLT3mut (ITD and TKD) AML, where 34% of patients achieved CR or CR with partial hematologic recovery (CRh) compared with 15% in the conventional salvage chemotherapy group with improved OS (median 9.3 months vs. 5.6 months) [79]. Considering the FLT3-ITD NGS-MRD data from the phase 1/2 CHRYSALIS study [80], gilteritinib resulted in FLT3-ITD NGS-MRDneg (<0.01%) in 8 (16%) of 49 patients who achieved CR/CRi, which was associated with a longer OS (median 131.4 weeks vs. 43.3 weeks, p = 0.066). To further improve the response in R/R FLT3-mutated AML, the combination of VEN and gilteritinib was recently studied in 56 patients, of which 75% achieved a composite morphologic remission (including CR/CRi/CRp/MLFS): 15 (60%), 11 (44%) and 5 (20%) of 25 evaluable achieved an NGS-MRD < 1%, <0.1%, and 0.01%, respectively. However, OS among those with an FLT3-ITD NGS-MRD of <1% was not significantly different (median OS 11.6 months vs. 8.2 months, p = 0.16). More data are needed to conclusively determine the impact of FLT3-ITD NGS-MRD clearance on the outcomes of patients with R/R FLT3-mutated AML [81].
Preliminary data from the QuANTUM-First study showed that the upfront addition of quizartinib to intensive chemotherapy in patients with an FLT3-ITD mutant AML resulted in improved OS with placebo (median 31.9 months vs. 15.1 months, p = 0.03) and a lower post-induction FLT3-ITD NGS-MRD level (median VAF 0.01% vs. 0.03%, p = 0.02). FLT3-ITD MRD level < 0.01% post-induction, which was observed in 24.6% of quizartinib and 21.4% of placebo-treated patients, was associated with a longer OS (median OS NR vs. 29.4 months) irrespective of treatment arm. Interestingly, attaining an undetectable MRD level (<0.001%), which was observed in a higher proportion of patients on quizartinib (13.8% vs. 7.4%), did not seem to confer a significant survival benefit [82].
These studies highlight the feasibility and importance of the prospective incorporation of a highly sensitive FLT3-ITD NGS-MRD assessment in clinical trials. Further studies are required to further inform its kinetics and prognostic utility, particularly in the setting of FLT3 inhibitors and R/R AML, and to predict those with emerging therapeutic resistance when treated with other novel therapeutics.

3.3. IDH1 and IDH2 Inhibitors

Ivosidenib (IDH1 inhibitor) and enasidenib (IDH2 inhibitor) were both FDA-approved for R/R IDH-mutated AML based on the respective single-arm monotherapy studies [83,84,85]. Additionally, ivosidenib was also approved for newly diagnosed AML in unfit patients [83,86]. Ivosidenib monotherapy in R/R AML resulted in CR/CRh rates of 30%, of which 21% achieved IDH1-MRD clearance, associated with improved DOR (median 11.1 months vs. 6.5 months) and survival (median OS 14.5 months vs. 10.2 months) [83]. In the newly diagnosed setting and treatment with ivosidenib monotherapy, CR/CRh rate was 42.4%, of which the rate of IDH1-MRDneg was 64% [86]. Similarly, enasidenib monotherapy in R/R AML resulted in an overall response rate of 38.8% (including MLFS and partial remission), of which 12% achieved IDH2-MRDneg, associated with a significant survival benefit (median OS 22.9 months vs. 8.8 months, p = 0.0153) [85].
IDH inhibitors have also been studied in combination studies. In newly diagnosed patients with AML unfit for intensive chemotherapy (phase 3 AGILE study), ivosidenib and AZA resulted in higher rates of IDH1-MRDneg as compared with ivosidenib and a placebo (17/33 [51.5%] vs. 3/10 [30.0%]) [87]. All baseline co-mutations were also cleared among 73% of patients who achieved CR [88]. Similarly, an enasidenib–AZA combination in unfit newly diagnosed AML (phase 1b/2 AG221-AML-005) resulted in greater IDH2 mutation reduction; the median VAF was 0.023% in CR vs. 0.233% in CRi/CRp/PR/MLFS vs. 0.872% in no response [89]. Combining ivosidenib with VEN ± AZA in both newly diagnosed and R/R patients in a phase 1b/2 study resulted in IDH1-MRDneg and MFC-MRDneg in 67% and 63% of patients, respectively, resulting in significant improvement in survival (median OS NR vs. 8 months, p = 0.002) [90]. In newly diagnosed AML, ivosidenib or enasidenib in combination with intensive chemotherapy resulted in IDH1 and IDH2-MRDneg of 16/41 (39%) and 15/64 (23%) and MFC-MRDneg in 16/20 (80%) and 10/16 (63%) among patients achieving CR/CRi/CRp, respectively [91].

3.4. Gemtuzumab Ozogamicin (GO)

GO is a CD33 antibody–drug conjugate that has demonstrated survival benefits when added to standard intensive chemotherapy in patients with non-adverse karyotype AML [92,93]. AML with mutated NPM1 is one of the AML subtypes with the highest CD33 expression, which has been associated with the efficacy of GO [94]. Molecular MRD responses among patients with NPM1mut AML receiving upfront GO have been examined by the ALFA-0701 (n = 77) and AMLSG 09-09 trials (n = 469 evaluable out of 588) [95,96]. In the ALFA-0701 study, patients aged 50–70 years were randomized to 7 + 3 induction chemotherapies ± fractionated doses of GO, followed by two consolidation cycles with daunorubicin and cytarabine ± GO. The proportion of patients achieving NPM1 qPCR-MRD < 0.1% was significantly higher in patients treated with GO, both post-induction (39% vs. 7%, p = 0.006) and at the end of treatment (91% vs. 61%, p = 0.028) [95].
In the AMLSG 09-09 trial, patients received two cycles of induction therapy with ATRA, idarubicin, cytarabine, and etoposide, followed by up to three consolidation cycles of high dose cytarabine and ATRA. Patients randomized to GO received this on day 1 of two induction cycles and the first consolidation cycle; significantly lower NPM1 qPCR-MRD levels were found in BM and PB post-induction and maintained throughout subsequent treatment cycles. This was also reflected by a significantly lower 4-year CIR in patients who received GO (31.6% vs. 43.9%, p = 0.015) and a superior relapse-free survival (RFS) (60.5% vs. 48.9%; p = 0.028) [96].
In a smaller cohort of patients with core-binding factor (CBF) AML on the UK MRC AML15 trial, the addition of GO increased molecular MRD reduction after only one course of induction in patients with t(8;21), but this was not mirrored in the inv(16) cohort and with no perceptible effect on relapse or survival [97].

3.5. CC-486

CC-486, an oral AZA formulation, was granted FDA approval in 2020 as maintenance therapy in patients who are unable to complete intensive chemotherapy after achieving remission post-intensive induction, based on OS benefit in the phase 3 QUAZAR study [98]. This benefit was seen irrespective of the presence of MFC-MRDpos (≥0.1%) in 46% of patients at study entry. On serial assessment, CC-486 resulted in an increased rate of MFC-MRDneg (<0.1%) compared with placebo at 37% vs. 19%, with 76% of patients achieving this within 6 months of treatment [99]. Achievement of MFC-MRDneg (including 60 patients on CC-486 or placebo) in the study was associated with longer survival: median OS 41.3 months vs. 9.0 months and median RFS 20.4 months vs. 2.8 months (MRD-MFCneg vs. remained MFC-MRDpos). Post-hoc analyses of the NPM1mut subgroup showed that CC-486 was able to eradicate flow MRD (<0.1%) in 63% of patients vs. 33% in placebo (p = 0.051) and with longer duration of sustained MFC-MRDneg compared with placebo (15.6 months vs. 7.1 months; p = 0.006). Similarly, patients who were FLT3mut had a numerically higher rate of MRD conversion to MFC-MRDneg (50% in CC-486 vs. 18% in the placebo arm, p-value not provided) [100].
Table 3. Impact of novel therapies on MRD endpoints.
Table 3. Impact of novel therapies on MRD endpoints.
TherapyPopulationNMRD MarkerThresholds (% of N)TimepointOutcomes (neg vs. pos) or
(< vs. ≥ Threshold)
Venetoclax (VEN)
VEN-AZA [68]New AML164MFCBM < 0.1% (41)AnyMedian OS NR vs. 19 m
1 y OS 94% vs. 68%
VEN-DEC10 [69]New AML83MFCBM < 0.1% (54)1–4 mMedian OS 25 m vs. 7 m
(2 m timepoint)
VEN-AZA [70]New AML63MFCBM < 0.1% (38)1–3 m18 m CIR 13% vs. 57%
18 m OS 70% vs. 35%
Median OS 26.5 m vs. 10 m
VEN-HMA/LDAC [71]New AML55NPM1 cDNABM < 0.005% (49)≤6 m18 m OS 87% vs. 39%
VEN-CLAD-LDAC alternating with VEN-AZA [72]New AML51MFCBM < 0.1% (84)Any2 y OS 80% vs. 45%
Median DFS NR vs. 5.9 m
VEN-FLAG-IDA [73]New AML40MFCBM < 0.1% (93)AnyMedian OS NR vs. 16 m
FLT3 inhibitor
Quizartinib [82]New AML161FLT3-ITDBM < 0.01% (24.6)PC1Median OS NR vs. 29.4 m
Gilteritinib [80]R/R49FLT3-ITDBM < 0.01% (16)AnyMedian OS 131.4 weeks vs. 43.3 weeks
VEN-Gilteritinib [81]R/R25FLT3-ITDBM < 0.1%
(60; 20% < 0.01%)
AnyMedian OS 11.6 m vs. 8.2 m
IDH1/2 inhibitors
Ivosidenib + intensive chemotherapy [91]New AML41IDH1BM < 0.02–0.04% (39)AnyNot reported
20MFCBM < 0.1% (80)AnyNot reported
Ivosidenib [83]R/R34IDH1BM < 0.02–0.04% (21)AnyMedian OS 11.1 m vs. 6.5 m
Ivosidenib [86]New AML14IDH1BM < 0.02–0.04% (64)AnyNot reported
Ivosidenib + VEN ± AZA [90]New AML or R/R31IDH1BM < 0.1–0.25% (67)AnyMedian OS NR vs. 8 m
MFCBM < 0.1% (63)
Enasidenib + intensive chemotherapy [91]New AML64IDH2BM < 0.02–0.04% (23)AnyNot reported
Enasidenib [85]R/R101IDH2BM < 0.02–0.04% (12)AnyMedian OS 22.9 m vs. 8.8 m
Gemtuzumab ozogamicin
ALFA-0701 [95]New AML61NPM1 cDNABM < 0.1% (25)PC12 y CIR 21% vs. 55%
BM < 0.1% (78)EOT2 y CIR 45% vs. 67%
AMLSG 09-09 [96]New AML232NPM1 cDNABM reduction ≥ 3 log10 (87)PC24 y CIR 28.5% vs. 60%
PB neg (53)PC24 y CIR 18% vs. 53%
PB neg (78)EOT4 y CIR 28% vs. 70%
CC-486
CC-486 [99]AML in first remission236MFCBM < 0.1% (37)AnyMedian OS 41.3 m vs. 9.0 m
Median RFS 20.4 m vs. 2.8 m
Abbreviations: AZA, azacitidine; BM bone marrow; CLAD, cladribine; CIR, cumulative incidence of relapse; DEC10, ten-day decitabine; DFS, disease-free survival; ddPCR, digital droplet polymerase chain reaction; EOT, end of treatment; FLAG-IDA, fludarabine, cytarabine, idarubicin, and filgrastim; LDAC, low dose cytarabine; m, months; MFC, multiparameter flow cytometry; MRD, measurable residual disease; neg, negative; NR, not reached; OS, overall survival; PB, peripheral blood; PC1, post course 1; PC2, post course 2; pos, positive; RT-qPCR, reverse transcription quantitative polymerase chain reaction; RFS, relapse-free survival; and R/R, relapsed/refractory.

4. MRD-Guided Therapy in AML

MRD assessment improves risk stratification in AML beyond the baseline patient and disease characteristics [101,102], but the key question remains “does it merely portend fate”? In other words, is MRD a predictive biomarker for therapeutic targeting that translates to an improvement in RFS and/or OS after accounting for lead time bias when compared with intensive salvage chemotherapy (followed by HCT) in patients with frank hematologic relapse? The benefit of relapse prevention needs to be balanced with treatment toxicities. An ideal MRD marker (Table 1) will reliably predict relapse in ensuing weeks/months, allowing time for intervention (including donor preparation), in addition to being prognostic regarding an increased risk of relapse over the next months to years. In parallel, a clinician will be readier to pre-emptively treat an emerging relapse if the therapy has fewer toxicities (see Section 3 on the novel therapies). The use of MRD to guide HCT in CR1, particularly in patients with favorable and intermediate-risk AML, has been reviewed elsewhere: patient selection vs. deference, donor selection, conditioning intensity/regimen, and post-HCT strategies, including immunosuppressant, disease monitoring, and therapeutic intervention [103,104,105,106]. Despite an earlier retrospective study by Araki et al. showing similarly poor outcomes post-myeloablative HCT between those with MFC-MRDpos morphologic remission and active morphologic disease [107], more recent studies, including both retrospective (European Society for Blood and Marrow Transplantation registry) [108] and prospective randomized-controlled trials (BMT-CTN 0901 study) [29], demonstrated improved outcomes following myeloablative conditioning HCT in those with pre-HCT MRD positivity of various markers.
This section will review the MRD-guided non-HCT therapeutic approach (summarized in Table 4), noting that the strategy of MRD eradication/reduction pre-HCT (vs. direct HCT) is unproven but rapidly evolving.

4.1. MRD-Directed Therapy Using AZA

One of the earliest descriptions of MRD-directed therapy (excluding acute promyelocytic leukemia) was the use of AZA in 10 patients with NPM1 qPCR-MRD relapse or persistence > 1%, of which seven (70%) had an MRD response (≥1 log10 reduction) and remained in morphologic remission at a median follow up of 10 months (range 2–12) [109]. This concept was extended to the prospective phase 2 RELAZA2 study, where 53 patients with falling post-HCT donor chimerism < 80% or MRD transcript levels > 1% at any time post-chemotherapy or HCT were pre-emptively treated with AZA. Sixty percent of patients were NPM1mut, and overall, 31 (58%) patients had an MRD response, including 19 (36%) who achieved MRDneg. The study met its primary endpoint, with 58% of patients remaining relapse-free at 6 months from therapy initiation. Overall, the 2-year RFS was 46% [14].
The value of MRD-guided therapeutic decision-making was further highlighted in a retrospective analysis by Short et al., where 55 patients with MFC-MRD relapse either continued current therapy (n = 36; including three with no further therapy) vs. those who had a change of therapy to direct HCT (n = 9) or HMA-based treatment (n = 7). Survival outcomes were significantly better among those who had a change in therapy; the 5-year RFS was 31% vs. 5% (p = 0.01), and OS was 45% vs. 17% (p = 0.01) [111]. Among the seven patients who received HMA-based treatment, three achieved MRD negativity (followed by HCT in 2), one remained MRD positive (then HCT), and three relapsed.

4.2. MRD-Directed Therapy Using Intensive Chemotherapy

The CETLAM group performed NPM1 qPCR-MRD monitoring on 110 patients with ELN favorable NPM1mut AML in first remission (CR1) after standard chemotherapy, of which 33 patients experienced molecular failure, defined as failure to achieve NPM1-MRD ≤ 0.05% after consolidation therapy (n = 11) or MRD relapse (n = 22). MRD-directed therapy was at the discretion of the treating clinician: 13 direct HCT, 12 salvage chemotherapy or HMA (10 followed by HCT), and 8 had interim morphologic relapse [110]. Eighty percent of patients receiving MRD-directed intensive chemotherapy achieved negative MRD. HCT was realized in 70% and 52% of patients with molecular failure and morphologic relapse, respectively. A survival benefit was observed in patients treated with molecular failure with 2-year OS 86% vs. 42% (p = 0.0014). With the limitation of small numbers, no significant survival difference was observed between patients who received pre-transplant MRD-directed therapy compared with direct HCT. Important caveats include that the NPM1 qPCR-MRD threshold of 0.05% at the end of consolidation is considered MRD detectable at a low level and might not be associated with increased relapse risk [2], and NPM1 persistence after intensive chemotherapy has been shown to spontaneously resolve or persist at a low level in ~39% of patients [115].
In the NCRI AML17 trial, intensive chemotherapy (usually FLAG-IDA) was given to 27 patients with molecular relapse, of whom 16 (59%) achieved MRD negativity prior to HCT. This approach was at the discretion of the clinician. A comparison with HCT was not possible as only three other patients with molecular relapse directly proceeded to HCT without attempted MRD eradication [112].

4.3. MRD-Directed Therapy Using VEN

Several studies have applied VEN as MRD-directed therapy, predominantly in the NPM1mut cohort, which is recognized as a predictive biomarker of response to VEN [49]. In a small cohort of seven patients treated with VEN and AZA or LDAC for NPM1 qPCR-MRD relapse after prior intensive chemotherapy, MRDneg was achieved in 86% of patients within 1–2 cycles, with ongoing molecular remission at a median time of 10.8 months follow-up [11]. We further studied this in the prospective phase 2 VALDAC study, utilizing VEN-LDAC in 48 patients with either MRD relapse (n = 26; 20 were NPM1mut) or early morphologic relapse (n = 22). MRD response (≥1 log10 reduction) was achieved in 69% (after a median of 1 cycle), including 54% achieving MRDneg (after a median of 2 cycles), resulting in an estimated 2-year EFS of 55% and OS of 73% [12].
In a UK-wide program reporting on outcomes of VEN-based low-intensity combinations (AZA, LDAC, another agent, or monotherapy), 19 patients were treated with MRD failure (18 MRD relapse and one persistent MRD), with 84% achieving molecular remission. In an indirect comparison with 103 patients treated in parallel with a morphologic disease, patients treated with molecular MRD failure demonstrated superior OS (median 18.4 months vs. 7.1 months, p = 0.004) [113].

4.4. MRD-Directed Therapy Using FLT3 Inhibitors

The use of FLT3 inhibitors (gilteritinib, quizartinib, or sorafenib) was described in 48 patients with known FLT3-ITD at baseline, who experienced a molecular MRD failure: NPM1 (81%), NUP98::NSD1 (8%), DEK::NUP214 (6%), and one of each CBFB::MYH11 and RUNX1::RUNX1T1 (4%). MRD failure was defined as persistence > 2% (n = 6) or MRD relapse (n = 42). In 47 patients evaluable for response, MRD response was achieved in 64%, and 40% achieved MRDneg. Fifty percent of patients received subsequent HCT. Two-year OS and EFS were 80% and 73%, respectively [114].

4.5. Future Directions in MRD-Directed Therapy

The future direction in MRD-directed therapy comprises integrated and coordinated frameworks to ascertain, firstly, the clinical actionability of various markers and urgently assess the survival benefit of treating MRD failure to circumvent protracted research timelines, high cost of singular clinical trials, and high attrition rates. Two large clinical trials and initiatives with the goal of harmonizing MRD monitoring and assessing the role of pre-emptive therapy are currently underway. The AMLM26 INTERCEPT study (ACTRN12621000439842) is a platform trial in Australia and the US, has been actively recruiting since August 2022, and aims to examine novel therapies as MRD-directed intervention in patients in first/second remission with MRD response as a primary endpoint, and key secondary endpoints of RFS, time to and duration of MRD response and OS [116]. Refer to Figure 1 for the AMLM26 INTERCEPT trial schema. This trial is linked to a coordinated MRD monitoring framework guided by an MRD reference committee once patients enter the platform in first/second remission. If suspected MRD failure occurs, this is confirmed by centralized laboratories, and patients are then allocated to one of many biomarker-directed treatment arms; the same is applied to patients where morphologic failure eventuates during MRD monitoring. For example, a patient with NPM1mut MRD relapse will be allocated to VEN-LDAC, or if with FLT3-ITDmut MRD relapse, to VEN-gilteritinib. Close clonal tracking using molecular assays ± MFC-MRD or leukemic stem cell flow MRD employed to assess response and clonal evolution is incorporated. Patients who experience MRD or morphologic relapse can rotate to another biomarker-driven treatment arm in the study. The MyeloMATCH precision medicine clinical trial (NCT05564390) is an umbrella trial comprising four tiers, which aims to enroll patients with newly diagnosed AML/MDS, who are then followed throughout their treatment journey with planned activation in mid-2023 [117,118]. The highest tier (tier 4) is aimed at targeting MRD, with incorporated technologies such as MFC and duplex sequencing.
With the increasing use of novel agents in the MRD failure setting, it is imperative that ongoing efforts are focused on the identification of clonal evolution, recognition of treatment-emergent resistance mechanisms, and unraveling the intrinsic and extrinsic biological differences between leukemia at the stage of MRD vs. morphologic failure.

5. Conclusions

At this stage, emerging molecular MRD markers have not consistently yielded a high positive predictive value for relapse or oncogenic potential, and beyond technical optimization, which focuses on achieving a higher sensitivity and lower limit of detection, ongoing work will need to dissect the genetic heterogeneity of AML at single-cell level and assess if the putative MRD clone resides within the leukemic compartment in the MRD setting [119,120]. These efforts are currently prohibited by the high cost of multiomic technologies and technical complexity. The ultimate goal is that the right assay for the right mutation in the right scenario and at the right timepoint is used.
Our article has also shown that novel therapies have the capacity to induce high rates of MRD response in patients attaining morphologic remission when used in the bulk disease setting, with exploratory analyses demonstrating a survival benefit in patients attaining MRD negativity. Moving forward, the prognostic value of historical MRD thresholds at specific timepoints will require prospective revalidation in the setting of novel therapeutics, as many of the aforementioned studies are limited by small numbers for MRD analyses.
Finally, MRD-directed therapy in AML is now the mainstay of two large, harmonized, biomarker-driven trial initiatives aimed at the efficient evaluation of pre-emptive therapy using a multi-modal approach. Novel therapies have shown promise in inducing high MRD response rates and with the possibility of lower toxicity. Whilst HCT remains the gold standard for potential cure in AML, the area needing urgent resolution is the value of pre-HCT MRD-directed therapy in patients with pre-HCT MRD detection vs. direct to HCT and whether or not this will alter post-HCT fate. This will hopefully provide an answer to the age-old question of whether MRD-directed intervention in AML is just delaying the inevitable or if it will ultimately improve patients’ long-term outcomes.

Author Contributions

I.S.T. and S.L. performed the literature review and wrote and approved the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

I.S.T.: speaker’s bureau (Amgen, Pfizer, and Servier) and consultancy (Pfizer and Servier); and S.L.: None to disclose.

References

  1. Rollig, C.; Bornhauser, M.; Thiede, C.; Taube, F.; Kramer, M.; Mohr, B.; Aulitzky, W.; Bodenstein, H.; Tischler, H.J.; Stuhlmann, R.; et al. Long-term prognosis of acute myeloid leukemia according to the new genetic risk classification of the European LeukemiaNet recommendations: Evaluation of the proposed reporting system. J. Clin. Oncol. 2011, 29, 2758–2765. [Google Scholar] [CrossRef] [PubMed]
  2. Heuser, M.; Freeman, S.D.; Ossenkoppele, G.J.; Buccisano, F.; Hourigan, C.S.; Ngai, L.L.; Tettero, J.M.; Bachas, C.; Baer, C.; Béné, M.C.; et al. 2021 Update on MRD in acute myeloid leukemia: A consensus document from the European LeukemiaNet MRD Working Party. Blood 2021, 138, 2753–2767. [Google Scholar] [CrossRef] [PubMed]
  3. Schuurhuis, G.J.; Heuser, M.; Freeman, S.; Bene, M.C.; Buccisano, F.; Cloos, J.; Grimwade, D.; Haferlach, T.; Hills, R.K.; Hourigan, C.S.; et al. Minimal/measurable residual disease in AML: A consensus document from the European LeukemiaNet MRD Working Party. Blood 2018, 131, 1275–1291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Dohner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef] [PubMed]
  5. Kronos Bio Speeds Development of Genetically Targeted Leukemia Drug with Unique Trial Design. Available online: https://www.statnews.com/2021/03/04/kronos-bio-speeds-development-of-genetically-targeted-leukemia-drug-with-unique-trial-design (accessed on 6 May 2021).
  6. Hirsch, P.; Zhang, Y.; Tang, R.; Joulin, V.; Boutroux, H.; Pronier, E.; Moatti, H.; Flandrin, P.; Marzac, C.; Bories, D.; et al. Genetic hierarchy and temporal variegation in the clonal history of acute myeloid leukaemia. Nat. Commun. 2016, 7, 12475. [Google Scholar] [CrossRef] [PubMed]
  7. Buckley, S.A.; Wood, B.L.; Othus, M.; Hourigan, C.S.; Ustun, C.; Linden, M.A.; DeFor, T.E.; Malagola, M.; Anthias, C.; Valkova, V.; et al. Minimal residual disease prior to allogeneic hematopoietic cell transplantation in acute myeloid leukemia: A meta-analysis. Haematologica 2017, 102, 865–873. [Google Scholar] [CrossRef] [Green Version]
  8. Short, N.J.; Fu, C.; Berry, D.A.; Walter, R.B.; Freeman, S.D.; Hourigan, C.S.; Huang, X.; Gonzalez, G.N.; Hwang, H.; Qi, X.; et al. Association of hematologic response and assay sensitivity on the prognostic impact of measurable residual disease in acute myeloid leukemia: A systematic review and meta-analysis. Leukemia 2022, 36, 2817–2826. [Google Scholar] [CrossRef]
  9. Wei, A.H.; Tiong, I.S. Midostaurin, enasidenib, CPX-351, gemtuzumab ozogamicin, and venetoclax bring new hope to AML. Blood 2017, 130, 2469–2474. [Google Scholar] [CrossRef] [Green Version]
  10. Kayser, S.; Levis, M.J. Updates on targeted therapies for acute myeloid leukaemia. Br. J. Haematol. 2022, 196, 316–328. [Google Scholar] [CrossRef]
  11. Tiong, I.S.; Dillon, R.; Ivey, A.; Teh, T.C.; Nguyen, P.; Cummings, N.; Taussig, D.C.; Latif, A.L.; Potter, N.E.; Runglall, M.; et al. Venetoclax induces rapid elimination of NPM1 mutant measurable residual disease in combination with low-intensity chemotherapy in acute myeloid leukaemia. Br. J. Haematol. 2021, 192, 1026–1030. [Google Scholar] [CrossRef]
  12. Tiong, I.S.; Hiwase, D.; Abro, E.U.; Bajel, A.; Palfreyman, E.; Loo, S.; Fleming, S.; Fong, C.Y.; Teh, T.-C.; Ivey, A.; et al. A Prospective Phase 2 Study of Venetoclax and Low Dose Ara-C (VALDAC) to Target Rising Molecular Measurable Residual Disease and Early Relapse in Acute Myeloid Leukemia. Blood 2022, 140, 1453–1455. [Google Scholar] [CrossRef]
  13. Platzbecker, U.; Wermke, M.; Radke, J.; Oelschlaegel, U.; Seltmann, F.; Kiani, A.; Klut, I.M.; Knoth, H.; Rollig, C.; Schetelig, 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. [Google Scholar] [CrossRef] [PubMed]
  14. Platzbecker, U.; Middeke, J.M.; Sockel, K.; Herbst, R.; Wolf, D.; Baldus, C.D.; Oelschlagel, U.; Mutherig, A.; Fransecky, L.; Noppeney, R.; et al. Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): An open-label, multicentre, phase 2 trial. Lancet Oncol. 2018, 19, 1668–1679. [Google Scholar] [CrossRef] [PubMed]
  15. Grimwade, D.; Jovanovic, J.V.; Hills, R.K.; Nugent, E.A.; Patel, Y.; Flora, R.; Diverio, D.; Jones, K.; Aslett, H.; Batson, E.; et al. Prospective minimal residual disease monitoring to predict relapse of acute promyelocytic leukemia and to direct pre-emptive arsenic trioxide therapy. J. Clin. Oncol. 2009, 27, 3650–3658. [Google Scholar] [CrossRef] [PubMed]
  16. Gokbuget, N.; Dombret, H.; Bonifacio, M.; Reichle, A.; Graux, C.; Faul, C.; Diedrich, H.; Topp, M.S.; Bruggemann, M.; Horst, H.A.; et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood 2018, 131, 1522–1531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Grimwade, D.; Hills, R.K.; Moorman, A.V.; Walker, H.; Chatters, S.; Goldstone, A.H.; Wheatley, K.; Harrison, C.J.; Burnett, A.K.; National Cancer Research Institute Adult Leukaemia Working Group. Refinement of cytogenetic classification in acute myeloid leukemia: Determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010, 116, 354–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Scholl, C.; Schlenk, R.F.; Eiwen, K.; Dohner, H.; Frohling, S.; Dohner, K.; AML Study Group. The prognostic value of MLL-AF9 detection in patients with t(9;11)(p22;q23)-positive acute myeloid leukemia. Haematologica 2005, 90, 1626–1634. [Google Scholar]
  19. Issa, G.C.; Aldoss, I.; Di Persio, J.F.; Cuglievan, B.; Stone, R.M.; Arellano, M.L.; Thirman, M.J.; Patel, M.R.; Dickens, D.; Shenoy, S.; et al. The menin inhibitor SNDX-5613 (revumenib) leads to durable responses in patients (pts) with KMT2A-rearranged or NPM1 mutant AML: Updated results of a phase (Ph) 1 study. Blood 2022, 140, 150–152. [Google Scholar] [CrossRef]
  20. Ommen, H.B.; Touzart, A.; MacIntyre, E.; Kern, W.; Haferlach, T.; Haferlach, C.; Tobal, K.; Hokland, P.; Schnittger, S. The kinetics of relapse in DEK-NUP214-positive acute myeloid leukemia patients. Eur. J. Haematol. 2015, 95, 436–441. [Google Scholar] [CrossRef]
  21. Welch, J.S.; Ley, T.J.; Link, D.C.; Miller, C.A.; Larson, D.E.; Koboldt, D.C.; Wartman, L.D.; Lamprecht, T.L.; Liu, F.; Xia, J.; et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 2012, 150, 264–278. [Google Scholar] [CrossRef] [Green Version]
  22. Papaemmanuil, E.; Gerstung, M.; Bullinger, L.; Gaidzik, V.I.; Paschka, P.; Roberts, N.D.; Potter, N.E.; Heuser, M.; Thol, F.; Bolli, N. Genomic classification and prognosis in acute myeloid leukemia. N. Engl. J. Med. 2016, 374, 2209–2221. [Google Scholar] [CrossRef] [PubMed]
  23. Jongen-Lavrencic, M.; Grob, T.; Hanekamp, D.; Kavelaars, F.G.; Al Hinai, A.; Zeilemaker, A.; Erpelinck-Verschueren, C.A.J.; Gradowska, P.L.; Meijer, R.; Cloos, J.; et al. Molecular minimal residual disease in acute myeloid leukemia. N. Engl. J. Med. 2018, 378, 1189–1199. [Google Scholar] [CrossRef] [PubMed]
  24. Klco, J.M.; Miller, C.A.; Griffith, M.; Petti, A.; Spencer, D.H.; Ketkar-Kulkarni, S.; Wartman, L.D.; Christopher, M.; Lamprecht, T.L.; Helton, N.M. Association between mutation clearance after induction therapy and outcomes in acute myeloid leukemia. JAMA 2015, 314, 811–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Morita, K.; Kantarjian, H.M.; Wang, F.; Yan, Y.; Bueso-Ramos, C.; Sasaki, K.; Issa, G.C.; Wang, S.; Jorgensen, J.; Song, X.; et al. Clearance of somatic mutations at remission and the risk of relapse in acute myeloid leukemia. J. Clin. Oncol. 2018, 36, 1788–1797. [Google Scholar] [CrossRef] [PubMed]
  26. Rothenberg-Thurley, M.; Amler, S.; Goerlich, D.; Kohnke, T.; Konstandin, N.P.; Schneider, S.; Sauerland, M.C.; Herold, T.; Hubmann, M.; Ksienzyk, B.; et al. Persistence of pre-leukemic clones during first remission and risk of relapse in acute myeloid leukemia. Leukemia 2018, 32, 1598–1608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Thol, F.; Gabdoulline, R.; Liebich, A.; Klement, P.; Schiller, J.; Kandziora, C.; Hambach, L.; Stadler, M.; Koenecke, C.; Flintrop, M.; et al. Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML. Blood 2018, 132, 1703–1713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Kim, T.; Moon, J.H.; Ahn, J.S.; Kim, Y.K.; Lee, S.S.; Ahn, S.Y.; Jung, S.H.; Yang, D.H.; Lee, J.J.; Choi, S.H.; et al. Next-generation sequencing-based posttransplant monitoring of acute myeloid leukemia identifies patients at high risk of relapse. Blood 2018, 132, 1604–1613. [Google Scholar] [CrossRef] [Green Version]
  29. Hourigan, C.S.; Dillon, L.W.; Gui, G.; Logan, B.R.; Fei, M.; Ghannam, J.; Li, Y.; Licon, A.; Alyea, E.P.; Bashey, A.; et al. Impact of conditioning intensity of allogeneic transplantation for acute myeloid leukemia with genomic evidence of residual disease. J. Clin. Oncol. 2020, 38, 1273–1283. [Google Scholar] [CrossRef]
  30. Heuser, M.; Heida, B.; Buttner, K.; Wienecke, C.P.; Teich, K.; Funke, C.; Brandes, M.; Klement, P.; Liebich, A.; Wichmann, M.; et al. Posttransplantation MRD monitoring in patients with AML by next-generation sequencing using DTA and non-DTA mutations. Blood Adv. 2021, 5, 2294–2304. [Google Scholar] [CrossRef]
  31. Patkar, N.; Kakirde, C.; Shaikh, A.F.; Salve, R.; Bhanshe, P.; Chatterjee, G.; Rajpal, S.; Joshi, S.; Chaudhary, S.; Kodgule, R.; et al. Clinical impact of panel-based error-corrected next generation sequencing versus flow cytometry to detect measurable residual disease (MRD) in acute myeloid leukemia (AML). Leukemia 2021, 35, 1392–1404. [Google Scholar] [CrossRef]
  32. Tsai, C.H.; Tang, J.L.; Tien, F.M.; Kuo, Y.Y.; Wu, D.C.; Lin, C.C.; Tseng, M.H.; Peng, Y.L.; Hou, M.F.; Chuang, Y.K.; et al. Clinical implications of sequential MRD monitoring by NGS at 2 time points after chemotherapy in patients with AML. Blood Adv. 2021, 5, 2456–2466. [Google Scholar] [CrossRef] [PubMed]
  33. Hourigan, C.S.; Dillon, L.W.; Gui, G.; Ravindra, N.; Wong, Z.; Andrew, G.; Mukherjee, D.; El Chaer, F.; Yoon, C.; Spellman, S.; et al. Pre-MEASURE: Multicenter evaluation of the prognostic significance of measurable residual disease testing prior to allogeneic transplantation for adult patients with AML in first remission. J. Clin. Oncol. 2022, 40, 7006. [Google Scholar] [CrossRef]
  34. Hirsch, P.; Lambert, J.; Bucci, M.; Deswarte, C.; Lambert, J.; Fenwarth, L.; Dombret, H.; Duployez, N.; Preudhomme, C.; Itzykson, R.; et al. Persistence of mutations in complete remission including DNMT3A, TET2 and ASXL1 mutations is associated with worse prognosis in patients with acute myeloid leukemia treated in ALFA 0702 study. Blood 2022, 140, 549–550. [Google Scholar] [CrossRef]
  35. Metzeler, K.H.; Herold, T.; Rothenberg-Thurley, M.; Amler, S.; Sauerland, M.C.; Gorlich, D.; Schneider, S.; Konstandin, N.P.; Dufour, A.; Braundl, K.; et al. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood 2016, 128, 686–698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. The Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 2013, 368, 2059–2074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Jaiswal, S.; Fontanillas, P.; Flannick, J.; Manning, A.; Grauman, P.V.; Mar, B.G.; Lindsley, R.C.; Mermel, C.H.; Burtt, N.; Chavez, A.; et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 2014, 371, 2488–2498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Genovese, G.; Kahler, A.K.; Handsaker, R.E.; Lindberg, J.; Rose, S.A.; Bakhoum, S.F.; Chambert, K.; Mick, E.; Neale, B.M.; Fromer, M.; et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 2014, 371, 2477–2487. [Google Scholar] [CrossRef] [Green Version]
  39. Desai, P.; Mencia-Trinchant, N.; Savenkov, O.; Simon, M.S.; Cheang, G.; Lee, S.; Samuel, M.; Ritchie, E.K.; Guzman, M.L.; Ballman, K.V.; et al. Somatic mutations precede acute myeloid leukemia years before diagnosis. Nat. Med. 2018, 24, 1015–1023. [Google Scholar] [CrossRef]
  40. Ferret, Y.; Boissel, N.; Helevaut, N.; Madic, J.; Nibourel, O.; Marceau-Renaut, A.; Bucci, M.; Geffroy, S.; Celli-Lebras, K.; Castaigne, S.; et al. Clinical relevance of IDH1/2 mutant allele burden during follow-up in acute myeloid leukemia. A study by the French ALFA group. Haematologica 2018, 103, 822–829. [Google Scholar] [CrossRef] [Green Version]
  41. Ok, C.Y.; Loghavi, S.; Sui, D.; Wei, P.; Kanagal-Shamanna, R.; Yin, C.C.; Zuo, Z.; Routbort, M.J.; Tang, G.; Tang, Z.; et al. Persistent IDH1/2 mutation in remission can predict relapse in patients with acute myeloid leukemia. Haematologica 2018, 104, 305–311. [Google Scholar] [CrossRef] [Green Version]
  42. Cappelli, L.V.; Meggendorfer, M.; Baer, C.; Nadarajah, N.; Hutter, S.; Jeromin, S.; Dicker, F.; Kern, W.; Haferlach, T.; Haferlach, C.; et al. Indeterminate and oncogenic potential: CHIP vs CHOP mutations in AML with NPM1 alteration. Leukemia 2021, 36, 394–402. [Google Scholar] [CrossRef] [PubMed]
  43. Wiseman, D.H.; Williams, E.L.; Wilks, D.P.; Sun Leong, H.; Somerville, T.D.; Dennis, M.W.; Struys, E.A.; Bakkali, A.; Salomons, G.S.; Somervaille, T.C. Frequent reconstitution of IDH2(R140Q) mutant clonal multilineage hematopoiesis following chemotherapy for acute myeloid leukemia. Leukemia 2016, 30, 1946–1950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Bill, M.; Jentzsch, M.; Bischof, L.; Kohlschmidt, J.; Grimm, J.; Schmalbrock, L.K.; Backhaus, D.; Brauer, D.; Goldmann, K.; Franke, G.N.; et al. Impact of IDH1 and IDH2 mutation detection at diagnosis and in remission in patients with AML receiving allogeneic transplantation. Blood Adv. 2023, 7, 436–444. [Google Scholar] [CrossRef] [PubMed]
  45. Cloos, J.; Goemans, B.F.; Hess, C.J.; van Oostveen, J.W.; Waisfisz, Q.; Corthals, S.; de Lange, D.; Boeckx, N.; Hahlen, K.; Reinhardt, D.; et al. Stability and prognostic influence of FLT3 mutations in paired initial and relapsed AML samples. Leukemia 2006, 20, 1217–1220. [Google Scholar] [CrossRef] [PubMed]
  46. Kottaridis, P.D.; Gale, R.E.; Langabeer, S.E.; Frew, M.E.; Bowen, D.T.; Linch, D.C. 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, 2393–2398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Amatangelo, M.D.; Quek, L.; Shih, A.; Stein, E.M.; Roshal, M.; David, M.D.; Marteyn, B.; Farnoud, N.R.; de Botton, S.; Bernard, O.A.; et al. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response. Blood 2017, 130, 732–741. [Google Scholar] [CrossRef] [PubMed]
  48. McMahon, C.M.; Ferng, T.; Canaani, J.; Wang, E.S.; Morrissette, J.J.D.; Eastburn, D.J.; Pellegrino, M.; Durruthy-Durruthy, R.; Watt, C.D.; Asthana, S.; et al. Clonal selection with RAS pathway activation mediates secondary clinical resistance to selective FLT3 inhibition in acute myeloid leukemia. Cancer Discov. 2019, 9, 1050–1063. [Google Scholar] [CrossRef]
  49. DiNardo, C.D.; Tiong, I.S.; Quaglieri, A.; MacRaild, S.; Loghavi, S.; Brown, F.C.; Thijssen, R.; Pomilio, G.; Ivey, A.; Salmon, J.M.; et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood 2020, 135, 791–803. [Google Scholar] [CrossRef]
  50. Helbig, G.; Koclęga, A.; Wieczorkiewicz-Kabut, A.; Woźniczka, K.; Kopińska, A.; Boral, K.; Grygoruk-Wiśniowska, I.; Stachowicz, M.; Karolczyk, A. Pre-transplant FLT3/ITD status predicts outcome in FLT3-mutated acute myeloid leukemia following allogeneic stem cell transplantation. Ann. Hematol. 2020, 99, 1845–1853. [Google Scholar] [CrossRef] [Green Version]
  51. Gaballa, S.; Saliba, R.; Oran, B.; Brammer, J.E.; Chen, J.; Rondon, G.; Alousi, A.M.; Kebriaei, P.; Marin, D.; Popat, U.R.; et al. Relapse risk and survival in patients with FLT3 mutated acute myeloid leukemia undergoing stem cell transplantation. Am. J. Hematol. 2017, 92, 331–337. [Google Scholar] [CrossRef] [Green Version]
  52. Spencer, D.H.; Abel, H.J.; Lockwood, C.M.; Payton, J.E.; Szankasi, P.; Kelley, T.W.; Kulkarni, S.; Pfeifer, J.D.; Duncavage, E.J. Detection of FLT3 internal tandem duplication in targeted, short-read-length, next-generation sequencing data. J. Mol. Diagn. 2013, 15, 81–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Bibault, J.E.; Figeac, M.; Helevaut, N.; Rodriguez, C.; Quief, S.; Sebda, S.; Renneville, A.; Nibourel, O.; Rousselot, P.; Gruson, B.; et al. Next-generation sequencing of FLT3 internal tandem duplications for minimal residual disease monitoring in acute myeloid leukemia. Oncotarget 2015, 6, 22812–22821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Levis, M.J.; Perl, A.E.; Altman, J.K.; Gocke, C.D.; Bahceci, E.; Hill, J.; Liu, C.; Xie, Z.; Carson, A.R.; McClain, V.; et al. A next-generation sequencing-based assay for minimal residual disease assessment in AML patients with FLT3-ITD mutations. Blood Adv. 2018, 2, 825–831. [Google Scholar] [CrossRef] [PubMed]
  55. He, R.; Devine, D.J.; Tu, Z.J.; Mai, M.; Chen, D.; Nguyen, P.L.; Oliveira, J.L.; Hoyer, J.D.; Reichard, K.K.; Ollila, P.L.; et al. Hybridization capture-based next generation sequencing reliably detects FLT3 mutations and classifies FLT3-internal tandem duplication allelic ratio in acute myeloid leukemia: A comparative study to standard fragment analysis. Mod. Pathol. 2020, 33, 334–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ma, L.; Cao, Y.; Jiang, Y.; Cong, X.; Lu, S.; Shen, J.; Liu, Q.; Han, C.; Zhan, Y.; Cao, Y. Detection the frequency and characteristics of FLT3 internal tandem duplication mutations by capillary electrophoresis assay and next-generation sequencing in. Clin. Lab. 2016, 62, 2065–2072. [Google Scholar] [CrossRef]
  57. Blatte, T.J.; Schmalbrock, L.K.; Skambraks, S.; Lux, S.; Cocciardi, S.; Dolnik, A.; Dohner, H.; Dohner, K.; Bullinger, L. getITD for FLT3-ITD-based MRD monitoring in AML. Leukemia 2019, 33, 2535–2539. [Google Scholar] [CrossRef] [Green Version]
  58. Loo, S.; Dillon, R.; Ivey, A.; Anstee, N.S.; Othman, J.; Tiong, I.S.; Potter, N.; Jovanovic, J.; Runglall, M.; Chong, C.C.; et al. Pretransplant FLT3-ITD MRD assessed by high-sensitivity PCR-NGS determines posttransplant clinical outcome. Blood 2022, 140, 2407–2411. [Google Scholar] [CrossRef]
  59. Lee, J.M.; Park, S.; Hwang, I.; Kang, D.; Cho, B.S.; Kim, H.J.; Ahn, A.; Kim, M.; Kim, Y. FLT3-ITD measurable residual disease monitoring in acute myeloid leukemia using next-generation sequencing. Cancers 2022, 14, 6121. [Google Scholar] [CrossRef]
  60. Dillon, L.W.; Gui, G.; Ravindra, N.; Wong, Z.; Andrew, G.; Mukherjee, D.; Zeger, S.L.; El Chaer, F.; Spellman, S.; Howard, A.; et al. Pre-MEASURE: FLT3-ITD and mutated NPM1 measurable residual disease before allogeneic transplant in adults with AML in first remission. medRxiv 2022. preprint. [Google Scholar] [CrossRef]
  61. Grob, T.; Sanders, M.A.; Vonk, C.M.; Kavelaars, F.G.; Rijken, M.; Hanekamp, D.W.; Gradowska, P.L.; Cloos, J.; Floisand, Y.; van Marwijk Kooy, M.; et al. Prognostic value of FLT3-internal tandem duplication residual disease in acute myeloid leukemia. J. Clin. Oncol. 2022, 41, 756–765. [Google Scholar] [CrossRef]
  62. Schranz, K.; Hubmann, M.; Harin, E.; Vosberg, S.; Herold, T.; Metzeler, K.H.; Rothenberg-Thurley, M.; Janke, H.; Braundl, K.; Ksienzyk, B.; et al. Clonal heterogeneity of FLT3-ITD detected by high-throughput amplicon sequencing correlates with adverse prognosis in acute myeloid leukemia. Oncotarget 2018, 9, 30128–30145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yokoyama, S.; Onozawa, M.; Yoshida, S.; Miyashita, N.; Kimura, H.; Takahashi, S.; Matsukawa, T.; Fujisawa, S.; Miki, K.; Hidaka, D.; et al. Subclinical minute FLT3-ITD clone can be detected in clinically FLT3-ITD-negative acute myeloid leukemia. Blood 2022, 140, 9094–9095. [Google Scholar] [CrossRef]
  64. Rucker, F.G.; Du, L.; Luck, T.J.; Benner, A.; Krzykalla, J.; Gathmann, I.; Voso, M.T.; Amadori, S.; Prior, T.W.; Brandwein, J.M.; et al. Molecular landscape and prognostic impact of FLT3-ITD insertion site in acute myeloid leukemia: RATIFY study results. Leukemia 2022, 36, 90–99. [Google Scholar] [CrossRef] [PubMed]
  65. DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Dohner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef] [PubMed]
  66. Wei, A.H.; Montesinos, P.; Ivanov, V.; DiNardo, C.D.; Novak, J.; Laribi, K.; Kim, I.; Stevens, D.A.; Fiedler, W.; Pagoni, M.; et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: A phase 3 randomized placebo-controlled trial. Blood 2020, 135, 2137–2145. [Google Scholar] [CrossRef]
  67. Pratz, K.; Jonas, B.A.; Pullarkat, V.A.; Thirman, M.J.; Garcia, J.S.; Fiedler, W.; Yamamoto, K.; Wang, J.; Yoon, S.S.; Wolach, O.; et al. Long-term follow-up of the phase 3 Viale-a clinical trial of venetoclax plus azacitidine for patients with untreated acute myeloid leukemia ineligible for intensive chemotherapy. Blood 2022, 135, 2137–2145. [Google Scholar] [CrossRef]
  68. Pratz, K.W.; Jonas, B.A.; Pullarkat, V.; Recher, C.; Schuh, A.C.; Thirman, M.J.; Garcia, J.S.; DiNardo, C.D.; Vorobyev, V.; Fracchiolla, N.S.; et al. Measurable residual disease response and prognosis in treatment-naive acute myeloid leukemia with venetoclax and azacitidine. J. Clin. Oncol. 2022, 40, 855–865. [Google Scholar] [CrossRef]
  69. Maiti, A.; DiNardo, C.D.; Wang, S.A.; Jorgensen, J.; Kadia, T.M.; Daver, N.G.; Short, N.J.; Yilmaz, M.; Pemmaraju, N.; Borthakur, G.; et al. Prognostic value of measurable residual disease after venetoclax and decitabine in acute myeloid leukemia. Blood Adv. 2021, 5, 1876–1883. [Google Scholar] [CrossRef]
  70. Ong, S.Y.; Tan Si Yun, M.; Abdul Halim, N.A.; Christopher, D.; Jen, W.Y.; Gallardo, C.; Tan Hwee Yim, A.; Woon, Y.K.; Ng, H.J.; Ooi, M.; et al. Real-world experience of measurable residual disease response and prognosis in acute myeloid leukemia treated with venetoclax and azacitidine. Cancers 2022, 14, 3576. [Google Scholar] [CrossRef]
  71. Othman, J.; Tiong, I.S.; Mokretar, K.; Ivey, A.; Austin, M.J.; Latif, A.L.; Crawley, C.; Amer, M.; Crolla, F.; Cross, J.W.; et al. Molecular MRD assessment is strongly prognostic in patients with NPM1 mutated AML receiving venetoclax based non-intensive therapy. Blood 2022, 140, 2033–2035. [Google Scholar] [CrossRef]
  72. Kadia, T.M.; Reville, P.K.; Wang, X.; Rausch, C.R.; Borthakur, G.; Pemmaraju, N.; Daver, N.G.; DiNardo, C.D.; Sasaki, K.; Issa, G.C.; et al. Phase II study of venetoclax added to cladribine plus low-dose cytarabine alternating with 5-azacitidine in older patients with newly diagnosed acute myeloid leukemia. J. Clin. Oncol. 2022, 40, 3848–3857. [Google Scholar] [CrossRef] [PubMed]
  73. DiNardo, C.D.; Lachowiez, C.A.; Takahashi, K.; Loghavi, S.; Kadia, T.; Daver, N.; Xiao, L.; Adeoti, M.; Short, N.J.; Sasaki, K.; et al. Venetoclax combined with FLAG-IDA induction and consolidation in newly diagnosed acute myeloid leukemia. Am. J. Hematol. 2022, 97, 1035–1043. [Google Scholar] [CrossRef] [PubMed]
  74. Chua, C.C.; Hammond, D.; Kent, A.; Tiong, I.S.; Konopleva, M.Y.; Pollyea, D.A.; DiNardo, C.D.; Wei, A.H. Treatment-free remission after ceasing venetoclax-based therapy in patients with acute myeloid leukemia. Blood Adv. 2022, 6, 3879–3883. [Google Scholar] [CrossRef] [PubMed]
  75. Gutman, J.A.; Winters, A.C.; Kent, A.; Amaya, M.L.; McMahon, C.M.; Smith, C.; Jordan, C.T.; Stevens, B.M.; Minhajuddin, M.; Pei, S.; et al. Higher-dose venetoclax with measurable residual disease-guided azacitidine discontinuation in newly diagnosed patients with acute myeloid leukemia: Phase 2 Hiddav study. Blood 2022, 140, 3275–3276. [Google Scholar] [CrossRef]
  76. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Dohner, K.; Marcucci, G.; et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef]
  77. Levis, M.; Shi, W.; Chang, K.; Laing, C.; Pollner, R.; Gocke, C.; Adams, E.; Berisha, F.; Lameh, J.; Lesegretain, A. FLT3 inhibitors added to induction therapy induce deeper remissions. Blood 2020, 135, 75–78. [Google Scholar] [CrossRef]
  78. Herzig, J.K.; Rücker, F.G.; Schmalbrock, L.K.; Blätte, T.J.; Weber, D.; Skambraks, S.; Kapp-Schwoerer, S.; Corbacioglu, A.; Gaidzik, V.I.; Paschka, P.; et al. Next-Generation Sequencing (NGS)-Based Measurable Residual Disease (MRD) Monitoring in Acute Myeloid Leukemia with FLT3 Internal Tandem Duplication (FLT3-ITD+ AML) Treated with Additional Midostaurin. Blood 2020, 136, 21–22. [Google Scholar] [CrossRef]
  79. Perl, A.E.; Martinelli, G.; Cortes, J.E.; Neubauer, A.; Berman, E.; Paolini, S.; Montesinos, P.; Baer, M.R.; Larson, R.A.; Ustun, C. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N. Engl. J. Med. 2019, 381, 1728–1740. [Google Scholar] [CrossRef]
  80. Altman, J.K.; Perl, A.E.; Hill, J.E.; Rosales, M.; Bahceci, E.; Levis, M.J. The impact of FLT3 mutation clearance and treatment response after gilteritinib therapy on overall survival in patients with FLT3 mutation-positive relapsed/refractory acute myeloid leukemia. Cancer Med. 2021, 10, 797–805. [Google Scholar] [CrossRef]
  81. Daver, N.; Perl, A.E.; Maly, J.; Levis, M.; Ritchie, E.; Litzow, M.; McCloskey, J.; Smith, C.C.; Schiller, G.; Bradley, T.; et al. Venetoclax plus gilteritinib for FLT3-mutated relapsed/refractory acute myeloid leukemia. J. Clin. Oncol. 2022, 40, 4048–4059. [Google Scholar] [CrossRef]
  82. Levis, M.J.; Erba, H.P.; Montesinos, P.; Vrhovac, R.; Patkowska, E.; Kim, H.J.; Zak, P.; Wang, P.N.; Rohrbach, J.E.C.; Chang, K.C.N.; et al. Quantum-First trial: FLT3-ITD-specific MRD clearance is associated with improved overall survival. Blood 2022, 140, 546–548. [Google Scholar] [CrossRef]
  83. DiNardo, C.D.; Stein, E.M.; de Botton, S.; Roboz, G.J.; Altman, J.K.; Mims, A.S.; Swords, R.; Collins, R.H.; Mannis, G.N.; Pollyea, D.A.; et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N. Engl. J. Med. 2018, 378, 2386–2398. [Google Scholar] [CrossRef] [PubMed]
  84. Stein, E.M.; DiNardo, C.D.; Pollyea, D.A.; Fathi, A.T.; Roboz, G.J.; Altman, J.K.; Stone, R.M.; DeAngelo, D.J.; Levine, R.L.; Flinn, I.W.; et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017, 130, 722–731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Stein, E.M.; DiNardo, C.D.; Fathi, A.T.; Pollyea, D.A.; Stone, R.M.; Altman, J.K.; Roboz, G.J.; Patel, M.R.; Collins, R.; Flinn, I.W. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood 2019, 133, 676–687. [Google Scholar] [CrossRef] [Green Version]
  86. Roboz, G.J.; DiNardo, C.D.; Stein, E.M.; de Botton, S.; Mims, A.S.; Prince, G.T.; Altman, J.K.; Arellano, M.L.; Donnellan, W.; Erba, H.P.; et al. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood 2020, 135, 463–471. [Google Scholar] [CrossRef] [Green Version]
  87. Montesinos, P.; Recher, C.; Vives, S.; Zarzycka, E.; Wang, J.; Bertani, G.; Heuser, M.; Calado, R.T.; Schuh, A.C.; Yeh, S.P.; et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia. N. Engl. J. Med. 2022, 386, 1519–1531. [Google Scholar] [CrossRef]
  88. Dohner, H.; Marchione, D.M.; Choe, S.; Montesinos, P.; Recher, C.; Vives, S.; Zarzycka, E.; Wang, J.; Cerchione, C.; Heuser, M.; et al. Molecular characterization of clinical response and relapse in patients with IDH1m ND-AML treated with ivo+AZA in the AGILE study. Blood 2022, 539–542. [Google Scholar] [CrossRef]
  89. DiNardo, C.D.; Schuh, A.C.; Stein, E.M.; Montesinos, P.; Wei, A.H.; de Botton, S.; Zeidan, A.M.; Fathi, A.T.; Kantarjian, H.M.; Bennett, J.M.; et al. Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): A single-arm, phase 1b and randomised, phase 2 trial. Lancet Oncol. 2021, 22, 1597–1608. [Google Scholar] [CrossRef]
  90. Lachowiez, C.A.; Garcia, J.S.; Borthakur, G.; Loghavi, S.; Zeng, Z.; Tippett, G.D.; Kadia, T.; Masarova, L.; Yilmaz, M.; Maiti, A.; et al. A phase Ib/II study of ivosidenib with venetoclax +/− azacitidine in IDH1-mutated hematologic malignancies. J. Clin. Oncol. 2022, 39, 7012. [Google Scholar] [CrossRef]
  91. Stein, E.M.; DiNardo, C.D.; Fathi, A.T.; Mims, A.S.; Pratz, K.W.; Savona, M.R.; Stein, A.S.; Stone, R.M.; Winer, E.S.; Seet, C.S.; et al. Ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed AML: A phase 1 study. Blood 2021, 137, 1792–1803. [Google Scholar] [CrossRef]
  92. Hills, R.K.; Castaigne, S.; Appelbaum, F.R.; Delaunay, J.; Petersdorf, S.; Othus, M.; Estey, E.H.; Dombret, H.; Chevret, S.; Ifrah, N.; et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: A meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014, 15, 986–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Lambert, J.; Pautas, C.; Terre, C.; Raffoux, E.; Turlure, P.; Caillot, D.; Legrand, O.; Thomas, X.; Gardin, C.; Gogat-Marchant, K.; et al. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: Final efficacy and safety updates from the open-label, phase III ALFA-0701 trial. Haematologica 2019, 104, 113–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Khan, N.; Hills, R.K.; Virgo, P.; Couzens, S.; Clark, N.; Gilkes, A.; Richardson, P.; Knapper, S.; Grimwade, D.; Russell, N.H.; et al. Expression of CD33 is a predictive factor for effect of gemtuzumab ozogamicin at different doses in adult acute myeloid leukaemia. Leukemia 2017, 31, 1059–1068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lambert, J.; Lambert, J.; Nibourel, O.; Pautas, C.; Hayette, S.; Cayuela, J.M.; Terre, C.; Rousselot, P.; Dombret, H.; Chevret, S.; 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, 6280–6288. [Google Scholar] [CrossRef] [Green Version]
  96. Kapp-Schwoerer, S.; Weber, D.; Corbacioglu, A.; Gaidzik, V.I.; Paschka, P.; Kronke, J.; Theis, F.; Rucker, F.G.; Teleanu, M.V.; Panina, E.; 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, 3041–3050. [Google Scholar] [CrossRef]
  97. Yin, J.A.; O’Brien, M.A.; Hills, R.K.; Daly, S.B.; Wheatley, K.; Burnett, A.K. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: Results of the United Kingdom MRC AML-15 trial. Blood 2012, 120, 2826–2835. [Google Scholar] [CrossRef] [Green Version]
  98. Wei, A.H.; Dohner, H.; Pocock, C.; Montesinos, P.; Afanasyev, B.; Dombret, H.; Ravandi, F.; Sayar, H.; Jang, J.H.; Porkka, K.; et al. Oral azacitidine maintenance therapy for acute myeloid leukemia in first remission. N. Engl. J. Med. 2020, 383, 2526–2537. [Google Scholar] [CrossRef]
  99. Roboz, G.J.; Ravandi, F.; Wei, A.H.; Dombret, H.; Thol, F.; Voso, M.T.; Schuh, A.C.; Porkka, K.; La Torre, I.; Skikne, B.; et al. Oral azacitidine prolongs survival of patients with AML in remission independently of measurable residual disease status. Blood 2022, 139, 2145–2155. [Google Scholar] [CrossRef]
  100. Dohner, H.; Wei, A.H.; Roboz, G.J.; Montesinos, P.; Thol, F.R.; Ravandi, F.; Dombret, H.; Porkka, K.; Sandhu, I.; Skikne, B.; et al. Prognostic impact of NPM1 and FLT3 mutations in patients with AML in first remission treated with oral azacitidine. Blood 2022, 140, 1674–1685. [Google Scholar] [CrossRef]
  101. Short, N.J.; Zhou, S.; Fu, C.; Berry, D.A.; Walter, R.B.; Freeman, S.D.; Hourigan, C.S.; Huang, X.; Nogueras Gonzalez, G.; Hwang, H.; et al. Association of measurable residual disease with survival outcomes in patients with acute myeloid leukemia: A systematic review and meta-analysis. JAMA Oncol. 2020, 6, 1890–1899. [Google Scholar] [CrossRef]
  102. Yu, S.; Fan, Z.; Ma, L.; Wang, Y.; Huang, F.; Zhang, Q.; Huang, J.; Wang, S.; Xu, N.; Xuan, L.; et al. Association between measurable residual disease in patients with intermediate-risk acute myeloid leukemia and first remission, treatment, and outcomes. JAMA Netw. Open 2021, 4, e2115991. [Google Scholar] [CrossRef] [PubMed]
  103. Craddock, C. Transplant in AML with measurable residual disease: Proceed or defer? Hematology 2022, 2022, 528–533. [Google Scholar] [CrossRef] [PubMed]
  104. Loke, J.; Vyas, H.; Craddock, C. Optimizing transplant approaches and post-transplant strategies for patients with acute myeloid leukemia. Front. Oncol. 2021, 11, 666091. [Google Scholar] [CrossRef] [PubMed]
  105. Spyridonidis, A. How I treat measurable (minimal) residual disease in acute leukemia after allogeneic hematopoietic cell transplantation. Blood 2020, 135, 1639–1649. [Google Scholar] [CrossRef] [PubMed]
  106. Gaut, D.; Mead, M. Measurable residual disease in hematopoietic stem cell transplantation-eligible patients with acute myeloid leukemia: Clinical significance and promising therapeutic strategies. Leuk. Lymphoma 2021, 62, 8–31. [Google Scholar] [CrossRef]
  107. Araki, D.; Wood, B.L.; Othus, M.; Radich, J.P.; Halpern, A.B.; Zhou, Y.; Mielcarek, M.; Estey, E.H.; Appelbaum, F.R.; Walter, R.B. Allogeneic hematopoietic cell transplantation for acute myeloid leukemia: Time to move toward a minimal residual disease-based definition of complete remission? J. Clin. Oncol. 2016, 34, 329–336. [Google Scholar] [CrossRef] [Green Version]
  108. Gilleece, M.H.; Shimoni, A.; Labopin, M.; Robinson, S.; Beelen, D.; Socie, G.; Unal, A.; Ganser, A.; Vitek, A.; Sengeloev, H.; et al. Measurable residual disease status and outcome of transplant in acute myeloid leukemia in second complete remission: A study by the acute leukemia working party of the EBMT. Blood Cancer J. 2021, 11, 88. [Google Scholar] [CrossRef]
  109. Sockel, K.; Wermke, M.; Radke, J.; Kiani, A.; Schaich, M.; Bornhauser, M.; Ehninger, G.; Thiede, C.; Platzbecker, U. Minimal residual disease-directed preemptive treatment with azacitidine in patients with NPM1-mutant acute myeloid leukemia and molecular relapse. Haematologica 2011, 96, 1568–1570. [Google Scholar] [CrossRef]
  110. Bataller, A.; Onate, G.; Diaz-Beya, M.; Guijarro, F.; Garrido, A.; Vives, S.; Tormo, M.; Arnan, M.; Salamero, O.; Sampol, A.; et al. Acute myeloid leukemia with NPM1 mutation and favorable European LeukemiaNet category: Outcome after preemptive intervention based on measurable residual disease. Br. J. Haematol. 2020, 191, 52–61. [Google Scholar] [CrossRef]
  111. Short, N.J.; Macaron, W.; Kadia, T.; Dinardo, C.; Issa, G.C.; Daver, N.; Wang, S.; Jorgensen, J.; Nguyen, D.; Bidikian, A.; et al. Clinical outcomes and impact of therapeutic intervention in patients with acute myeloid leukemia who experience measurable residual disease (MRD) recurrence following MRD-negative remission. Am. J. Hematol. 2022, 97, E408–E411. [Google Scholar] [CrossRef]
  112. Dillon, R.; Hills, R.; Freeman, S.; Potter, N.; Jovanovic, J.; Ivey, A.; Kanda, A.S.; Runglall, M.; Foot, N.; Valganon, M.; et al. Molecular MRD status and outcome after transplantation in NPM1-mutated AML. Blood 2020, 135, 680–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Wood, H.; Bourlon, C.; Kulasekararaj, A.; Borg, A.; Pavlu, J.; Elder, P.; Taussig, D.; Veitch, S.; Knapper, S. Venetoclax-based non-intensive combinations successfully salvage molecular relapse of acute myeloid leukemia and are an important bridge to cellular therapy in relapsed/refractory disease—Real-world data from a UK-wide programme. Blood 2022, 140, 9016–9018. [Google Scholar] [CrossRef]
  114. Othman, J.; Potter, N.; Mokretar, K.; Taussig, D.; Khan, A.; Krishnamurthy, P.; Latif, A.L.; Cahalin, P.; Aries, J.; Amer, M.; et al. High molecular response rate and overall survival with FLT3 inhibitors as MRD-guided salvage treatment for molecular failure in AML. Blood 2022, 140, 2002–2004. [Google Scholar] [CrossRef]
  115. Tiong, I.S.; Dillon, R.; Ivey, A.; Kuzich, J.A.; Thiagarajah, N.; Sharplin, K.M.; Kok, C.H.; Tedjaseputra, A.; Rowland, J.P.; Grove, C.S.; et al. Clinical impact of NPM1-mutant molecular persistence after chemotherapy for acute myeloid leukemia. Blood Adv. 2021, 5, 5107–5111. [Google Scholar] [CrossRef] [PubMed]
  116. Wei, A.; Iland, H.; Reynolds, J.; Loo, S.; Chua, C.C.; Westerman, D.; Tiong, I.S.; Ivey, A.; Blombery, P.; Anstee, N.S.; et al. ALLG AMLM26 Phase 1B/2 study investigating novel therapies to target early relapse and clonal evolution as pre-emptive therapy in AML (INTERCEPT): A multi-arm, precision-based, recursive, platform trial. Blood 2022, 3341–3343. [Google Scholar] [CrossRef]
  117. Hourigan, C.S. Achieving MRD negativity in AML: How important is this and how do we get there? Hematol. Am. Soc. Hematol. Educ. Program 2022, 2022, 9–14. [Google Scholar] [CrossRef] [PubMed]
  118. Little, R.F.; Othus, M.; Assouline, S.; Ansher, S.; Atallah, E.L.; Lindsley, R.C.; Freidlin, B.; Gore, S.D.; Harris, L.; Hourigan, C.S.; et al. Umbrella trial in myeloid malignancies: The Myelomatch National Clinical Trials Network Precision Medicine Initiative. Blood 2022, 140, 9057–9060. [Google Scholar] [CrossRef]
  119. Mission Bio. Mission Bio Launches Early Access Program for Single-Cell Measurable Residual Disease Detection in Blood Cancers. Available online: https://missionbio.com/press/single-cell-mrd-early-access-program/ (accessed on 28 July 2022).
  120. Robinson, T.; Bowman, R.L.; Persaud, S.; Liu, Y.; Gao, Q.; Zhang, J.; Sun, X. Single cell genotypic and phenotypic analysis of measurable residual disease in acute myeloid leukemia. Blood 2022, 140, 2276–2277. [Google Scholar] [CrossRef]
Figure 1. Trial schema of the AMLM26 INTERCEPT for MRD-directed therapy in AML. Abbreviations: AML, acute myeloid leukemia; AZA, azacitidine; LDAC, low dose cytarabine; MRD, measurable residual disease; and VEN, venetoclax.
Figure 1. Trial schema of the AMLM26 INTERCEPT for MRD-directed therapy in AML. Abbreviations: AML, acute myeloid leukemia; AZA, azacitidine; LDAC, low dose cytarabine; MRD, measurable residual disease; and VEN, venetoclax.
Ijms 24 04790 g001
Table 1. Established and emerging molecular markers according to mutation type.
Table 1. Established and emerging molecular markers according to mutation type.
Mutation TypesClonal HematopoiesisAML-DefiningSignaling Pathway
Characteristics
ExamplesDNMT3A, TET2, ASXL1, IDH1, IDH2, TP53, SRSF2, and RUNX1, among othersNPM1, CBFB::MYH11, RUNX1::RUNX1T1, PML::RARA, and KMT2A fusions, among othersFLT3 (ITD and TKD), NRAS, KRAS, KIT, CBL, and PTPN11, among others
Clonal hierarchyPreleukemic, but overlaps with leukemia-initiating cloneLeukemia-initiating cloneSubclone
Presence in clonal hematopoiesisYes, but some mutations are deemed higher risk than others
(“high risk CCUS”)
RareRare
Clearance post morphological remissionFrequently persistsYesYes
Predictive valueLow value for DTA mutations (see text), and some likely have more oncogenic potentialHigh if detectable above the threshold or if serially increasing (MRD relapse)High positive predictive value but low negative predictive value
Stable at relapseYesYes, but note branched evolutionNo
Sample sourcegDNAUsually cDNAgDNA
Molecular assaysTargeted NGS panel
ddPCR (only for hotspots)
RT-qPCR (or RT-dPCR)
NGS-MRD panel (gDNA)
Targeted NGS panel
FLT3-ITD NGS-MRD panel
Abbreviations: CCUS, clonal cytopenias of undetermined significance; cDNA, complementary DNA; ddPCR, digital droplet PCR; DTA, DNMTA, TET2, and ASXL1; gDNA, genomic DNA; ITD, internal tandem duplication; MRD, measurable residual disease; NGS, next-generation sequencing; PCR, polymerase chain reaction; RT-qPCR, reverse transcription quantitative PCR; RT-dPCR, reverse transcription digital PCR; TKD, tyrosine kinase domain.
Table 2. Summary of studies evaluating MRD by targeted NGS panel in AML.
Table 2. Summary of studies evaluating MRD by targeted NGS panel in AML.
StudyNPB vs. BMTimepointMutationsThresholds
(% of N)
Risks (neg vs. pos)
(or < vs. > Threshold))
Klco, 2015 [24]50BMPC1Any<2.5%
(52)
Median OS 42.2 m vs. 10.5 m
Morita, 2018 [25]122BMPC1Any aNegative
(48)
2 y CIR 24% vs. 46%
2 y OS 77% vs. 60%
Jongen-Lavrencic, 2018 [23]430EitherPC2Non-DTANegative
(72)
4 y CIR 31.9% vs. 55.4%
4 y RFS 58.1% vs. 36.6%
4 y OS 66.1% vs. 41.9%
Rothenberg-Thurley, 2018 [26]126EitherFirst remissionAny b<2%
(60)
Median RFS 55.7 m vs. 11.7 m
Median OS NR vs. 31.3 m
Thol, 2018 [27]96EitherPre-HCTNon-DNMT3ANegative
(55)
5 y CIR 17% vs. 66%
5 y RFS 74% vs. 31%
5 y OS 78% vs. 41%
Kim, 2018 [28]104BMPost-HCT D21Any<2%
(85)
3 y CIR 16.0% vs. 56.2%
3 y OS 67.0% vs. 36.5%
Hourigan, 2020 [29]190PBPre-HCTAny cNegative
(35)
Neg: 3 y OS 56% vs. 63% (MAC vs. RIC)
Pos: 3 y OS 61% vs. 43% (MAC vs. RIC)
Pos: 3 y CIR 19% vs. 67% (MAC vs. RIC)
Heuser, 2021 [30]131EitherPost-HCT D90 ± 180Non-DTANegative
(80)
5 y CIR 25% vs. 62%
5 y RFS 68% vs. 35%
5 y OS 73% vs. 49%
Patkar, 2021 [31]201BMPC1AnyNegative
(29)
3 y CIR 25.7% vs. 47.5%
Median RFS NR vs. 17 m
Median OS NR vs. 27 m
Tsai, 2021 [32]335BMPC2Non-DTANegative
(71)
Median CIR 4.8 y—NR vs. 0.6–1.1 y
Median OS NR vs. 3.1–3.6 y
Hourigan, 2022 (Pre-MEASURE) [33]1075PBPre-HCTFLT3, NPM1, IDH1, IDH2, and KIT<0.01%
(70)
3 y CIR 23% vs. 62%
3 y RFS 59% vs. 25%
3 y OS 66% vs. 36%
Abbreviations: BM, bone marrow; CIR, cumulative incidence of relapse; EOT, end of treatment; HCT, hematopoietic cell transplantation; m, month; MAC, myeloablative conditioning; n.s., nonsignificant; neg, negative; NR, not reached; OS, overall survival; PB, peripheral blood; PC1, post-course 1; pos, positive; RFS, relapse-free survival; RIC: reduced-intensity conditioning; and y, year. a Additional analysis showed a stronger prognostic association when DTA was removed from the analysis. However, a separate analysis was not performed to compare the outcomes among those with DTA persistence vs. clearance. b Separate analysis showed no prognostic significance of DNMT3A mutation persistence. c Includes only 13 genes: DNMT3A, TET2, ASXL1, TP53, RUNX1, NRAS, KIT, IDH1, JAK2, SF3B1, IDH2, FLT3, and NPM1.
Table 4. Summary of MRD-guided therapy in AML.
Table 4. Summary of MRD-guided therapy in AML.
StudyPopulationNTreatmentMRD ResponseSurvival
Sockel, 2011 [109]NPM1 MRD relapse or persistence > 1%10AZA≥1 log10 decrease (70%)Not reported
Platzbecker, 2018 [14]RT-qPCR > 1% or donor chimerism loss53
(32 NPM1mut)
AZA36% MRDneg1 y RFS 46%
Bataller, 2020 [110]NPM1 MRD failure
(ELN 2017 favorable risk)
33
(Eight morphologic relapses)
20 chemo/HMA ± HCT
13 direct HCT
80% MRDneg
(8/10 chemo)
2 y OS 86%
Short, 2022 [111]MFC-MRD relapse16Seven HMA-based chemo
Nine direct HCT
43% MRDneg
(3/7 chemo)
5 y RFS 31%
5 y OS 45%
Dillon, 2020 [112]NPM1 MRD relapse3027 chemo + HCT
Three direct HCT
59% MRDneg
(16/27 chemo)
2 y OS 63%
Tiong, 2021 [11]NPM1 MRD relapseSevenVEN + HMA/LDAC86% CR MRDnegNot reported
Wood, 2022 [113]Molecular MRD failure (marker not specified)19
(103 morphologic disease)
VEN ± LDAC or HMA or other84% molecular remissionMedian OS 18.4 m
Tiong, 2022 [12]MRD relapse26
(20 NPM1mut)
VEN-LDAC≥1 log10 decrease (69%)
54% MRDneg
2 y EFS 54%
2 y OS 73%
Othman, 2022 [114]MRD (NPM1 or other gene fusions) failure with baseline FLT3mut48
(39 NPM1mut)
32 gilteritinib
Eight quizaritinib
Eight sorafenib
40% MRDneg2 y OS 80%
Abbreviations: AZA, azacitidine; chemo, chemotherapy; EFS, event-free survival; HCT, hematopoietic cell transplant; HMA, hypomethylating agent; LDAC, low dose cytarabine; m, months; MRD, measurable residual disease; mut, mutant; neg, negative; OS, overall survival; RFS, relapse-free survival; RT-qPCR, reverse transcription-quantitative polymerase chain reaction; VEN, venetoclax; and y, year.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tiong, I.S.; Loo, S. Targeting Measurable Residual Disease (MRD) in Acute Myeloid Leukemia (AML): Moving beyond Prognostication. Int. J. Mol. Sci. 2023, 24, 4790. https://doi.org/10.3390/ijms24054790

AMA Style

Tiong IS, Loo S. Targeting Measurable Residual Disease (MRD) in Acute Myeloid Leukemia (AML): Moving beyond Prognostication. International Journal of Molecular Sciences. 2023; 24(5):4790. https://doi.org/10.3390/ijms24054790

Chicago/Turabian Style

Tiong, Ing S., and Sun Loo. 2023. "Targeting Measurable Residual Disease (MRD) in Acute Myeloid Leukemia (AML): Moving beyond Prognostication" International Journal of Molecular Sciences 24, no. 5: 4790. https://doi.org/10.3390/ijms24054790

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop