Next Article in Journal
Brain Tumor Detection Based on Deep Learning Approaches and Magnetic Resonance Imaging
Next Article in Special Issue
Retinoid Therapy for Neuroblastoma: Historical Overview, Regulatory Challenges, and Prospects
Previous Article in Journal
Obstetric Results after Fertility-Sparing Management of Non-Epithelial Ovarian Cancer
Previous Article in Special Issue
Impact of Time to Surgery on Outcome in Wilms Tumor Treated with Preoperative Chemotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Risk-Stratified Therapy for Pediatric Acute Myeloid Leukemia

by
Daisuke Tomizawa
1,* and
Shin-Ichi Tsujimoto
2
1
Division of Leukemia and Lymphoma, Children’s Cancer Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
2
Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(16), 4171; https://doi.org/10.3390/cancers15164171
Submission received: 26 June 2023 / Revised: 8 August 2023 / Accepted: 12 August 2023 / Published: 18 August 2023
(This article belongs to the Special Issue Updates on Management and Clinical Trials in Pediatric Oncology)

Abstract

:

Simple Summary

Owing to the 40-year worldwide efforts for improving diagnosis and therapy for acute myeloid leukemia (AML), the second most common type of leukemia in children, overall survival rates of children with AML have now reached 70% to 80% in developed countries. This review article comprehensively describes the history and advances in the current state-of-the-art risk-stratified therapy for AML in children. However, it is likely that the traditional approaches have already reached their limits, and therefore, novel approaches are absolutely essential. The current state and future directions for incorporating novel molecular-targeted drugs into contemporary therapy through international collaboration are also extensively discussed. These aspects present key solutions for further improvements in outcomes of children with AML.

Abstract

Acute Myeloid Leukemia (AML) is the second most common type of leukemia in children. Recent advances in high-resolution genomic profiling techniques have uncovered the mutational landscape of pediatric AML as distinct from adult AML. Overall survival rates of children with AML have dramatically improved in the past 40 years, currently reaching 70% to 80% in developed countries. This was accomplished by the intensification of conventional chemotherapy, improvement in risk stratification using leukemia-specific cytogenetics/molecular genetics and measurable residual disease, appropriate use of allogeneic hematopoietic stem cell transplantation, and improvement in supportive care. However, the principle therapeutic approach for pediatric AML has not changed substantially for decades and improvement in event-free survival is rather modest. Further refinements in risk stratification and the introduction of emerging novel therapies to contemporary therapy, through international collaboration, would be key solutions for further improvements in outcomes.

1. Introduction

Acute Myeloid Leukemia (AML) is a form of hematopoietic malignancy characterized by clonal proliferation of immature myeloid cells. As can be seen from the fact that the median diagnostic age of AML is over 60 years old, AML is the most common type of leukemia in adults, whereas it is the second most common leukemia subtype in children, accounting for 20–25% of pediatric leukemia cases, with an incidence of approximately seven cases per 1,000,000 children per year [1]. There are no sex differences in prevalence of AML in children. Although AML in children may arise from certain constitutional chromosomal abnormalities (e.g., Down syndrome [trisomy 21]), familial predisposition syndromes or inherited gene mutations/translocations (e.g., inherited bone marrow failure syndromes), acquired conditions (e.g., myelodysplastic syndrome [MDS]), or exposure to chemotherapy/radiotherapy (therapy-related myeloid neoplasms), most of the children develop AML as a de novo disease without apparent etiology. A multi-step process of an accumulation of chromosomal and genomic alterations within immature myeloid cells results in the development of AML. Recently, novel AML classifications (Fifth edition of the World Health Organization [WHO] Classification of Haematolymphoid Tumours and International Consensus Classification [ICC] of Myeloid Neoplasms and Acute Leukemias) have been proposed [2,3,4]. Despite some existing differences, both classifications place more emphasis on molecular/genetic criteria compared to the previous ones. However, one should note that recent evidence suggests that AML in children and that in adults are distinct at least in terms of mutational landscape [5].
Overall Survival (OS) rates of children with AML have dramatically improved in the past 40 years, currently reaching 70–80% in developed countries [6,7,8,9,10,11,12,13,14,15,16,17]. This was accomplished mainly by the intensification of conventional chemotherapy, improvement in risk stratification, appropriate use of allogeneic Hematopoietic Stem Cell Transplantation (HSCT), and improvement in supportive care. However, despite the cytogenetic/mutational heterogeneity of the disease, the principle treatment for pediatric AML has not changed substantially for decades and improvement in Event-Free Survival (EFS) is rather modest [18]. In this review, the state-of-the-art risk-stratified therapy for children with AML other than Acute Promyelocytic Leukemia (APL) and Myeloid Leukemia associated with Down Syndrome (ML-DS) will be highlighted, including the historical background and future perspectives emphasizing risk stratification and molecularly targeted therapies.

2. Prognostic Factors and Risk Stratification in Pediatric AML

Risk stratification is one of the key elements for successful treatment in AML, and its aim is (A) to assign patients to therapies with sufficient intensity, (B) to avoid excess toxicities by avoiding therapies with unnecessary intensity, and recently, (C) to identify targetable lesions to incorporate targeted therapies. To properly risk-stratify patients, it is necessary to predict the treatment failure risk of patients by evaluating various prognostic factors. Prognostic factors can be subdivided into patient-associated factors (e.g., age, ethnicity) and disease-related factors (e.g., leukemia-specific cytogenetics/molecular genetics, drug resistance). Age at diagnosis is prognostic, i.e., the survival rate of children is significantly better than young adults, and that of young adults is better than older adults. However, within children < 15 years old, the impact of age difference is not significant [19]. Regarding ethnicities, analysis of Children’s Oncology Group (COG) studies mainly involving North America showed that Hispanic and African-American children had significantly worse OS rates compared to Caucasian children, and that access to chemotherapy, differences in supportive care, leukemic phenotype, and reduced compliance were unlikely to be the explanations [20]. There is no data that directly analyzed differences between Asian children and other ethnicities; however, the literature shows a higher prevalence of t(8;21) (RUNX1::RUNX1T1)-positive AML in Asian populations (approximately 30% compared to 12–14% of the U.S. or European patients) [1,21,22]. Overall, the impact of patient-associated factors is not as large as the disease-related factors in children with AML.

2.1. Leukemia-Specific Cytogenetics/Molecular Genetics

Leukemia-associated genetic profiles of 369 patients in the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) trial AML-05 are listed in Figure 1 [22]. The distribution of genetic profiles is similar to the other groups in the U.S. or Europe except that the proportion of patients with RUNX1::RUNX1T1 is high in the Japanese cohort, as mentioned previously [1].
Analyses of clinical trials conducted from the late 1980s to the early 2000s revealed the prognostic significance of recurrent chromosomal aberrations in AML. In the United Kingdom (UK) studies MRC-AML10 and MRC-AML12, children with t(8;21)(q22;q22) (RUNX1::RUNX1T1) and inv(16)(p13q22) (CBFB::MYH11), the core-binding factor (CBF)-AML, had the best prognosis (80% OS rate), and the patients with chromosome 12 or 5q abnormalities, t(6;9)(p23;q34) (DEK::NUP214), monosomy 7, and t(9;22)(q34;q11) (BCR::ABL1) had the worst prognosis (36% OS rate) [23]. However, the majority (nearly 70%) of the patients were classified as intermediate risk (56% OS rate), which includes patients with normal karyotypes, chromosome 11q23 abnormalities, Acute Megakaryoblastic Leukemia (AMKL), and others. International collaborative efforts have significantly contributed to further uncovering the prognoses of certain AML subtypes. One of the first successes was the retrospective analysis of chromosome 11q23 abnormalities by the International BFM study group (I-BFM) consisting of 11 cooperative study groups in 15 countries [24]. 11q23 abnormalities or KMT2A gene rearrangements (KMT2A-r) account for 15–20% of pediatric AML, and recent studies have identified more than 100 fusion gene partners [25]. The 756 patients included in this study showed an “intermediate” prognosis of 44% EFS and 56% OS rates, but large EFS/OS differences were identified among the following different translocation partners: t(1;11)(q21;q23) (KMT2A::MLLT11) showed the best prognosis and t(6;11)(q27;q23) (KMT2A::AFDN), t(10;11)(p12;q23) (KMT2A::MLLT10), and t(10;11)(p11.2;q23) (KMT2A::ABI1) showed unfavorable prognoses. The other following I-BFM projects, such as t(8;16)(p11;p13)/CREBBP::KAT6A (intermediate OS, spontaneous remission in neonatal cases), t(6;9)(p23;q34)/DEK::NUP214 (high-risk of relapse, improved EFS by HSCT), KIT and RAS mutations in t(8;21) (not associated with a worse outcome), t(16;21)(p11;q22)/FUS::ERG (extremely poor prognosis), t(16;21)(q24;q22)/RUNX1::CBFA2T3 (favorable outcome), and hypodiploidy (poor prognosis), have also elucidated the clinical features and prognosis of these relatively rare subsets [26,27,28,29,30].
Finally, advances in molecular/genetic analyses have revealed many of the prognostic genetic markers in pediatric AML. Among non-DS AMKL (rare in adults, but 4–15% in children), inv(16)(p13q24)/CBFA2T3::GLIS2 (18.4%), KMT2A-r (17.2%), and t(11;12)(p15;p13)/NUP98::KDM5A (11.5%) formed a poor prognostic subgroup, and mutations of the GATA1 gene that generate the short form of GATA1 (GATA1s; 9.2%) and t(1;22)(p13;q13)/RBM15::MRTFA (10.2%) formed a good prognostic group [31,32,33]. Many of the important genetic prognostic markers were identified among the cytogenetically “normal” AML (CN-AML; approximately 40% in adult AML and 20% in pediatric AML) as well. As a poor prognostic marker, internal tandem duplication (ITD) of the FLT3 gene (FLT3-ITD) is found in approximately 10% of pediatric AML and 20–30% of adult AML and is also important as a targetable marker [34]. As favorable prognostic markers, a mutation of the NPM1 gene that generates cytoplasmic NPM1 (NPM1c) is found in approximately 5–8% of pediatric AML and 20–30% in adult AML, and biallelic CEBPA mutations in approximately 5% of both pediatric and adult AML [35,36]. Recent studies have revealed that CEBPA-basic leucine zipper (CEBPA-bZip) mutations are associated with favorable clinical outcomes regardless of monoallelic or biallelic mutational status (80% of the patients have a double mutation) [37]. Mutations in IDH1, IDH2, and DNMT3A are found in 7–14%, 8–19%, and 18–22%, respectively, of adult AML (most frequently seen in CN-AML) [38]. The prognostic significance of these mutations are not fully established, but all these mutations are extremely rare in children [39,40]. Many of the newly discovered gene mutations are less frequent in children. In addition to the already mentioned gene mutations, the TP53 mutation is found in approximately 8% of adult AML, is associated with older age, has complex and monosomal karyotypes, has a very poor outcome, and has been given a strong emphasis in recently proposed European LeukemiaNet (ELN) 2022 recommendations, but it is rarely seen in pediatric AML [41,42]. However, NUP98::NSD1 encoded by cryptic t(5;11)(q35;p15.5) was discovered in 16.1% of pediatric CN-AML, whereas it was only 2% of CN-AML in adults [43]. This fusion is associated with high leukocyte count, monocytic leukemia (M4 or M5 in French-American-British [FAB] classification), FLT3-ITD, and a very poor prognosis. A recent study analyzing the COG trial cohorts showed poor outcomes of not only NUP98::NSD1 and NUP98::KDM5A cases but of cases with other NUP98-fusion-positive AML (unlike NSD1 and KDM5A cases, other NUP98-fusions are typically not cryptic) [44]. Tandem duplication in the UBTF gene (UBTF-TD) is another example of a mutation that is predominant in pediatric AML (approximately 4% of newly diagnosed and 9% of relapsed pediatric AML). This mutation is associated with normal karyotype or trisomy 8 with co-occurring WT1 mutations or FLT3-ITD and confers an unfavorable prognosis [45]. International cooperation will become increasingly important to further identify a subgroup of pediatric AML with prognostic impact, which generally would include small numbers of patients.

2.2. Treatment Response Including Measurable Residual Disease

Assessment of treatment response is regarded as an in vivo method to measure leukemia drug resistance and is widely used to risk-stratify patients with AML. In the UK MRC-AML10 study, bone marrow morphological response after initial induction therapy was significantly associated with both OS and relapse rates, and ≥15% bone marrow blasts after initial induction without favorable genetic abnormalities were allocated to the poor-risk arm in the MRC-AML12 study [46,47]. In the German BFM studies, analysis of AML-BFM83 and AML-BFM87 studies showed that residual bone marrow blasts (≥5%) at day 15 of initial induction were associated with reduced EFS rates and was therefore included in the risk group definition since the AML-BFM93 study [48]. A similar approach to risk stratification using morphological bone marrow response was used in other pediatric AML studies as well after the 1990s.
Although >85% of children with AML achieve morphological remission after one or two courses of induction therapy, 30–40% of the patients eventually experience overt relapse. A growing need for more accurate methods to assess treatment response led to the development of molecular or immunophenotypic determination of Measurable Residual Disease (MRD) in the late 1990s to early 2000s. Regarding molecular MRD assessment, a Polymerase Chain Reaction (PCR) approach, and recently a next-generation sequencing (NGS) approach, targeting certain AML-specific genetic markers (fusion transcripts or gene mutations) can be taken with a sensitivity of 0.01–0.001% in PCR and 0.01–0.0001% in NGS. The most problematic issue of molecular MRD is its limited applicability in children, i.e., major fusions (e.g., RUNX1::RUNX1T1, CBFB::MYH11, PML::RARA, KMT2A::MLLT3) are found in less than 40% of children with AML and major gene mutations found in adults (e.g., NPM1c, FLT3-ITD) are far less prevalent in children. In addition, it is well recognized that RUNX1::RUNX1T1 and CBFB::MYH11 transcripts may persist in the patient’s bone marrow while in long-term remission, and because of this, false-positive results may come out. The St. Jude Children’s Research Hospital (SJCRH) study in the U.S., which compared flow-cytometric MRD and molecular MRD, showed discrepant results (only 9.6% of the PCR-positive samples were flow-positive) and that PCR-MRD results did not have a prognostic impact when flow-MRD was negative [49]. The ELN MRD Working Party suggests a failure to reach a 3-log to 4-log reduction between the sample at diagnosis and at the end of treatment in adult patients with CBF-AML would be a relevant marker for subsequent relapse, but caution is needed whether it could be applied to children as well [50]. As a consequence, multiparametric flow cytometry (MFC)-MRD is considered to be the more preferred method for children with AML because of its wide applicability (>95% of the patients), although sensitivity is potentially lower (0.1–0.01%) than molecular MRD. Currently, there are two MFC-MRD approaches to target leukemia cells, namely, the leukemia-associated immunophenotype (LAIP) approach used in many study groups and the different-from-normal (DfN) aberrant immunophenotype approach used in the COG studies [51,52,53,54,55,56,57]. The LAIP approach is more complicated because it needs to select a patient-specific antigen combination. In contrast, the DfN approach employs a standardized panel which could potentially be applied to all patients regardless of the leukemia blast immunophenotype at diagnosis (i.e., does not require access to the diagnostic specimen) and has the strength that the method does not rely on the stability of a diagnostic LAIP during treatment, and therefore, the blasts can be detected even if an immunophenotypic shift occurs. Whichever approaches are applied, positive MRD at the end of one or two courses of induction therapies is shown to be the strongest predictor of poor outcomes in every previously reported clinical trial. Notably, the SJCRH AML02 study using the LAIP approach and the COG AAML0531 study using the DfN approach both showed the limited impact of morphological remission status on the negative MRD condition at the end of initial induction therapy [49,58].

2.3. Risk Stratification in Pediatric AML

Current risk stratifications used in most of the pediatric AML studies are based on combinations of leukemia-specific cytogenetic/molecular genetic abnormalities and MRD-based treatment response (Table 1). The NOPHO-DBH AML 2012 study (NCT01828489) by the Nordic Society of Paediatric Haematology and Oncology (NOPHO), Belgium, the Netherlands, and others are quite unique in that their risk stratification is strongly based on treatment response. Pediatric AML risk stratification has been focusing on determining the high-risk subsets of the patients assigned to receive HSCT in the first CR. Importantly, one should note whether HSCT truly improves the outcome of the high-risk patients. The outcome of a certain subset of the patients (e.g., FUS::ERG) has not improved by simply allocating the patients to receive HSCT and novel therapeutic strategies are urgently needed for these patients [59]. In the current genomic era, future risk stratification should focus more on identifying targetable lesions to incorporate molecularly targeted therapies. Success for APL using an introduction of All-Trans-Retinoic Acid (ATRA)-combined chemotherapy, and more recently of ATRA/arsenic trioxide combination therapy, is an ideal model, and many groups are starting to take this approach, as is the case with FLT3-ITD.

3. Current Standard Therapy for Pediatric AML

3.1. Chemotherapy

Multi-agent combination chemotherapy is still a mainstay of treating children with AML. Key drugs are cytarabine and anthracyclines. Similar to adult AML, standard initial induction chemotherapy in children is based on the “3 + 7” regimen (seven days of low-to-intermediate dose cytarabine [LDAC] concurrent with three days of anthracyclines), but a third drug (e.g., etoposide) is often combined although its role is not fully established (Figure 2). The Japanese group introduced a unique prolonged schedule of induction therapy “ECM” (12 days in total) in the ANLL91 study in the early 1990s, on the basis of a high proportion of FAB-M4/M5 subtypes in children and frequent high leukocyte presentation and expected high efficacy of etoposide against monocytic AML [60]. Since then, this regimen has been used in the Japanese trials (AML99, AML-05, AML-12, and the currently ongoing AML-20), and was recently adopted in the NOPHO-DBH AML 2012 study as well [6,7,8,61,62]. Because of the significant prognostic impact of induction therapies and the limited number of pediatric AML patients, randomized questions to improve the outcome of children with AML have been mainly set at induction phases in the past pediatric AML studies worldwide. However, most of the study questions raised in the past trials have failed to show the impact on improved survival in pediatric AML (Table 2), specifically, the role of high-dose cytarabine (HDAC) [8,11,63,64,65], use of different types of anthracyclines [13,47,66], and addition of other cytotoxic drugs [10,12,67]. An exception is the addition of gemtuzumab ozogamicin (GO), an anti-CD33 antibody-drug conjugate (ADC). Cell surface antigen CD33 is expressed in more than 80% of the patients with AML, which makes this agent an attractive option. A randomized study, COG AAML0531, showed that the addition of GO 3 mg/m2 to ADE induction (and also to the second consolidation course) significantly improved the EFS (but not OS) of children with newly diagnosed AML [9]. Consequently, GO is currently approved by the U.S. Food and Drug Agency (FDA) for use in this setting and is regarded as a standard of care in the U.S. In the MyeChild01 study by the UK, Ireland, and France, a dose-finding study of GO (cohort 1, GO 3 mg/m2/dose on day 4 of initial induction consisting of cytarabine and mitoxantrone; cohort 2, GO on days 4 and 7; cohort 3, GO on days 1, 4, and 7) is being conducted (NCT02724163).
Post-induction chemotherapies, namely, consolidation or intensification courses, are provided to all the patients who have achieved morphological CR to further consolidate the remission status. Drugs used are almost the same as induction chemotherapies, consisting mainly of cytarabine (generally includes HDAC) with or without anthracyclines and/or other additional drugs. Many of the questions regarding post-induction chemotherapies remain unsolved (Table 2), including the number of chemotherapy courses [6,47,70] and the addition of other cytotoxic drugs. Regarding the role of GO in post-induction therapy, COG AAML0531 showed the benefit of adding GO in a second consolidation course with HDAC and mitoxantrone (also in initial induction) [9]. However, the addition of GO (5 mg/m2/dose on days 1 and 21) at the very end of consolidation chemotherapies failed to improve both EFS and OS in the NOPHO-AML2004 study [71]. Currently, the Japan Children’s Cancer Group (JCCG) is evaluating the role of GO in post-induction phases by randomizing the intermediate-risk and high-risk patients to receive three additional courses of GO (3 mg/m2/dose)-combined or non-combined consolidation chemotherapy (jRCTs041210015) [62]
Unlike the treatment of ALL, maintenance therapy is not a part of the standard of care for AML [72]. However, maintenance therapy using targeted drugs, including FLT3 inhibitors, might offer a benefit [73].
As mentioned previously, there is no clear evidence for the best anthracyclines of choice in AML chemotherapy. However, when comparing cumulative doses of different anthracyclines, the equivalence ratio is an issue, particularly in terms of late cardiotoxicity risks. For mitoxantrone and idarubicin, the ratio of 1:4–5 has been generally used for doxorubicin-equivalent doses. Recently, Feijen et al. reported a higher mean mitoxantrone conversion ratio of 10.5 (ratio of 0.5 for daunorubicin) based on cardiomyopathy risk assessment of the 28,423 childhood cancer survivors from the Childhood Cancer Survivor Study (CCSS), St. Jude Lifetime (SJLIFE), and Dutch Children’s Oncology Group (DCOG)-LATER study cohorts [69]. This data should be taken cautiously because the survivors included in this study were treated quite a long time ago (mostly between the 1960s and late 1990s) with various disease backgrounds (not only AML), and there have been no reports on increased cardiotoxicities from the UK or the Japanese group that had used mitoxantrone in their AML protocol since the late 1990s. The risk of late cardiotoxicity is something one should take into account for choosing kinds and doses of anthracyclines.
Because the principle AML therapy has been systemic therapy, the need for local therapy, including central nervous system (CNS)-directed therapy, is not clear. Most groups usually include intrathecal therapy (ITT) with cytarabine with or without methotrexate and corticosteroids in every chemotherapy course, but it is not evidence-based. In fact, adult AML studies generally do not include ITT. However, it is well recognized that children with AML (compared to adults) possess features with a higher risk of CNS disease and/or CNS relapse, such as higher leukocyte count at diagnosis and a higher prevalence of monocytic leukemia [74].

3.2. Hematopoietic Stem Cell Transplantation

The anti-leukemia effect of allogeneic HSCT relies on the cytotoxic effect of conditioning therapy and the immunological graft-versus-leukemia (GVL) effect by donor-derived cytotoxic immune cells; an approximately 60–70% DFS rate and 10–15% treatment-related mortality (TRM) is expected if transplanted in first or second CR [74]. Despite its potential risk of both acute and late toxicities, HSCT still plays an important role as a curative post-remission therapy for children with AML, although its indication is restricted to the high-risk subset (Table 1) [75]. Historically, both total body irradiation (TBI)-based and non-TBI-based (usually busulfan-based) myeloablative conditioning (MAC) were used in HSCT for children with AML. However, unlike ALL, both pediatric and adult studies (mainly retrospective studies) have shown similar or better results in favor of non-TBI-based conditioning for transplanting children with AML in terms of both efficacy and toxicity [76,77,78]. Therefore, intravenous busulfan (IV-BU)-based MAC is currently considered as standard; IV-BU in combination with melphalan and/or cyclophosphamide is generally used. Reduced-intensity conditioning (RIC) is an attractive option for children with AML, particularly in terms of reducing risks of late effects. Several retrospective analyses showed comparable outcomes between MAC and RIC, but an adult phase 3 randomized study for AML and MDS by the Blood and Marrow Transplant Clinical Trials Network (BMT CTN) demonstrated statistically significantly better relapse-free survival (RFS) in MAC [79,80,81]. BU-based MAC and RIC were randomly compared in children with AML in the MyeChild01 study, currently awaiting results.

4. Novel Therapy for Pediatric AML

The outcomes of recently conducted pediatric AML studies are listed in Table 3. EFS ranges from 45% to 63% and OS from 65% to 80% [6,7,8,9,10,11,12,13,14,15,16,17]. As further improvements in outcomes for children with AML by conventional approaches are unlikely, the introduction of effective novel therapies to the current standard AML therapy would be a key solution. Several new classes of agents currently under development will be discussed.

4.1. FLT3 Inhibitors

FLT3 is a transmembrane ligand-activated receptor tyrosine kinase that is normally expressed by hematopoietic stem or progenitor cells and plays an important role in the early stages of both myeloid and lymphoid lineage development. An extracellular ligand binds and activates FLT3, promoting cell survival, proliferation, and differentiation through various signaling pathways, including PI3K, RAS, and STAT5. Mutations of the FLT3 gene are found in approximately 30% of newly diagnosed adult AML cases (25% as ITDs and 10% as point mutations in the tyrosine kinase domain [TKD]) [38]. Frequencies in children with AML are lower, i.e., ITDs are found in 10% and TKD mutations in 6% of the cases [34]. Both FLT3-ITD and TKD mutations constitutively activate FLT3 kinase activity, resulting in proliferation and survival of AML. The presence of FLT3-ITD, not FLT3-TKD, is associated with poor outcomes both in children and adults with AML. FLT3 inhibitors are molecular-targeted agents that inhibit FLT3 signaling and are of two types. Type I inhibitors (midostaurin, gilteritinib) bind the FLT3 receptor in both the active and inactive conformational state of the FLT3 kinase domain, either near the activation loop or the ATP binding pocket, and are active against both ITD and TKD mutations. Type II inhibitors (sorafenib, quizartinib) bind specifically for the inactive conformation in a region adjacent to the ATP-binding domain. As a result of this binding affinity, type II FLT3 inhibitors prevent the activity of only ITD mutations but do not target TKD mutations. In terms of development history, midostaurin and sorafenib belong to the first generation, which was identified to have an FLT3 target among the various compounds with multi-targets. Quizartinib and gilteritinib belong to the second generation, which was originally designed to target FLT3, and therefore, more FLT3-specific compared to the first-generation inhibitors. Midostaurin is not active when used as monotherapy, but was developed for use in combination therapy [82]. Sorafenib, quizartinib, and gilteritinib all showed approximately 50% response rate for relapsed/refractory AML as monotherapy [83,84,85]. Each drug has specific toxicities, such as skin rash in sorafenib and QTcF prolongation in quizartinib for example. Current development of FLT3 inhibitors is focused on combination chemotherapy. In the QuANTUM-First trial, a randomized phase 3 study for newly diagnosed FLT3-ITD positive adult AML on a quizartinib combination, the addition of quizartinib significantly improved EFS and OS, and toxicities were comparable between the two arms [86]. Pediatric development of FLT3 inhibitors is lagging behind compared to adults. However, in the COG AAML1031 study, children with newly diagnosed high allelic ratio FLT3-ITD positive AML were eligible for receiving sorafenib combined therapy, and improved EFS was observed for the 72 children who took sorafenib compared to the 76 children who did not [71]. Table 4 shows the ongoing FLT3 inhibitor trials for children with AML.

4.2. BCL2 Inhibitors

B-cell/CLL lymphoma-2 (BCL-2) family proteins regulate the intrinsic apoptosis pathway by integrating diverse pro-survival or pro-apoptotic intracellular signals. In AML, increased expression of BCL2 family proteins in leukemic blasts has been reported, and the majority of AML stem cells express aberrantly high levels of BCL2 and are dependent on BCL2 for survival. Furthermore, high expression of BCL2 has been associated with an inferior response to chemotherapy and poor survival among patients with AML. Venetoclax, a selective small-molecule BCL2 inhibitor, has been shown in preclinical studies to induce apoptosis in malignant cells that are dependent on BCL2 for survival. However, the single-agent venetoclax has had only modest activity in AML. Through downregulation of myeloid-cell leukemia 1 (MCL1) and induced expression of the pro-death proteins NOXA and PUMA, azacitidine or cytarabine synergistically inhibits the pro-survival proteins MCL1 and BCL-XL, thereby increasing the dependence of leukemia cells on BCL2. In fact, venetoclax combined with azacytidine or in combination with LDAC significantly prolonged the survival of adult patients with AML unfit for standard chemotherapy [87,88]. In the SJCRH phase 1 study for children with relapsed/refractory AML, venetoclax in combination with LDAC or HDAC was tested, and 360 mg/m2 venetoclax in combination with HDAC with or without idarubicin was determined to be the recommended phase 2 dose [89]. The overall response was 69%. Febrile neutropenia and invasive fungal infections were observed in 16% of the patients and one treatment-related death was observed. However, the treatment was tolerable overall. Currently, a phase 3 trial of venetoclax in combination with fludarabine, cytarabine, and GO for children with relapsed AML is ongoing (NCT05183035). This study is conducted as one of the sub-studies of PedAL/EuPAL initiatives, a global precision medicine master clinical trial that will test multiple targeted therapies simultaneously at various clinical sites, mainly in the U.S. and Europe.

4.3. Menin Inhibitors

KMT2A-r AML accounts for 20–25% of pediatric AML. Recently, the updated retrospective study by the I-BFM was published; notably, nearly 50% of the patients failed the therapy even if their MRD was negative after the second induction [90]. Menin inhibitors are the most attractive class of agents for leukemia with KMT2A-r. Menin is a product of the MEN1 tumor suppressor gene, which binds to the rearranged KMT2A complex and leads to the upregulation of leukemogenic genes (such as HOX and MEIS1), and thus to the subsequent development of acute leukemia. Menin inhibitors have shown selective, profound single-agent activity in KMT2A-r PDX models [91]. Menin inhibitors are potentially active against other subtypes of AML, such as NPM1c and NUP98-rearranged AML. NPM1c AML accounts for 20–30% of adult AML and 6% of pediatric AML and is generally associated with good prognosis. NPM1-coding protein nucleophosmin shuttles between the nucleus and cytoplasm during the cell cycle and is involved in diverse cellular processes, such as ribosome biogenesis, centrosome duplication, protein chaperoning, histone assembly, cell proliferation, and regulation of tumor suppressors TP53. However, mutated NPM1 (NPM1c) persists in the cytoplasm, and although the mechanism is not clear (but presumed to be a loss of function), NPM1c is associated with upregulation of HOX genes in a menin-dependent manner [92]. NUP98-rearranged AML accounts for less than 1% of adult AML and 7% of pediatric AML and is associated with unfavorable outcomes. It is known that NUP98 fusion proteins interact with KMT2A chromatin complexes and promote leukemogenesis. Inhibition of menin-KMT2A impairs leukemogenic gene expression and disrupts chromatin binding of menin, KMT2A, and NUP98 fusion proteins in mouse models [93]. Given the strong preclinical rationale justifying the use of menin inhibitors as a novel class of targeted therapy in acute leukemias, multiple clinical trials with these agents are in progress. The Syndax trial AUGMENT-101 is an industry-initiated first-in-human phase 1 clinical trial of the oral menin-inhibitor product revumenib (SNDX-5613) for both adults and children with relapsed/refractory acute leukemia with KMT2A-r or NPM1c [94]. Because revumenib is a substrate of cytochrome P450 3A4 (CYP3A4), two parallel dose-escalation cohorts, one without (Arm A) and one with (Arm B) strong CYP3A4 inhibitors, were conducted. There were no discontinuations or deaths due to treatment-related adverse events. Dose-limiting toxicities were asymptomatic grade 3 prolonged QTc in both arms. Notably, differentiation syndrome was observed in 16% of the patients. Overall response rate was 59%, and 73% of the patients achieving CR/CRh were MRD negative. Other menin inhibitor trials in children are also in progress.

4.4. Others

In addition to the cell signaling inhibitors against FLT3, BCL2, and menin, examples of novel therapies of interest for pediatric AML are immunotherapies (e.g., ADC, bispecific antibodies/T-cell engagers, chimeric antigen receptor T-cells [CAR-T]) targeting CD123 (expressed in nearly all AML subsets and leukemia stem cells), CD33, FLT3, or FOLR1 (targeting CBFA2T3::GLIS2 fusion-positive AML), checkpoint inhibitors, cell-signaling inhibitors targeting MEK (NRAS and KRAS mutations are among the most common mutations in pediatric AML), and epigenetic modifiers (e.g., DNA methyltransferase inhibitors, histone deacetylase inhibitors, IDH1/IDH2 inhibitors) [95]. Finally, owing to the limited number of patients within each AML subgroup with a specific targetable disease, international cooperation (e.g., the PedAL/EuPAL initiatives) is crucial for effective drug development.

5. Conclusions and Future Directions

A quarter century of global efforts on clinical trials have contributed to improved outcomes for children with AML but are still tentative. Refinement in risk stratification based on leukemia biology and MRD, as well as the introduction of emerging novel therapies to contemporary therapy, through international collaboration, would be a key solution for further improvement in outcomes.

Author Contributions

Conception and design, D.T.; writing—original draft preparation, D.T., writing—review and editing, all authors; funding acquisition, D.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the Japan Agency for Medical Research and Development (AMED) (grant number JP23ck0106853 to Daisuke Tomizawa).

Acknowledgments

The authors thank the senior medical editor from the Center for Postgraduate Education and Training of the National Center for Child Health and Development for editing this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Creutzig, U.; van den Heuvel-Eibrink, M.M.; Gibson, B.; Dworzak, M.N.; Adachi, S.; de Bont, E.; Harbott, J.; Hasle, H.; Johnston, D.; Kinoshita, A.; et al. Diagnosis and management of acute myeloid leukemia in children and adolescents: Recommendations from an international expert panel. Blood 2012, 120, 3187–3205. [Google Scholar] [CrossRef] [PubMed]
  2. Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef] [PubMed]
  3. Pfister, S.M.; Reyes-Múgica, M.; Chan, J.K.; Hasle, H.; Lazar, A.J.; Rossi, S.; Ferrari, A.; Jarzembowski, J.A.; Pritchard-Jones, K.; Hill, D.A.; et al. A Summary of the Inaugural WHO Classification of Pediatric Tumors: Transitioning from the Optical into the Molecular Era. Cancer Discov. 2022, 12, 331–355. [Google Scholar] [CrossRef] [PubMed]
  4. Arber, D.A.; Orazi, A.; Hasserjian, R.P.; Borowitz, M.J.; Calvo, K.R.; Kvasnicka, H.-M.; Wang, S.A.; Bagg, A.; Barbui, T.; Branford, S.; et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: Integrating morphologic, clinical, and genomic data. Blood 2022, 140, 1200–1228. [Google Scholar] [CrossRef]
  5. Bolouri, H.; Farrar, J.E.; Triche, T., Jr.; Ries, R.E.; Lim, E.L.; Alonzo, T.A.; Ma, Y.; Moore, R.; Mungall, A.J.; Marra, M.A.; et al. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat. Med. 2018, 24, 103–112. [Google Scholar] [CrossRef]
  6. Tomizawa, D.; Tawa, A.; Watanabe, T.; Saito, A.M.; Kudo, K.; Taga, T.; Iwamoto, S.; Shimada, A.; Terui, K.; Moritake, H.; et al. Excess treatment reduction including anthracyclines results in higher incidence of relapse in core binding factor acute myeloid leukemia in children. Leukemia 2013, 27, 2413–2416. [Google Scholar] [CrossRef]
  7. Hasegawa, D.; Tawa, A.; Tomizawa, D.; Watanabe, T.; Saito, A.M.; Kudo, K.; Taga, T.; Iwamoto, S.; Shimada, A.; Terui, K.; et al. Attempts to optimize postinduction treatment in childhood acute myeloid leukemia without core-binding factors: A report from the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG). Pediatr. Blood Cancer 2020, 67, e28692. [Google Scholar] [CrossRef]
  8. Tomizawa, D.; Tanaka, S.; Hasegawa, D.; Iwamoto, S.; Hiramatsu, H.; Kiyokawa, N.; Miyachi, H.; Horibe, K.; Saito, A.M.; Taga, T.; et al. Evaluation of high-dose cytarabine in induction therapy for children with de novo acute myeloid leukemia: A study protocol of the Japan Children’s Cancer Group Multi-Center Seamless Phase II–III Randomized Trial (JPLSG AML-12). Jpn. J. Clin. Oncol. 2018, 48, 587–593. [Google Scholar] [CrossRef]
  9. Gamis, A.S.; Alonzo, T.A.; Meshinchi, S.; Sung, L.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Kahwash, S.B.; Heerema-McKenney, A.; Winter, L.; et al. Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: Results from the randomized phase III Children’s Oncology Group trial AAML0531. J. Clin. Oncol. 2014, 32, 3021–3032. [Google Scholar] [CrossRef]
  10. Aplenc, R.; Meshinchi, S.; Sung, L.; Alonzo, T.; Choi, J.; Fisher, B.; Gerbing, R.; Hirsch, B.; Horton, T.; Kahwash, S.; et al. Bortezomib with standard chemotherapy for children with acute myeloid leukemia does not improve treatment outcomes: A report from the Children’s Oncology Group. Haematologica 2020, 105, 1879–1886. [Google Scholar] [CrossRef]
  11. Rubnitz, J.E.; Inaba, H.; Dahl, G.; Ribeiro, R.C.; Bowman, W.P.; Taub, J.; Pounds, S.; Razzouk, B.I.; Lacayo, N.J.; Cao, X.; et al. Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: Results of the AML02 multicentre trial. Lancet Oncol. 2010, 11, 543–552. [Google Scholar] [CrossRef] [PubMed]
  12. Rubnitz, J.E.; Lacayo, N.J.; Inaba, H.; Heym, K.; Ribeiro, R.C.; Taub, J.; McNeer, J.; Degar, B.; Schiff, D.; Yeoh, A.E.-J.; et al. Clofarabine Can Replace Anthracyclines and Etoposide in Remission Induction Therapy for Childhood Acute Myeloid Leukemia: The AML08 Multicenter, Randomized Phase III Trial. J. Clin. Oncol. 2019, 37, 2072–2081. [Google Scholar] [CrossRef] [PubMed]
  13. Creutzig, U.; Zimmermann, M.; Bourquin, J.-P.; Dworzak, M.N.; Fleischhack, G.; Graf, N.; Klingebiel, T.; Kremens, B.; Lehrnbecher, T.; Von Neuhoff, C.; et al. Randomized trial comparing liposomal daunorubicin with idarubicin as induction for pediatric acute myeloid leukemia: Results from Study AML-BFM 2004. Blood 2013, 122, 37–43. [Google Scholar] [CrossRef] [PubMed]
  14. Pession, A.; Masetti, R.; Rizzari, C.; Putti, M.C.; Casale, F.; Fagioli, F.; Luciani, M.; Nigro, L.L.; Menna, G.; Micalizzi, C.; et al. Results of the AIEOP AML 2002/01 multicenter prospective trial for the treatment of children with acute myeloid leukemia. Blood 2013, 122, 170–178. [Google Scholar] [CrossRef] [PubMed]
  15. Abrahamsson, J.; Forestier, E.; Heldrup, J.; Jahnukainen, K.; Jónsson, G.; Lausen, B.; Palle, J.; Zeller, B.; Hasle, H. Response-guided induction therapy in pediatric acute myeloid leukemia with excellent remission rate. J. Clin. Oncol. 2011, 29, 310–315. [Google Scholar] [CrossRef]
  16. De Moerloose, B.; Reedijk, A.; De Bock, G.H.; Lammens, T.; de Haas, V.; Denys, B.; Dedeken, L.; van den Heuvel-Eibrink, L.L.; te Loo, M.; Uyttebroeck, A.; et al. Response-guided chemotherapy for pediatric acute myeloid leukemia without hematopoietic stem cell transplantation in first complete remission: Results from protocol DB AML-01. Pediatr. Blood Cancer 2019, 66, e27605. [Google Scholar] [CrossRef]
  17. Petit, A.; Ducassou, S.; Leblanc, T.; Pasquet, M.; Rousseau, A.; Ragu, C.; Cachanado, M.; Nelken, B.; Bertrand, Y.; Michel, G.; et al. Maintenance Therapy With Interleukin-2 for Childhood AML: Results of ELAM02 Phase III Randomized Trial. Hemasphere 2018, 2, e159. [Google Scholar] [CrossRef]
  18. Rasche, M.; Zimmermann, M.; Borschel, L.; Bourquin, J.-P.; Dworzak, M.; Klingebiel, T.; Lehrnbecher, T.; Creutzig, U.; Klusmann, J.-H.; Reinhardt, D. Successes and challenges in the treatment of pediatric acute myeloid leukemia: A retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia 2018, 32, 2167–2177. [Google Scholar] [CrossRef]
  19. Creutzig, U.; Kutny, M.A.; Barr, R.; Schlenk, R.F.; Ribeiro, R.C. Acute myelogenous leukemia in adolescents and young adults. Pediatr. Blood Cancer 2018, 65, e27089. [Google Scholar] [CrossRef]
  20. Aplenc, R.; Alonzo, T.A.; Gerbing, R.B.; Smith, F.O.; Meshinchi, S.; Ross, J.A.; Perentesis, J.; Woods, W.G.; Lange, B.J.; Davies, S.M. Ethnicity and survival in childhood acute myeloid leukemia: A report from the Children’s Oncology Group. Blood 2006, 108, 74–80. [Google Scholar] [CrossRef]
  21. Liu, T.; Rao, J.; Hu, W.; Cui, B.; Cai, J.; Liu, Y.; Sun, H.; Chen, X.; Tang, Y.; Chen, J.; et al. Distinct genomic landscape of Chinese pediatric acute myeloid leukemia impacts clinical risk classification. Nat. Commun. 2022, 13, 1640. [Google Scholar] [CrossRef] [PubMed]
  22. Shiba, N.; Yoshida, K.; Hara, Y.; Yamato, G.; Shiraishi, Y.; Matsuo, H.; Okuno, Y.; Chiba, K.; Tanaka, H.; Kaburagi, T.; et al. Transcriptome analysis offers a comprehensive illustration of the genetic background of pediatric acute myeloid leukemia. Blood Adv. 2019, 3, 3157–3169. [Google Scholar] [CrossRef] [PubMed]
  23. Harrison, C.J.; Hills, R.K.; Moorman, A.V.; Grimwade, D.J.; Hann, I.; Webb, D.K.; Wheatley, K.; de Graaf, S.S.; Berg, E.v.D.; Burnett, A.K.; et al. Cytogenetics of childhood acute myeloid leukemia: United Kingdom Medical Research Council Treatment trials AML 10 and 12. J. Clin. Oncol. 2010, 28, 2674–2681. [Google Scholar] [CrossRef]
  24. Balgobind, B.V.; Raimondi, S.C.; Harbott, J.; Zimmermann, M.; Alonzo, T.A.; Auvrignon, A.; Beverloo, H.B.; Chang, M.; Creutzig, U.; Dworzak, M.N.; et al. Novel prognostic subgroups in childhood 11q23/MLL-rearranged acute myeloid leukemia: Results of an international retrospective study. Blood 2009, 114, 2489–2496. [Google Scholar] [CrossRef] [PubMed]
  25. Meyer, C.; Larghero, P.; Lopes, B.A.; Burmeister, T.; Gröger, D.; Sutton, R.; Venn, N.C.; Cazzaniga, G.; Abascal, L.C.; Tsaur, G.; et al. The KMT2A recombinome of acute leukemias in 2023. Leukemia 2023, 37, 988–1005. [Google Scholar] [CrossRef]
  26. Coenen, E.A.; Zwaan, C.M.; Reinhardt, D.; Harrison, C.J.; Haas, O.A.; de Haas, V.; Mihál, V.; De Moerloose, B.; Jeison, M.; Rubnitz, J.E.; et al. Pediatric acute myeloid leukemia with t(8;16)(p11;p13), a distinct clinical and biological entity: A collaborative study by the International-Berlin-Frankfurt-Munster AML-study group. Blood 2013, 122, 2704–2713. [Google Scholar] [CrossRef]
  27. Sandahl, J.D.; Coenen, E.A.; Forestier, E.; Harbott, J.; Johansson, B.; Kerndrup, G.; Adachi, S.; Auvrignon, A.; Beverloo, H.B.; Cayuela, J.-M.; et al. t(6;9)(p22;q34)/DEK-NUP214-rearranged pediatric myeloid leukemia: An international study of 62 patients. Haematologica 2014, 99, 865–872. [Google Scholar] [CrossRef]
  28. Klein, K.; Kaspers, G.; Harrison, C.J.; Beverloo, H.B.; Reedijk, A.; Bongers, M.; Cloos, J.; Pession, A.; Reinhardt, D.; Zimmerman, M.; et al. Clinical Impact of Additional Cytogenetic Aberrations, cKIT and RAS Mutations, and Treatment Elements in Pediatric t(8;21)-AML: Results From an International Retrospective Study by the International Berlin-Frankfurt-Munster Study Group. J. Clin. Oncol. 2015, 33, 4247–4258. [Google Scholar] [CrossRef]
  29. Noort, S.; Zimmermann, M.; Reinhardt, D.; Cuccuini, W.; Pigazzi, M.; Smith, J.; Ries, R.E.; Alonzo, T.A.; Hirsch, B.; Tomizawa, D.; et al. Prognostic impact of t(16;21)(p11;q22) and t(16;21)(q24;q22) in pediatric AML: A retrospective study by the I-BFM Study Group. Blood 2018, 132, 1584–1592. [Google Scholar] [CrossRef]
  30. Hammer, A.S.B.; Juul-Dam, K.L.; Sandahl, J.D.; Abrahamsson, J.; Czogala, M.; Delabesse, E.; Haltrich, I.; Jahnukainen, K.; Kolb, E.A.; Kovács, G.T.; et al. Hypodiploidy has unfavorable impact on survival in pediatric acute myeloid leukemia: An I-BFM Study Group collaboration. Blood Adv. 2023, 7, 1045–1055. [Google Scholar] [CrossRef]
  31. Gruber, T.A.; Gedman, A.L.; Zhang, J.; Koss, C.S.; Marada, S.; Ta, H.Q.; Chen, S.-C.; Su, X.; Ogden, S.K.; Dang, J.; et al. An Inv(16)(p13.3q24.3)-encoded CBFA2T3-GLIS2 fusion protein defines an aggressive subtype of pediatric acute megakaryoblastic leukemia. Cancer Cell 2012, 22, 683–697. [Google Scholar] [CrossRef] [PubMed]
  32. de Rooij, J.D.; Hollink, I.H.; Arentsen-Peters, S.T.C.J.M.; Van Galen, J.F.; Berna Beverloo, H.; Baruchel, A.; Trka, J.; Reinhardt, D.; Sonneveld, E.; Zimmermann, M.; et al. NUP98/JARID1A is a novel recurrent abnormality in pediatric acute megakaryoblastic leukemia with a distinct HOX gene expression pattern. Leukemia 2013, 27, 2280–2288. [Google Scholar] [CrossRef] [PubMed]
  33. de Rooij, J.D.; Branstetter, C.; Ma, J.; Li, Y.; Walsh, M.P.; Cheng, J.; Obulkasim, A.; Dang, J.; Easton, J.; Verboon, L.J.; et al. Pediatric non-Down syndrome acute megakaryoblastic leukemia is characterized by distinct genomic subsets with varying outcomes. Nat. Genet. 2017, 49, 451–456. [Google Scholar] [CrossRef] [PubMed]
  34. Meshinchi, S.; Alonzo, T.A.; Stirewalt, D.L.; Zwaan, M.; Zimmerman, M.; Reinhardt, D.; Kaspers, G.J.L.; Heerema, N.A.; Gerbing, R.; Lange, B.J.; et al. Clinical implications of FLT3 mutations in pediatric AML. Blood 2006, 108, 3654–3661. [Google Scholar] [CrossRef] [PubMed]
  35. Hollink, I.H.I.M.; Zwaan, C.M.; Zimmermann, M.; Arentsen-Peters, T.C.J.M.; Pieters, R.; Cloos, J.; Kaspers, G.J.L.; de Graaf, S.S.N.; Harbott, J.; Creutzig, U.; et al. Favorable prognostic impact of NPM1 gene mutations in childhood acute myeloid leukemia, with emphasis on cytogenetically normal AML. Leukemia 2009, 23, 262–270. [Google Scholar] [CrossRef] [PubMed]
  36. Hollink, I.H.; van den Heuvel-Eibrink, M.M.; Arentsen-Peters, S.T.C.J.M.; Zimmermann, M.; Peeters, J.K.; Valk, P.J.; Balgobind, B.V.; Sonneveld, E.; Kaspers, G.J.; de Bont, E.S.; et al. Characterization of CEBPA mutations and promoter hypermethylation in pediatric acute myeloid leukemia. Haematologica 2011, 96, 384–392. [Google Scholar] [CrossRef] [PubMed]
  37. Tarlock, K.; Lamble, A.J.; Wang, Y.C.; Gerbing, R.B.; Ries, R.E.; Loken, M.R.; Brodersen, L.E.; Pardo, L.; Leonti, A.; Smith, J.L.; et al. CEBPA-bZip mutations are associated with favorable prognosis in de novo AML: A report from the Children’s Oncology Group. Blood 2021, 138, 1137–1147. [Google Scholar] [CrossRef]
  38. Dohner, H.; Weisdorf, D.J.; Bloomfield, C.D. Acute Myeloid Leukemia. N. Engl. J. Med. 2015, 373, 1136–1152. [Google Scholar] [CrossRef]
  39. Damm, F.; Thol, F.; Hollink, I.; Zimmermann, M.; Reinhardt, K.; van den Heuvel-Eibrink, M.M.; Zwaan, C.M.; de Haas, V.; Creutzig, U.; Klusmann, J.-H.; et al. Prevalence and prognostic value of IDH1 and IDH2 mutations in childhood AML: A study of the AML-BFM and DCOG study groups. Leukemia 2011, 25, 1704–1710. [Google Scholar] [CrossRef]
  40. Hollink, I.H.I.M.; Feng, Q.; Danen-van Oorschot, A.A.; Arentsen-Peters, S.T.C.J.M.; Verboon, L.J.; Zhang, P.; de Haas, V.; Reinhardt, D.; Creutzig, U.; Trka, J.; et al. Low frequency of DNMT3A mutations in pediatric AML, and the identification of the OCI-AML3 cell line as an in vitro model. Leukemia 2012, 26, 371–373. [Google Scholar] [CrossRef]
  41. Döhner, 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]
  42. Cucchi, D.G.J.; Bachas, C.; Klein, K.; Huttenhuis, S.; Zwaan, C.M.; Ossenkoppele, G.J.; Janssen, J.M.W.M.; Kaspers, G.L.; Cloos, J. TP53 mutations and relevance of expression of TP53 pathway genes in paediatric acute myeloid leukaemia. Br. J. Haematol. 2020, 188, 736–739. [Google Scholar] [CrossRef]
  43. Hollink, I.H.; van den Heuvel-Eibrink, M.M.; Arentsen-Peters, S.T.; Pratcorona, M.; Abbas, S.; Kuipers, J.E.; van Galen, J.F.; Beverloo, H.B.; Sonneveld, E.; Kaspers, G.-J.J.L.; et al. NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern. Blood 2011, 118, 3645–3656. [Google Scholar] [CrossRef] [PubMed]
  44. Bertrums, E.J.; Smith, J.L.; Harmon, L.; Ries, R.E.; Wang, Y.-C.J.; Alonzo, T.A.; Menssen, A.J.; Chisholm, K.M.; Leonti, A.R.; Tarlock, K.; et al. Comprehensive molecular and clinical characterization of NUP98 fusions in pediatric acute myeloid leukemia. Haematologica 2023, 108, 2044–2058. [Google Scholar] [CrossRef] [PubMed]
  45. Umeda, M.; Ma, J.; Huang, B.J.; Hagiwara, K.; Westover, T.; Abdelhamed, S.; Barajas, J.M.; Thomas, M.E.; Walsh, M.P.; Song, G.; et al. Integrated Genomic Analysis Identifies UBTF Tandem Duplications as a Recurrent Lesion in Pediatric Acute Myeloid Leukemia. Blood Cancer Discov. 2022, 3, 194–207. [Google Scholar] [CrossRef]
  46. Wheatley, K.; Burnett, A.K.; Goldstone, A.H.; Gray, R.G.; Hann, I.M.; Harrison, C.J.; Rees, J.K.H.; Stevens, R.F.; Walker, H. A simple, robust, validated and highly predictive index for the determination of risk-directed therapy in acute myeloid leukaemia derived from the MRC AML 10 trial. United Kingdom Medical Research Council’s Adult and Childhood Leukaemia Working Parties. Br. J. Haematol. 1999, 107, 69–79. [Google Scholar] [CrossRef]
  47. Gibson, B.E.S.; Webb, D.K.H.; Howman, A.J.; De Graaf, S.S.N.; Harrison, C.J.; Wheatley, K.; the United Kingdom Childhood Leukaemia Working Group and the Dutch Childhood Oncology Group. Results of a randomized trial in children with Acute Myeloid Leukaemia: Medical research council AML12 trial. Br. J. Haematol. 2011, 155, 366–376. [Google Scholar] [CrossRef]
  48. Creutzig, U.; Zimmermann, M.; Ritter, J.; Henze, G.; Graf, N.; Löffler, H.; Schellong, G. Definition of a standard-risk group in children with AML. Br. J. Haematol. 1999, 104, 630–639. [Google Scholar] [CrossRef]
  49. Inaba, H.; Coustan-Smith, E.; Cao, X.; Pounds, S.B.; Shurtleff, S.A.; Wang, K.Y.; Raimondi, S.C.; Onciu, M.; Jacobsen, J.; Ribeiro, R.C.; et al. Comparative analysis of different approaches to measure treatment response in acute myeloid leukemia. J. Clin. Oncol. 2012, 30, 3625–3632. [Google Scholar] [CrossRef]
  50. 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 Measurable Residual Disease in Acute Myeloid Leukemia: European LeukemiaNet Working Party Consensus Document. Blood 2021, 138, 2753–2767. [Google Scholar] [CrossRef]
  51. Coustan-Smith, E.; Ribeiro, R.C.; Rubnitz, J.E.; Razzouk, B.I.; Pui, C.-H.; Pounds, S.; Andreansky, M.; Behm, F.G.; Raimondi, S.C.; Shurtleff, S.A.; et al. Clinical significance of residual disease during treatment in childhood acute myeloid leukaemia. Br. J. Haematol. 2003, 123, 243–252. [Google Scholar] [CrossRef] [PubMed]
  52. Langebrake, C.; Creutzig, U.; Dworzak, M.; Hrusak, O.; Mejstrikova, E.; Griesinger, F.; Zimmermann, M.; Reinhardt, D. Residual disease monitoring in childhood acute myeloid leukemia by multiparameter flow cytometry: The MRD-AML-BFM Study Group. J. Clin. Oncol. 2006, 24, 3686–3692. [Google Scholar] [CrossRef] [PubMed]
  53. van der Velden, V.H.J.; van der Sluijs-Geling, A.; Gibson, B.E.S.; te Marvelde, J.G.; Hoogeveen, P.G.; Hop, W.C.J.; Wheatley, K.; Bierings, M.B.; Schuurhuis, G.J.; de Graaf, S.S.N.; et al. Clinical significance of flowcytometric minimal residual disease detection in pediatric acute myeloid leukemia patients treated according to the DCOG ANLL97/MRC AML12 protocol. Leukemia 2010, 24, 1599–1606. [Google Scholar] [CrossRef] [PubMed]
  54. Tierens, A.; Bjørklund, E.; Siitonen, S.; Marquart, H.V.; -Juergensen, G.W.; Pelliniemi, T.-T.; Forestier, E.; Hasle, H.; Jahnukainen, K.; Lausen, B.; et al. Residual disease detected by flow cytometry is an independent predictor of survival in childhood acute myeloid leukaemia; results of the NOPHO-AML 2004 study. Br. J. Haematol. 2016, 174, 600–609. [Google Scholar] [CrossRef] [PubMed]
  55. Buldini, B.; Rizzati, F.; Masetti, R.; Fagioli, F.; Menna, G.; Micalizzi, C.; Putti, M.C.; Rizzari, C.; Santoro, N.; Zecca, M.; et al. Prognostic significance of flow-cytometry evaluation of minimal residual disease in children with acute myeloid leukaemia treated according to the AIEOP-AML 2002/01 study protocol. Br. J. Haematol. 2017, 177, 116–126. [Google Scholar] [CrossRef] [PubMed]
  56. Sievers, E.L.; Lange, B.J.; Alonzo, T.A.; Gerbing, R.B.; Bernstein, I.D.; Smith, F.O.; Arceci, R.J.; Woods, W.G.; Loken, M.R. Immunophenotypic evidence of leukemia after induction therapy predicts relapse: Results from a prospective Children’s Cancer Group study of 252 patients with acute myeloid leukemia. Blood 2003, 101, 3398–3406. [Google Scholar] [CrossRef]
  57. Loken, M.R.; Alonzo, T.A.; Pardo, L.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Ho, P.A.; Franklin, J.; Cooper, T.M.; Gamis, A.S.; et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: A report from Children’s Oncology Group. Blood 2012, 120, 1581–1588. [Google Scholar] [CrossRef]
  58. Brodersen, L.E.; Gerbing, R.B.; Pardo, M.L.; Alonzo, T.A.; Paine, D.; Fritschle, W.; Hsu, F.-C.; Pollard, J.A.; Aplenc, R.; Kahwash, S.B.; et al. Morphologic remission status is limited compared to ΔN flow cytometry: A Children’s Oncology Group AAML0531 report. Blood Adv. 2020, 4, 5050–5061. [Google Scholar] [CrossRef]
  59. Tomizawa, D.; Yoshida, M.; Kondo, T.; Miyamura, T.; Taga, T.; Adachi, S.; Koh, K.; Noguchi, M.; Kakuda, H.; Watanabe, K.; et al. Allogeneic hematopoietic stem cell transplantation for children and adolescents with high-risk cytogenetic AML: Distinctly poor outcomes of FUS-ERG-positive cases. Bone Marrow Transplant. 2019, 54, 393–401. [Google Scholar] [CrossRef]
  60. Kigasawa, H. Progress of treatment of acute myeloid leukemia in children. Jpn. J. Pediatr. Hematol. 2000, 14, 288–297. [Google Scholar]
  61. Tsukimoto, I.; Tawa, A.; Horibe, K.; Tabuchi, K.; Kigasawa, H.; Tsuchida, M.; Yabe, H.; Nakayama, H.; Kudo, K.; Kobayashi, R.; et al. Risk-stratified therapy and the intensive use of cytarabine improves the outcome in childhood acute myeloid leukemia: The AML99 trial from the Japanese Childhood AML Cooperative Study Group. J. Clin. Oncol. 2009, 27, 4007–4013. [Google Scholar] [CrossRef] [PubMed]
  62. Tomizawa, D.; Tsujimoto, S.-I.; Tanaka, S.; Matsubayashi, J.; Aoki, T.; Iwamoto, S.; Hasegawa, D.; Nagai, K.; Nakashima, K.; Kawaguchi, K.; et al. A phase III clinical trial evaluating efficacy and safety of minimal residual disease-based risk stratification for children with acute myeloid leukemia, incorporating a randomized study of gemtuzumab ozogamicin in combination with post-induction chemotherapy for non-low-risk patients (JPLSG-AML-20). Jpn. J. Clin. Oncol. 2022, 52, 1225–1231. [Google Scholar]
  63. Becton, D.; Dahl, G.V.; Ravindranath, Y.; Chang, M.N.; Behm, F.G.; Raimondi, S.C.; Head, D.R.; Stine, K.C.; Lacayo, N.J.; Sikic, B.I.; et al. Randomized use of cyclosporin A (CsA) to modulate P-glycoprotein in children with AML in remission: Pediatric Oncology Group Study 9421. Blood 2006, 107, 1315–1324. [Google Scholar] [CrossRef] [PubMed]
  64. Creutzig, U.; Ritter, J.; Zimmermann, M.; Reinhardt, D.; Hermann, J.; Berthold, F.; Henze, G.; Jürgens, H.; Kabisch, H.; Havers, W.; et al. Improved treatment results in high-risk pediatric acute myeloid leukemia patients after intensification with high-dose cytarabine and mitoxantrone: Results of Study Acute Myeloid Leukemia-Berlin-Frankfurt-Munster 93. J. Clin. Oncol. 2001, 19, 2705–2713. [Google Scholar] [CrossRef] [PubMed]
  65. Creutzig, U.; Zimmermann, M.; Bourquin, J.-P.; Dworzak, M.N.; von Neuhoff, C.; Sander, A.; Schrauder, A.; Teigler-Schlegel, A.; Starý, J.; Corbacioglu, S.; et al. Second induction with high-dose cytarabine and mitoxantrone: Different impact on pediatric AML patients with t(8;21) and with inv(16). Blood 2011, 118, 5409–5415. [Google Scholar] [CrossRef] [PubMed]
  66. Creutzig, U.; Ritter, J.; Zimmermann, M.; Hermann, J.; Gadner, H.; Sawatzki, D.B.; Niemeyer, C.; Schwabe, D.; Selle, B.; Boos, J.; et al. Idarubicin improves blast cell clearance during induction therapy in children with AML: Results of study AML-BFM 93. AML-BFM Study Group. Leukemia 2001, 15, 348–354. [Google Scholar] [CrossRef] [PubMed]
  67. Stevens, R.F.; Hann, I.M.; Wheatley, K.; Gray, R.G. Marked improvements in outcome with chemotherapy alone in paediatric acute myeloid leukemia: Results of the United Kingdom Medical Research Council’s 10th AML trial. MRC Childhood Leukaemia Working Party. Br. J. Haematol. 1998, 101, 130–140. [Google Scholar] [CrossRef]
  68. Getz, K.D.; Alonzo, T.A.; Sung, L.; Meshinchi, S.; Gerbing, R.B.; Raimondi, S.; Hirsch, B.; Loken, M.; Brodersen, L.E.; Kahwash, S.; et al. Cytarabine dose reduction in patients with low-risk acute myeloid leukemia: A report from the Children’s Oncology Group. Pediatr. Blood Cancer 2021, 69, e29313. [Google Scholar] [CrossRef]
  69. Hasle, H.; Abrahamsson, J.; Forestier, E.; Ha, S.-Y.; Heldrup, J.; Jahnukainen, K.; Jónsson, G.; Lausen, B.; Palle, J.; Zeller, B. Gemtuzumab ozogamicin as postconsolidation therapy does not prevent relapse in children with AML: Results from NOPHO-AML 2004. Blood 2012, 120, 978–984. [Google Scholar] [CrossRef]
  70. Perel, Y.; Auvrignon, A.; Leblanc, T.; Vannier, J.-P.; Michel, G.; Nelken, B.; Gandemer, V.; Schmitt, C.; Lamagnere, J.-P.; De Lumley, L.; et al. Impact of addition of maintenance therapy to intensive induction and consolidation chemotherapy for childhood acute myeloblastic leukemia: Results of a prospective randomized trial, LAME 89/91. Leucamie Aique Myeloide Enfant. J. Clin. Oncol. 2002, 20, 2774–2782. [Google Scholar] [CrossRef]
  71. Pollard, J.A.; Alonzo, T.A.; Gerbing, R.; Brown, P.; Fox, E.; Choi, J.; Fisher, B.; Hirsch, B.; Kahwash, S.; Getz, K.; et al. Sorafenib in Combination With Standard Chemotherapy for Children With High Allelic Ratio FLT3/ITD+ Acute Myeloid Leukemia: A Report From the Children’s Oncology Group Protocol AAML1031. J. Clin. Oncol. 2022, 40, 2023–2035. [Google Scholar] [CrossRef]
  72. Feijen, E.A.M.; Leisenring, W.M.; Stratton, K.L.; Ness, K.K.; van der Pal, H.J.H.; van Dalen, E.C.; Armstrong, G.T.; Aune, G.J.; Green, D.M.; Hudson, M.M.; et al. Derivation of Anthracycline and Anthraquinone Equivalence Ratios to Doxorubicin for Late-Onset Cardiotoxicity. JAMA Oncol. 2019, 5, 864–871. [Google Scholar] [CrossRef] [PubMed]
  73. Creutzig, U.; Dworzak, M.N.; Zimmermann, M.; Reinhardt, D.; Sramkova, L.; Bourquin, J.P.; Hasle, H.; Abrahamsson, J.; Kaspers, G.; van den Heuvel-Eibrink, M.M.; et al. Characteristics and outcome in patients with central nervous system involvement treated in European pediatric acute myeloid leukemia study groups. Pediatr. Blood Cancer 2017, 64, e26664. [Google Scholar] [CrossRef]
  74. Sauer, M.G.; Lang, P.J.; Albert, M.H.; Bader, P.; Creutzig, U.; Eyrich, M.; Greil, J.; Gruhn, B.; Holter, W.; Klingebiel, T.; et al. Hematopoietic stem cell transplantation for children with acute myeloid leukemia-results of the AML SCT-BFM 2007 trial. Leukemia 2020, 34, 613–624. [Google Scholar] [CrossRef] [PubMed]
  75. Tarlock, K.; Sulis, M.L.; Chewning, J.H.; Pollard, J.A.; Cooper, T.; Gamis, A.; Shenoy, S.; Kutny, M.; Horan, J.; Meshinchi, S.; et al. Hematopoietic Cell Transplantation in the Treatment of Pediatric Acute Myeloid Leukemia and Myelodysplastic Syndromes: Guidelines from the American Society of Transplantation and Cellular Therapy. Transplant. Cell. Ther. 2022, 28, 530–545. [Google Scholar] [CrossRef]
  76. Copelan, E.A.; Hamilton, B.K.; Avalos, B.; Ahn, K.W.; Bolwell, B.J.; Zhu, X.; Aljurf, M.; van Besien, K.; Bredeson, C.; Cahn, J.-Y.; et al. Better leukemia-free and overall survival in AML in first remission following cyclophosphamide in combination with busulfan compared with TBI. Blood 2013, 122, 3863–3870. [Google Scholar] [CrossRef] [PubMed]
  77. de Berranger, E.; Cousien, A.; Petit, A.; de Latour, R.P.; Galambrun, C.; Bertrand, Y.; Salmon, A.; Rialland, F.; Rohrlich, P.-S.; Vannier, J.-P.; et al. Impact on long-term OS of conditioning regimen in allogeneic BMT for children with AML in first CR: TBI+CY versus BU+CY: A report from the Societe Francaise de Greffe de Moelle et de Therapie Cellulaire. Bone Marrow Transplant. 2014, 49, 382–388. [Google Scholar] [CrossRef]
  78. Ishida, H.; Kato, M.; Kudo, K.; Taga, T.; Tomizawa, D.; Miyamura, T.; Goto, H.; Inagaki, J.; Koh, K.; Terui, K.; et al. Comparison of Outcomes for Pediatric Patients With Acute Myeloid Leukemia in Remission and Undergoing Allogeneic Hematopoietic Cell Transplantation With Myeloablative Conditioning Regimens Based on Either Intravenous Busulfan or Total Body Irradiation: A Report From the Japanese Society for Hematopoietic Cell Transplantation. Biol. Blood Marrow Transplant. 2015, 21, 2141–2147. [Google Scholar]
  79. Bitan, M.; He, W.; Zhang, M.-J.; Abdel-Azim, H.; Ayas, M.F.; Bielorai, B.; Carpenter, P.A.; Cairo, M.S.; Diaz, M.A.; Horan, J.T.; et al. Transplantation for children with acute myeloid leukemia: A comparison of outcomes with reduced intensity and myeloablative regimens. Blood 2014, 123, 1615–1620. [Google Scholar] [CrossRef]
  80. Ishida, H.; Adachi, S.; Hasegawa, D.; Okamoto, Y.; Goto, H.; Inagaki, J.; Inoue, M.; Koh, K.; Yabe, H.; Kawa, K.; et al. Comparison of a fludarabine and melphalan combination-based reduced toxicity conditioning with myeloablative conditioning by radiation and/or busulfan in acute myeloid leukemia in Japanese children and adolescents. Pediatr. Blood Cancer 2015, 62, 883–889. [Google Scholar] [CrossRef]
  81. Scott, B.L.; Pasquini, M.C.; Logan, B.R.; Wu, J.; Devine, S.M.; Porter, D.L.; Maziarz, R.T.; Warlick, E.D.; Fernandez, H.F.; Alyea, E.P.; et al. Myeloablative Versus Reduced-Intensity Hematopoietic Cell Transplantation for Acute Myeloid Leukemia and Myelodysplastic Syndromes. J. Clin. Oncol. 2017, 35, 1154–1161. [Google Scholar] [CrossRef] [PubMed]
  82. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Döhner, 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]
  83. Inaba, H.; Rubnitz, J.E.; Coustan-Smith, E.; Li, L.; Furmanski, B.D.; Mascara, G.P.; Heym, K.M.; Christensen, R.; Onciu, M.; Shurtleff, S.A.; et al. Phase I pharmacokinetic and pharmacodynamic study of the multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in pediatric relapsed/refractory leukemia. J. Clin. Oncol. 2011, 29, 3293–3300. [Google Scholar] [CrossRef]
  84. Perl, A.E.; Martinelli, G.; Cortes, J.E.; Neubauer, A.; Berman, E.; Paolini, S.; Montesinos, P.; Baer, M.R.; Larson, R.A.; Ustun, C.; et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N. Engl. J. Med. 2019, 381, 1728–1740. [Google Scholar] [CrossRef]
  85. Cortes, J.E.; Khaled, S.; Martinelli, G.; Perl, A.E.; Ganguly, S.; Russell, N.; Krämer, A.; Dombret, H.; Hogge, D.; Jonas, B.A.; et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): A multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 984–997. [Google Scholar] [CrossRef]
  86. Erba, H.P.; Montesinos, P.; Kim, H.J.; Patkowska, E.; Vrhovac, R.; Žák, P.; Wang, P.-N.; Mitov, T.; Hanyok, J.; Kamel, Y.M.; et al. Quizartinib plus chemotherapy in newly diagnosed patients with FLT3-internal-tandem-duplication-positive acute myeloid leukaemia (QuANTUM-First): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2023, 401, 1571–1583. [Google Scholar] [CrossRef] [PubMed]
  87. Dinardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, 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]
  88. 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] [PubMed]
  89. Karol, S.E.; Alexander, T.B.; Budhraja, A.; Pounds, S.B.; Canavera, K.; Wang, L.; Wolf, J.; Klco, J.M.; Mead, P.E.; Das Gupta, S.; et al. Venetoclax in combination with cytarabine with or without idarubicin in children with relapsed or refractory acute myeloid leukaemia: A phase 1, dose-escalation study. Lancet Oncol. 2020, 21, 551–560. [Google Scholar] [CrossRef] [PubMed]
  90. van Weelderen, R.E.; Klein, K.; Harrison, C.J.; Jiang, Y.; Abrahamsson, J.; Arad-Cohen, N.; Bart-Delabesse, E.; Buldini, B.; De Moerloose, B.; Dworzak, M.N.; et al. Measurable Residual Disease and Fusion Partner Independently Predict Survival and Relapse Risk in Childhood KMT2A-Rearranged Acute Myeloid Leukemia: A Study by the International Berlin-Frankfurt-Münster Study Group. J. Clin. Oncol. 2023, 41, 2963–2974. [Google Scholar] [CrossRef]
  91. Krivtsov, A.V.; Evans, K.; Gadrey, J.Y.; Eschle, B.K.; Hatton, C.; Uckelmann, H.J.; Ross, K.N.; Perner, F.; Olsen, S.N.; Pritchard, T.; et al. A Menin-MLL Inhibitor Induces Specific Chromatin Changes and Eradicates Disease in Models of MLL-Rearranged Leukemia. Cancer Cell 2019, 36, 660–673.e11. [Google Scholar] [CrossRef] [PubMed]
  92. Issa, G.C.; Ravandi, F.; DiNardo, C.D.; Jabbour, E.; Kantarjian, H.M.; Andreeff, M. Therapeutic implications of menin inhibition in acute leukemias. Leukemia 2021, 35, 2482–2495. [Google Scholar] [CrossRef] [PubMed]
  93. Heikamp, E.B.; Henrich, J.A.; Perner, F.; Wong, E.M.; Hatton, C.; Wen, Y.; Barwe, S.P.; Gopalakrishnapillai, A.; Xu, H.; Uckelmann, H.J.; et al. The menin-MLL1 interaction is a molecular dependency in NUP98-rearranged AML. Blood 2022, 139, 894–906. [Google Scholar] [CrossRef] [PubMed]
  94. Issa, G.C.; Aldoss, I.; DiPersio, J.; Cuglievan, B.; Stone, R.; Arellano, M.; Thirman, M.J.; Patel, M.R.; Dickens, D.S.; Shenoy, S.; et al. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature 2023, 615, 920–924. [Google Scholar] [CrossRef]
  95. Pearson, A.D.J.; Zwaan, C.M.; Kolb, E.A.; Karres, D.; Guillot, J.; Kim, S.Y.; Marshall, L.; Tasian, S.K.; Smith, M.; Cooper, T.; et al. Paediatric Strategy Forum for medicinal product development for acute myeloid leukaemia in children and adolescents: ACCELERATE in collaboration with the European Medicines Agency with participation of the Food and Drug Administration. Eur. J. Cancer 2020, 136, 116–129. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Genetic profiles of pediatric AML. Data is from the JPLSG AML-05 study. * Percentage of the patients with FLT3-ITD per total patients (e.g., patients with RUNX1::RUNX1T1 and FLT3-ITD accounts for 0.8% of all 369 cases).
Figure 1. Genetic profiles of pediatric AML. Data is from the JPLSG AML-05 study. * Percentage of the patients with FLT3-ITD per total patients (e.g., patients with RUNX1::RUNX1T1 and FLT3-ITD accounts for 0.8% of all 369 cases).
Cancers 15 04171 g001
Figure 2. Induction therapy used in pediatric AML.
Figure 2. Induction therapy used in pediatric AML.
Cancers 15 04171 g002
Table 1. Examples of risk stratification used in recent and ongoing pediatric AML studies.
Table 1. Examples of risk stratification used in recent and ongoing pediatric AML studies.
COG AAML1831MyeChild 01NOPHO-DBH AML 2012JPLSG AML-20
SRLow Risk 1 (LR1)
  • CBF-AML
    -
    MRD@EOI1 < 0.05%
    -
    No KIT exon17 mutations
    -
    No other HR factors
  • Mutated NPM1/CEBPA-bZip
    -
    MRD@EOI1 < 0.05%
    -
    No other HR factors
Low risk 2 (LR2)
  • Other than LR1 or HR
Standard risk (SR)
  • Good-risk abnormalities *
    -
    MRD@EOI2 < 0.1%
  • Intermediate-risk abnormalities **
    -
    MRD@EOI1&2 < 0.1%
Intermediate risk (IR)
  • Good-risk abnormalities *
    -
    MRD@EOI2 > 0.1%
  • Intermediate-risk abnormalities **
    -
    MRD@EOI1 > 0.1% & EOI2 < 0.1%

* Good-risk abnormalities
  • CBF-AML
  • Mutated NPM1 (no FLT3-ITD)
  • CEBPA double mutation (no FLT3-ITD)
** Intermediate-risk abnormalities
  • t(9;11): KMT2A::MLLT3
  • t(11;19): KMT2A::MLLT1
  • Non-poor risk KMT2A-r
  • Non-good/poor risk abnormalities
  • No high-risk (HR) factors
  • MRD/BM blasts@EOI2 < 5%
  • SR patients with inv(16)/t(16;16) receive a reduced number of consolidation courses
Low risk (LR)
  • CBF-AML
    -
    No FLT3-ITD
    -
    MRD@EOI1 < 0.1%
Intermediate risk (IR)
  • CBF-AML & FLT3-ITD
  • CBF-AML & MRD@EOI1 ≥ 0.1%
  • Non-CBF-AML
    -
    No high-risk abnormalities †
    -
    MRD@EOI1 < 0.1%
HR
  • FLT3-ITD allelic ratio > 0.1
    -
    No NPM1/CEBPA-bZip mutation
  • FLT3-ITD allelic ratio > 0.1
    -
    Mutated NPM1/CEBPA-bZip
    -
    MRD@EOI1 ≥ 0.05%
  • Mutated non-ITD FLT3
    -
    MRD@EOI1 ≥ 0.05%
  • RAM phenotype
  • Unfavorable abnormalities:
    -
    inv(3)/t(3;3): RPN1::MECOM
    -
    t(3;21): RUNX1::MECOM
    -
    t(3;5): NPM1::MLF1
    -
    t(6;9): DEK::NUP214
    -
    t(8;16): KAT6A::CREBBP (≥90 days old)
    -
    t(16;21)(p11;q22): FUS::ERG
    -
    inv(16)(p13q24): CBFA2T3::GLIS2
    -
    t(4;11): KMT2A::AFF1
    -
    t(6;11): KMT2A::AFDN
    -
    t(10;11): KMT2A::MLLT10
    -
    t(10;11): KMT2A::ABI1
    -
    t(11;19): KMT2A::MLLT1
    -
    11p15-r: any NUP98 fusion
    -
    12p13-r: any ETV6 fusion
    -
    12pdeletion: ETV6 loss
    -
    −5/del(5q): EGR1 loss
    -
    Monosomy 7
    -
    10p12.3-r: any MLLT10 fusion
  • No favorable/unfavorable abnormalities
    -
    MRD@EOI1 ≥ 0.05%
  • Intermediate-risk abnormalities **
    -
    MRD@EOI2 > 0.1%
  • Good-risk abnormalities *
    -
    MRD@EOC3 > 0.1%
  • Poor-risk abnormalities
    -
    inv(3)/t(3;3)/abn(3q26)
    -
    −5/del(5q)
    -
    −7
    -
    t(6;9): DEK::NUP214
    -
    t(9;22): BCR::ABL1
    -
    12p abnormalities
    -
    t(4;11): KMT2A::AFF1
    -
    t(6;11): KMT2A::AFDN
    -
    t(10;11): KMT2A::MLLT10
    -
    t(5;11): NUP98::NSD1
    -
    t(7;12): MNX1::ETV6
    -
    inv(16)(p13q24): CBFA2T3::GLIS2
    -
    FLT3-ITD (no mutated NPM1, CBF)
  • Other poor-risk categories
  • Secondary leukemia without good-risk abnormalities
  • Induction failure@EOI1
  • MRD/BM blasts@d22 of induction 1 ≥ 15%
  • MRD@EOI2 ≥ 0.1–4.9%
  • FLT3-ITD without mutated NPM1
  • Non-CR @EOI1
  • Non-CBF-AML
    -
    No high-risk abnormalities †
    -
    MRD@EOI1 ≥ 0.1%
  • Non-CBF-AML
    -
    High-risk abnormalities †

† High-risk abnormalities:
  • Monosomy 7
  • −5/del(5q)
  • inv(3)/t(3;3)
  • FLT3-ITD (no CBF)
  • t(9;22): BCR::ABL1
  • t(4;11): KMT2A::AFF1
  • t(6;11): KMT2A::AFDN
  • t(10;11): KMT2A::MLLT10
  • t(6;9): DEK::NUP214
  • t(7;11): NUP98::HOXA9
  • t(5;11): NUP98::NSD1
  • t(11;12): NUP98::KDM5A
  • inv(16)(p13q24): CBFA2T3::GLIS2
  • t(16;21)(p11;q22): FUS::ERG
  • t(7;12): MNX1::ETV6
  • t(10;11): PICALM::MLLT10
  • TBL1XR1::RARB
BM, bone marrow; CBF, core binding factor; EOI1/2, end of induction 1/2; EOC3, end of course 3; MRD, measurable residual disease. * Definition of Good-risk abnormalities in MyeChild01 study. ** Definition of Intermediate-risk abnormalities in MyeChild01 study. † Definition of High-risk abnormalities in JPLSG AML-20 study.
Table 2. Evidence for current standard therapy for pediatric AML.
Table 2. Evidence for current standard therapy for pediatric AML.
Treatment FactorsSummarySpecific Data
Induction chemotherapy
Cytarabine dosesThree randomized studies showed that there is not a clear impact of high-dose cytarabine in initial induction compared to low-dose or standard-dose cytarabine. High-dose cytarabine in the second induction may improve the outcome.POG9421 [63] (n = 560): High-dose vs. standard-dose DAT in initial induction. No difference in CR and EFS.
SJCRH AML02 [11] (n = 230): High-dose vs. low-dose ADE in initial induction. No difference in day 22 MRD, EFS, and OS.
JPLSG AML-12 [8] (n = 324): High-dose vs. low-dose ECM in initial induction. No difference in end-of-induction MRD, EFS, and OS.
Improved EFS for high-risk patients (n = 310) in AML-BFM93 by introducing HAM as a second induction [64]. Better RR, EFS, and OS with second induction HAM in t(8;21) patients (n = 78) in AML-BFM98 [65].
AnthracyclinesOverall, there is no clear evidence for the best anthracyclines of choice.MRC AML12 [47] (n = 504): MAE vs. ADE. Use of mitoxantrone showed decreased RR and improved DFS over daunorubicin use, but no difference in EFS and OS.
AML-BFM93 [66] (n = 358): AIE vs. ADE. Better day 15 bone marrow blast reduction with idarubicin compared to daunorubicin, but no difference in EFS and DFS.
AML-BFM2004 [13] (n = 521): ADxE (liposomal daunorubicin) vs. AIE (idarubicin). No difference in RR, EFS, and OS.
Addition of other cytotoxic drugsNo clear evidence of adding cytotoxic drugs to cytarabine/anthracycline induction. However, one randomized study showed the benefit of adding GO to initial induction and second consolidation courses. Clofarabine may spare the use of anthracyclines and etoposide. Some groups use fludarabine to enhance the effect of cytarabine (FLA).MRC-AML10 [67] (n = 359): DAT (6-thioguanine) vs. ADE (etoposide). No difference in CR, RR, DFS, and OS.
COG AAML1031 [10] (n = 1097): Randomization to add bortezomib to each standard chemotherapy course failed to improve EFS and OS.
COG AAML0531 [9] (n = 1022): ADE + GO (3 mg/m2) vs. ADE. Improved EFS (but not OS) and reduced RR in GO arm.
SJCRH AML08 [12] (n = 262): Clofarabine + HDAC vs. high-dose ADE. No difference in EFS and OS.
DB-AML-01 [16] (n = 112): Patients with t(8;21) or day 15 marrow blasts ≥ 5% received FLA + liposomal daunorubicin as second induction.
Post-induction chemotherapy
Number of coursesA number of chemotherapy courses range from 4 to 6 (including induction) in recently conducted pediatric AML studies. Two retrospective analyses show benefit of an additional chemotherapy course for a subset of LR patients.MRC-AML12 [47] (n = 270): 4 vs. 5 courses. No survival benefit for a 5th course of chemotherapy.
Combined analysis of COG AAML0531 and AAML1031 studies [68] (n = 923) showed higher RR and lower DFS (but not OS) in a subset of LR patients who received 4 courses compared to those who received 5 courses.
In the JPLSG AML-05 study [6] (n = 154), a reduction to 5 from 6 courses in the AML99 study (n = 89) resulted in increased RR in CBF-AML patients.
Addition of other cytotoxic drugsNo clear evidence of adding cytotoxic drugs to cytarabine/anthracycline chemotherapy. However, one randomized study showed the benefit of adding GO to initial induction and second consolidation courses. COG AAML0531 [9] (n = 1022): MA + GO (3 mg/m2) vs. MA (second consolidation course). Improved EFS (but not OS) and reduced RR in GO arm.
NOPHO-AML2004 [69] (n = 120): Addition of GO (5 mg/m2/dose on days 1 and 21) vs. no further therapy following the end of consolidation chemotherapies. No improvement in EFS and OS.
COG AAML1031 [10] (n = 1097): Randomization to add bortezomib to each standard chemotherapy course failed to improve EFS and OS.
Maintenance therapyNo clear role of maintenance therapy. Major study groups no longer use maintenance therapy.LAME89/91 [70] (n = 268): Maintenance therapy was introduced in LAME89 and randomized to receive or not receive maintenance in LAME91. No difference in EFS and OS.
Central nervous system-directed therapy
CNS-directed therapyMost groups usually include intrathecal therapy (ITT) in every chemotherapy course, but it is not evidence-based.Previous AML-BFM studies included prophylactic CNS irradiation, due to the BFM-AML87 study results that the patients without CNS irradiation showed an increase in marrow relapses (not CNS relapses) compared to irradiated patients, but stopped since 2009 [18].
Abbreviations: BFM, Berlin-Frankfurt-Münster; COG, Children’s Oncology Group; DB, Dutch-Belgian; LAME, Enfant Leucemie Aigue Myeloblastique; JPLSG, Japanese Pediatric Leukemia/Lymphoma Study Group; MRC, Medical Research Council; NOPHO, Nordic Society of Paediatric Haematology and Oncology; SJCRH, St. Jude Children’s Research Hospital; ADE, cytarabine + daunorubicin + etoposide; ADxE, cytarabine + liposomal daunorubicin + etoposide; AIE, cytarabine + idarubicin + etoposide; AM, cytarabine + mitoxantrone; CNS, central nervous system; CR, complete remission rate; CBF, core-binding-factor; DAT, daunorubicin + cytarabine + 6-thioguanine; DFS, disease-free survival rate; EFS, event-free survival rate; FLA, fludarabine + cytarabine; GO, gemtuzumab ozogamicin; HAM, high-dose cytarabine + mitoxantrone; MAE, mitoxantrone + cytarabine + etoposide; MRD, measurable residual disease; OS, overall survival rate, RR, relapse rate.
Table 3. Comparison of recently completed pediatric AML studies.
Table 3. Comparison of recently completed pediatric AML studies.
Study
(Years of Accrual)
No. of PatientsRisk Group/
Treatment Arm
Cumulative Anthracycline DosesNo. (%) of Patients Treated with CR1 HSCTEFS, %
OS, %
(Years)
References
DaunorubicinMitoxantroneIdarubicinOthers
JPLSG AML-05
(2006–2010)
443LR252046 (10)54 (3)
73 (3)
Tomizawa et al., 2013 [6]
Hasegawa et al., 2020 [7]
IR/HRIR:55/HR40IR:20/HR:10
JPLSG AML-12
(2014–2018)
359CBF SR402040 (11)63.1 (3)
80.3 (3)
Tomizawa et al., 2018 [8]
nCBF-SR/HRnCBF-SR:55
HR:40
nCBF-SR:20
HR:10
COG AAML0531
(2006–2010)
1022No HSCT30048157 (15)53.1 (3) *
69.4 (3) *
Gamis et al., 2014 [9]
HSCT300
COG AAML1031
(2011–2016)
1097LR3004885 (8)45.9 (3)
65.4 (3)
Aplenc et al., 2020 [10]
HR300
SJCRH AML02
(2002–2008)
230No HSCT30020 * or 5059 (26)63.0 (3)
71.1 (3)
Rubnitz et al., 2010 [11]
HSCT300
SJCRH AML08
(2008–2017)
262HD-ADE3003681 (31)52.9 (3) **
74.8 (3) **
Rubnitz et al., 2019 [12]
Clo + Ara-C15036
AML-BFM2004
(2004–2010)
611ADxESR:20/HR:4014DNX: 240NA55 (5)
74 (5)
Creutzig et al., 2013 [13]
AIESR:20/HR:4050
AIEOP AML2002/01
(2002–2011)
4825060141 (29)55 (8)
68 (8)
Pession et al., 2013 [14]
NOPHO AML2004
(2004–2009)
151304822 (15)57 (3)
69 (3)
Abrahamsson et al., 2011 [15]
DB-AML-01
(2010–2013)
112AM3036NA52.6 (3)
74.0 (3)
De Moerloose et al., 2019 [16]
FLA-DNX36DNX: 180
ELAM02
(2005–2011)
438SR8060AMSA: 300119 (27)57 (4)
73 (4)
Petit et al., 2018 [17]
IR/HR60AMSA: 300
Abbreviations: AIEOP, Associazione Italiana di Ematologia e Oncologia Pediatrica; BFM, Berlin-Frankfurt-Münster; COG, Children’s Oncology Group; DB, Dutch-Belgian; ELAM, Enfant Leucemie Aigue Myeloblastique; JPLSG, Japanese Pediatric Leukemia/Lymphoma Study Group; NOPHO, Nordic Society of Paediatric Haematology and Oncology; SJCRH, St. Jude Children’s Research Hospital; AM, cytarabine + mitoxantrone; AMSA, amsacrine; Ara-C, cytarabine; Clo, clofarabine; CR1, first complete remission; CBF, core-binding-factor; DNX, daunoxome; EFS, event-free survival rate; FLA-DNX, fludarabine + cytarabine + daunoxome; HR, high-risk; HSCT, hematopoietic stem cell transplantation; IR, intermediate-risk; LR, low-risk; NA, not available; OS, overall survival rate, SR, standard-risk. * GO arm. ** Clo + Ara-C arm.
Table 4. Going FLT3 inhibitor trials in children with AML.
Table 4. Going FLT3 inhibitor trials in children with AML.
Trial
(ClinicalTrials.gov Identifier)
RegimenKey EligibilityPhase
(No. Patients)
Current Status
Novartis
(NCT03591510)
Midostaurin + chemoChildren (3 mo–17 yo)
FLT3-mutated AML
Phase 2
(n = 23)
Recruiting
33 sites: US, Austria, Czechia, Germany, Greece, Italy, Poland, Russia, Slovenia, Turkey, Jordan, Japan, Korea
COG AAML1831
(NCT04293562)
Gilteritinib + chemoChildren (2 yo–21 yo)
FLT3-ITD (AR > 0.1)+ AML
FLT3-TKD + AML
Phase 3Recruiting
Astellas
(NCT04240002)
Gilteritinib + chemoChildren, AYA (6 mo–21 yo)
r/r FLT3-ITD + AML
Phase 1/2
(n = 97)
Recruiting
19 sites: US, Canada, Germany, Italy, Spain, UK
Daiichi Sankyo/ITCC/COG
(NCT03793478)
Quizartinib + chemoChildren, AYA (1 mo–21 yo)
r/r FLT3-ITD + AML
Phase 1/2
(n = 65)
Recruiting
36 sites: US, Canada, Belgium, Denmark, France, Italy, Netherlands, Spain, Sweden, UK, Israel
Abbreviations: COG, Children’s Oncology Group; ITCC, Innovative Therapy for Childhood Cancer; AR, allelic ratio; AYA, adolescents and young adults; FLT3-ITD, internal tandem duplication of FLT3 gene; FLT3-TKD, tyrosine kinase domain mutation of FLT3 gene; r/r, relapsed/refractory.
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

Tomizawa, D.; Tsujimoto, S.-I. Risk-Stratified Therapy for Pediatric Acute Myeloid Leukemia. Cancers 2023, 15, 4171. https://doi.org/10.3390/cancers15164171

AMA Style

Tomizawa D, Tsujimoto S-I. Risk-Stratified Therapy for Pediatric Acute Myeloid Leukemia. Cancers. 2023; 15(16):4171. https://doi.org/10.3390/cancers15164171

Chicago/Turabian Style

Tomizawa, Daisuke, and Shin-Ichi Tsujimoto. 2023. "Risk-Stratified Therapy for Pediatric Acute Myeloid Leukemia" Cancers 15, no. 16: 4171. https://doi.org/10.3390/cancers15164171

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