Despite major improvements in outcome over the past decades, acute myeloid leukemia (AML) remains a life-threatening malignancy in children, with current survival rates of ∼ 70%. State-of-the-art recommendations in adult AML have recently been published in this journal by Döhner et al. The primary goal of an international expert panel of the International BFM Study Group AML Committee was to set standards for the management, diagnosis, response assessment, and treatment in childhood AML. This paper aims to discuss differences between childhood and adult AML, and to highlight recommendations that are specific to children. The particular relevance of new diagnostic and prognostic molecular markers in pediatric AML is presented. The general management of pediatric AML, the management of specific pediatric AML cohorts (such as infants) or subtypes of the disease occurring in children (such as Down syndrome related AML), as well as new therapeutic approaches, and the role of supportive care are discussed.

Childhood acute myeloid leukemia (AML) is a rare and heterogeneous disease, with an incidence of 7 cases per million children younger than 15 years. In high-income countries, intensive therapy in conjunction with effective supportive care has increased survival rates to ∼ 70%. In 1990 and 2003, expert working groups made recommendations for diagnosis, outcomes, standardization of response criteria, and reporting standards for AML.1,2  Recent improvements in identifying the molecular genetics and pathogenesis of AML have been implemented in the new World Health Organization (WHO) classification of AML.3  These changes, and the definition of new diagnostic and prognostic markers and their associated targeted therapies, have prompted the update of earlier recommendations by an international group, on behalf of the European LeukemiaNet for AML in adults in 2010.4 

Despite broad overlap in the diagnostic and treatment recommendations for AML for children and adults, there are important differences in both the diagnostic criteria and disease management, which merit age-specific recommendations. The absence of published recommendations specific for pediatric AML motivated an international group of pediatric hematologists and oncologists (panel and participating groups see “Appendix”) to develop evidence- and expert opinion-based consensus recommendations for the diagnosis and management of AML in children, incorporating emerging information on the biology of the disease. The scope of the review is presented in the “Appendix.” Recommendations for specific subgroups are also included. This article discusses diagnostic procedures and initial workup, prognostic factors, response criteria, and management, and in particular focuses on differences between adults and children with AML.

The recent WHO 2008 classification is applicable to both adult and pediatric AML3,5  and has been summarized by Döhner et al.4  The classification contains most, but not all, cytogenetic subgroups specific to children. Differences in genetic background between children and adults are given in Table 1 and discussed further in “Cytogenetics.”

Table 1

Comparison of biologic properties and genetic abnormalities in pediatric (children and adolescents < 18 years of age) and adult (age < 60 years) AML

Adult AML
Pediatric AML
Frequency/special featuresPrognosis 5-y survival (age 18-60 y)Frequency/special features/occurrencesPrognosis 5-y survival (age < 18 y)
Epidemiology 12 (20-39 y) to 30 (40-59 y) — 2 (< 1 y) — 
Incidence rate (age-adjusted/100 person-years)206  0.7 (5-19 y)  
Biology 
    De novo AML 83% 30%-40% > 95% 60%-75% 
    Secondary (MDS-related) AML207,208  17% Adverse 1% Adverse 
    Abnormal karyotypes 55% Depends on subgroups 70%-80% Depends on subgroups 
Adult AML
Pediatric AML
Frequency/special featuresPrognosis 5-y survival (age 18-60 y)Frequency/special features/occurrencesPrognosis 5-y survival (age < 18 y)
Epidemiology 12 (20-39 y) to 30 (40-59 y) — 2 (< 1 y) — 
Incidence rate (age-adjusted/100 person-years)206  0.7 (5-19 y)  
Biology 
    De novo AML 83% 30%-40% > 95% 60%-75% 
    Secondary (MDS-related) AML207,208  17% Adverse 1% Adverse 
    Abnormal karyotypes 55% Depends on subgroups 70%-80% Depends on subgroups 
WHO classification
AML with recurrent genetic abnormalities14,15,209–211  
    t(8;21)(q22;q22)/RUNX1-RUNX1T1 8% Favorable 12%-14% Favorable 
    inv(16)(p13.1q22)/CBFB-MYH11 5% Favorable 8% Favorable 
    t(15;17)(q22;q21)/PML-RARA 5%-10% Favorable 6%-10% Favorable 
    t(9;11)(p22;q23)/MLLT3-MLL 2% 50% 7% Intermediate or favorable (63%-77%) 
    t(10;11)(p12;q23)/MLLT10-MLL 1% Unclear 3%, mainly infants Adverse 
    AML with t(6;9)(p23;q34)/DEK-NUP214 1% Adverse < 2% Adverse 
    AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2)/RPN1-EVI1 1% Adverse < 1% Adverse 
    AML (megakaryoblastic) with t(1;22)(p13;q13)/RBM15-MKL1 < 1% Unclear AMKL only; infants Intermediate 
    Provisional entity: AML with mutated NPM133,212,213  35% (53% in CN) Favorable in case of FLT3-ITD negative 5%-10% (14%-22% in CN), type A mutation dominant, increasing by age Favorable 
    Provisional entity: AML with mutated CEBPA212  10% in CN Favorable 5% (14% in CN) Favorable 
    FLT3-ITD27,30,213–215  20%-40% (∼ 50% in CN) Adverse 10% (18% in CN) Context dependent* 
WHO classification
AML with recurrent genetic abnormalities14,15,209–211  
    t(8;21)(q22;q22)/RUNX1-RUNX1T1 8% Favorable 12%-14% Favorable 
    inv(16)(p13.1q22)/CBFB-MYH11 5% Favorable 8% Favorable 
    t(15;17)(q22;q21)/PML-RARA 5%-10% Favorable 6%-10% Favorable 
    t(9;11)(p22;q23)/MLLT3-MLL 2% 50% 7% Intermediate or favorable (63%-77%) 
    t(10;11)(p12;q23)/MLLT10-MLL 1% Unclear 3%, mainly infants Adverse 
    AML with t(6;9)(p23;q34)/DEK-NUP214 1% Adverse < 2% Adverse 
    AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2)/RPN1-EVI1 1% Adverse < 1% Adverse 
    AML (megakaryoblastic) with t(1;22)(p13;q13)/RBM15-MKL1 < 1% Unclear AMKL only; infants Intermediate 
    Provisional entity: AML with mutated NPM133,212,213  35% (53% in CN) Favorable in case of FLT3-ITD negative 5%-10% (14%-22% in CN), type A mutation dominant, increasing by age Favorable 
    Provisional entity: AML with mutated CEBPA212  10% in CN Favorable 5% (14% in CN) Favorable 
    FLT3-ITD27,30,213–215  20%-40% (∼ 50% in CN) Adverse 10% (18% in CN) Context dependent* 
Not included in WHO 2008
Adverse karyotypes/mutations 32%  5%-6%  
    t(7;12)(q36;p13)/t(7;12)(q32;p13) Not in adults — Infants Adverse 
    t(5;11)(q35;p15.5)/NUP98/NSD122  Not in adults — Mostly in CN Adverse 
New mutations 
    N-RAS35,40,216  10% No prognostic significance 20% (3% in CN) No prognostic significance 
    MLL-PTD45  3%-5% Adverse 3% Not yet defined 
    c-KIT40,43,87,217  17% in CBF leukemia Adverse in t(8;21) 25% in t(8;21) Unclear 
    WT131,36,214  10% in CN Adverse combined with FLT3-ITD 13% in CN Adverse combined with FLT3-ITD 
    PTPN1142  Not in adults — 5%-21%, infants only Unclear 
    IDH1/251  16% Context-dependent mutation Rare, 2%-3% No prognostic significance 
    TET2218  8%-17% Unclear Very rare Unclear 
    DNMT3A219  20% Unclear Rare Unclear 
Not included in WHO 2008
Adverse karyotypes/mutations 32%  5%-6%  
    t(7;12)(q36;p13)/t(7;12)(q32;p13) Not in adults — Infants Adverse 
    t(5;11)(q35;p15.5)/NUP98/NSD122  Not in adults — Mostly in CN Adverse 
New mutations 
    N-RAS35,40,216  10% No prognostic significance 20% (3% in CN) No prognostic significance 
    MLL-PTD45  3%-5% Adverse 3% Not yet defined 
    c-KIT40,43,87,217  17% in CBF leukemia Adverse in t(8;21) 25% in t(8;21) Unclear 
    WT131,36,214  10% in CN Adverse combined with FLT3-ITD 13% in CN Adverse combined with FLT3-ITD 
    PTPN1142  Not in adults — 5%-21%, infants only Unclear 
    IDH1/251  16% Context-dependent mutation Rare, 2%-3% No prognostic significance 
    TET2218  8%-17% Unclear Very rare Unclear 
    DNMT3A219  20% Unclear Rare Unclear 
WHO classification continued
AML with myelodysplasia-related changes220,221  48% Unfavorable Low No prognostic significance 
Therapy-related myeloid neoplasms213  6% Adverse 3.5% Adverse 
Acute myeloid leukemia, not otherwise specified (NOS)213,222;27  ∼ 17% Intermediate ∼ 15% Intermediate 
Myeloid sarcoma (syn.: extramedullary myeloid tumor; granulocytic sarcoma; chloroma) 1% — 2%-4%, leukemia cutis (infants), chloroma Favorable in case of favorable karyotypes 
Myeloid proliferations related to DS 
    Transient abnormal myelopoiesis (syn.: transient myeloproliferative disorder)223  Not in adults — In 5% of the newborns with DS Favorable 
    Myeloid leukemia associated with Down syndrome (ML-DS) Not in adults — 400-fold increased risk for AMKL 90% 
Blastic plasmacytic dendritic cell neoplasia Rare Adverse Not seen — 
    Acute leukemias of ambiguous lineage    — 
    MPAL134  Rare Adverse 4.5% 65% 
Therapy 
    Percentage in clinical trials < 40% — 50%-96% — 
    HSCT in first CR ∼ 60% — 13%-30% — 
    CNS Not analyzed — Intrathecal prophylaxis — 
WHO classification continued
AML with myelodysplasia-related changes220,221  48% Unfavorable Low No prognostic significance 
Therapy-related myeloid neoplasms213  6% Adverse 3.5% Adverse 
Acute myeloid leukemia, not otherwise specified (NOS)213,222;27  ∼ 17% Intermediate ∼ 15% Intermediate 
Myeloid sarcoma (syn.: extramedullary myeloid tumor; granulocytic sarcoma; chloroma) 1% — 2%-4%, leukemia cutis (infants), chloroma Favorable in case of favorable karyotypes 
Myeloid proliferations related to DS 
    Transient abnormal myelopoiesis (syn.: transient myeloproliferative disorder)223  Not in adults — In 5% of the newborns with DS Favorable 
    Myeloid leukemia associated with Down syndrome (ML-DS) Not in adults — 400-fold increased risk for AMKL 90% 
Blastic plasmacytic dendritic cell neoplasia Rare Adverse Not seen — 
    Acute leukemias of ambiguous lineage    — 
    MPAL134  Rare Adverse 4.5% 65% 
Therapy 
    Percentage in clinical trials < 40% — 50%-96% — 
    HSCT in first CR ∼ 60% — 13%-30% — 
    CNS Not analyzed — Intrathecal prophylaxis — 

Favorable, intermediate, and adverse were defined according to the definitions given14,15,224 : favorable indicates 5-year survival > 60% in adults and > 70% in children; intermediate, 23%-60% in adults and 50%-70% in children; and adverse, < 23% and < 50%, respectively, in children and adults.

— indicates not applicable; and AMKL, acute megakaryoblastic leukemia.

*

Different prognosis in combination with different other mutations.

Compared with previous classifications (European Group of Immunologic Characterization of Leukemias [EGIL], WHO 2001),6  the new WHO classification introduced a stringently defined subclass of acute leukemias of ambiguous lineage (mixed phenotype acute leukemias [MPALs]), mainly on the basis of detailed immunophenotypic criteria (Table 2) or presence of t(9;22)(q34;q11.2)/BCR-ABL1 or t(v;11q23)/MLL rearrangement.3,5,6  The new classification aims to create uniform subgroups defined by unifying molecular targets, which allow selection of specific treatment.

Table 2

Immunophenotyping panel for diagnosis of AML and mixed phenotype AML in children

Diagnosis of AML*  
    Precursor stage CD34, CD117, CD133, HLA-DR 
    Myelomonocytic markers CD4, CD11b, CD11c, CD13, CD14, CD15, CD33, CD36, CD64, CD65, CD184 (CXCR4), intracellular myeloperoxidase (iMPO), i-lysozyme 
    Megakaryocytic markers CD41, CD42, CD61 
    Erythroid marker CD235a 
    Leukemia specific antigen NG2 homolog 
    Lineage aberrant antigens CD2, CD7, CD19, CD56 
    Pan-leukocyte markers CD11a, CD45 
Diagnosis of MPAL§  
    Myeloid lineage iMPO or evidence of monocytic differentiation by at least two of i-lysozyme, CD11c, CD14, CD64 
    B-lineage CD19 (strong) with at least 1 of iCD79a, iCD22, CD10; or CD19 (weak) with at least 2 of iCD79a, iCD22, CD10 
    T-lineage iCD3 or surface CD3 
Diagnosis of AML*  
    Precursor stage CD34, CD117, CD133, HLA-DR 
    Myelomonocytic markers CD4, CD11b, CD11c, CD13, CD14, CD15, CD33, CD36, CD64, CD65, CD184 (CXCR4), intracellular myeloperoxidase (iMPO), i-lysozyme 
    Megakaryocytic markers CD41, CD42, CD61 
    Erythroid marker CD235a 
    Leukemia specific antigen NG2 homolog 
    Lineage aberrant antigens CD2, CD7, CD19, CD56 
    Pan-leukocyte markers CD11a, CD45 
Diagnosis of MPAL§  
    Myeloid lineage iMPO or evidence of monocytic differentiation by at least two of i-lysozyme, CD11c, CD14, CD64 
    B-lineage CD19 (strong) with at least 1 of iCD79a, iCD22, CD10; or CD19 (weak) with at least 2 of iCD79a, iCD22, CD10 
    T-lineage iCD3 or surface CD3 
*

The table provides a list of selected markers (bold type represents the mandatory minimal panel required to fulfill WHO and EGIL criteria3,6 ), differing from the adult panel reported by Döhner et al4,214  in only a few markers (italics), which may have diagnostic or therapeutic implications.

Intracellular expression is represented by prefix “i.”

Most cases with rearranged MLL gene express the NG2 homolog reacting with the monoclonal antibody 7.1.

§

The requirements for assigning MPAL are based on the WHO 2008 criteria.3  Note that MPAL can also be diagnosed if there are 2 or more blast populations from different lineages.

The minimal diagnostic requirements in childhood AML are morphology with cytochemistry, immunophenotyping, karyotyping, FISH, and specific molecular genetics in the bone marrow, which is comparable with common practice in adults. The initial diagnostic tests may be done on peripheral blood if the patient's condition contraindicates a bone marrow aspirate.

However, investigation of CNS involvement at diagnosis is not practiced routinely in adults but is considered necessary in children because specific treatment is required in case of CNS involvement (see “CNS-directed therapy”). However, in patients with a bleeding tendency, such as those with severe thrombocytopenia and/or coagulopathy and patients with acute promyelocytic leukemia (APL), lumbar puncture should be postponed until the risk of hemorrhage has passed. CNS positivity is generally defined by > 5 × 106/L white blood cells (WBCs) in the cerebrospinal fluid, with blasts present in a nonbloody tap.

Table 3 lists the recommended procedures and diagnostic tests in the initial workup of pediatric patients with AML. In the presence of high-risk conditions or premorbid conditions, additional tests may be required.

Table 3

Recommended tests and procedures in the initial workup of pediatric patients with AML

Test/procedureRationale
Tests to establish the diagnosis  
    Complete blood counts and differential count Diagnosis, initial risk 
    Bone marrow aspirate Diagnosis 
    Bone marrow trephine biopsy* Diagnosis 
    Lumbar puncture Diagnosis, treatment, prognosis 
    Immunophenotyping Diagnosis 
    Cytogenetics Diagnosis, treatment, prognosis 
    Molecular genetics/translocations/mutations (see Table 4Diagnosis, treatment, prognosis 
Additional tests/procedures at diagnosis  
    Demographics and medical history Initial risk 
    Performance status (WHO score) Initial risk 
    Physical examination Diagnosis, side effects 
    Syndromes, comorbidities Treatment, prognosis 
    Biochemistry, coagulation tests§ Initial risk 
    Serum pregnancy test Treatment 
    Hepatitis A, B, C; CMV, EBV, VZV, blood group Rh Diagnosis, side effects 
    Chest x-ray, 12-lead, electrocardiography (ECG); echocardiography Diagnosis, side effects 
Test/procedureRationale
Tests to establish the diagnosis  
    Complete blood counts and differential count Diagnosis, initial risk 
    Bone marrow aspirate Diagnosis 
    Bone marrow trephine biopsy* Diagnosis 
    Lumbar puncture Diagnosis, treatment, prognosis 
    Immunophenotyping Diagnosis 
    Cytogenetics Diagnosis, treatment, prognosis 
    Molecular genetics/translocations/mutations (see Table 4Diagnosis, treatment, prognosis 
Additional tests/procedures at diagnosis  
    Demographics and medical history Initial risk 
    Performance status (WHO score) Initial risk 
    Physical examination Diagnosis, side effects 
    Syndromes, comorbidities Treatment, prognosis 
    Biochemistry, coagulation tests§ Initial risk 
    Serum pregnancy test Treatment 
    Hepatitis A, B, C; CMV, EBV, VZV, blood group Rh Diagnosis, side effects 
    Chest x-ray, 12-lead, electrocardiography (ECG); echocardiography Diagnosis, side effects 

Table 3 is analogous to the table of Döhner et al.4 

*

Mandatory in patients with a dry tap (punctio sicca) and in case of dysplasia in the peripheral blood or bone marrow, or in the differentiation from MDS.

Required in all patients. Patients with clinical symptoms of suspected CNS involvement should be evaluated by imaging study for intracranial bleeding, leptomeningeal disease, and mass lesion; lumbar puncture should be postponed or is considered optional in other settings (eg, high WBC count or APL).

Includes race or ethnicity, family history, prior exposure to toxic agents, prior malignancy, therapy for prior malignancy, and information on smoking.

§

Biochemistry: sodium, potassium, calcium, phosphate, creatinine, aspartate aminotransferase (AST), alanine aminotransferase (ALT), lactate dehydrogenase, bilirubin, and uric acid; on indication, add other tests. Coagulation tests: prothrombin time (PTT), international normalized ratio (INR) where indicated, and activated partial thromboplastin time (aPTT).

In adolescents (female) of child-bearing potential.

Additional tests may be considered (eg, HIV-1, Parvo B19, and HLA typing).

Recommendations

At diagnosis, morphology, cytochemistry, immunophenotyping, and molecular and cytogenetics of the bone marrow aspirate are required. In the event of a dry tap or of a suspected underlying myelodysplastic syndrome (MDS), a bone marrow trephine biopsy has to be performed as well. All patients should have a cerebrospinal fluid examination for evidence of CNS involvement.

Morphology

The morphologic classification of AML is based on the lineage-associated phenotype (undifferentiated, myeloid, monoblastic, erythroblastic, or megakaryoblastic) and defined according to the French-American-British (FAB) classification.7  Morphologic studies reveal the percentages of undifferentiated, granulated or atypical blasts, intracellular structures, such as Auer rods, and presence of myelodysplasia. Cytochemistry confirms lineage affiliation and classifies myeloid (myeloperoxidase [MPO]-positive) and monoblastic differentiation (nonspecific esterase-positive). In the presence of ambiguous morphology and cytochemistry, immunophenotyping may further support the lineage definition. Acute megakaryoblastic leukemia (AMKL, FAB M7) and minimally differentiated AML (FAB M0) have to be confirmed by immunophenotyping, although the former may show typical morphologic features.7  The presence of myelofibrosis frequently associated with acute megakaryoblastic leukemia, and consequent sampling problems may lead to an underestimation of blasts by both morphology and immunophenotyping. In case of a low blast count (< 20%), repeated bone marrow sampling, including biopsy, needs to be done.

Differentiation between AML and MDS.

Differentiating between AML and advanced MDS may be difficult in children with a low percentage of blasts. In these patients, a trephine biopsy is necessary. Differentiating between AML and MDS is important for treatment allocation, as MDS can only be cured by hematopoietic stem cell transplantation (HSCT). In adults, a blast threshold of 20% is used to differentiate between these diseases, but in children blast percentages between 20% and 30% may be seen in MDS (refractory anemia with excess of blasts in transformation).8  AML-specific genetics, hyperleukocytosis, extramedullary disease, and progression within a short time frame (2-4 weeks) are supportive of AML rather than MDS. Children with Down syndrome (DS) should be diagnosed with AML in case of evidence for leukemic blasts in the bone marrow, even if the blast threshold of 20% is not reached. This is not considered MDS but myeloid leukemia of Down syndrome (ML-DS). The same exception is valid for AML with low blast counts and recurrent genetic abnormalities t(15;17), t(8;21), inv(16), or t(16;16).3 

Recommendation

To differentiate between AML with a low blast count and MDS, serial bone marrow aspirates and trephine biopsies are required, as well as detailed cytogenetic analyses.

Immunophenotyping

Immunophenotyping is a rapid method to determine lineage and distinguish between AML and acute lymphoblastic leukemia (ALL). It is required to classify the AML FAB subtypes M0 (negative MPO activity by cytochemistry, but positive by immunophenotyping for myeloid markers, such as MPO [proenzyme] and/or CD13, CD33, CD117) and FAB M7 (positive for platelet markers, such as CD41 and/or CD61). However, immunophenotyping does not usually substitute for morphologic classification according to FAB criteria and blast cell enumeration. According to the currently used WHO 2008 classification, only the markers MPO, lysozyme, CD11c, CD14, CD64, i(intracellular)CD3, CD19, iCD22, iCD79a, and CD10 are essential to assign lineage affiliations and to define otherwise not specified MPAL. The latter includes biphenotypic leukemia, bilineage leukemia with distinctly differentiated blast populations, and undifferentiated leukemia without any lineage commitment.

At present, there is no standardization of antibody panels used for immunophenotyping among the large trial groups. However, upcoming standards suggest the use of multicolor monoclonal antibody combinations that include CD45 to enable optimal gating and analysis of the blast population within the complex context of residual hematopoiesis.9  In addition, these procedures might improve flow cytometry techniques for assessing minimal residual disease (MRD) in pediatric AML.10 

Another methodologic transition concerns the interpretation of immunophenotypic expression data. Rigid cut-off points for determining marker positivity (eg, 10% or 20%) are being replaced by biologically more meaningful semiquantitative estimates of blast population appearances compared with those of normal populations.11 Table 2 summarizes the suggested antigen panel for immunophenotypic analysis during diagnosis of children with AML, which has been modified from that recommended for adults with AML.

Recommendation

The mandatory minimal panel required to fulfill WHO and EGIL criteria for AML includes CD34, CD117, CD11b, CD11c, CD13, CD14, CD15, CD33, CD64, CD65, iMPO, i-lysozyme, CD41, and CD61; and for MPAL: CD19, iCD79a, iCD22, CD10, and iCD3.

Conventional cytogenetics and FISH

Conventional cytogenetics can detect structural and numerical cytogenetic abnormalities in 70%-80% of children with AML. Certain fusion genes, products from cryptic translocations, or loss of chromosome material can only be reliably detected using FISH.

The diagnostic cytogenetic workup in children and adults is similar.4  The most frequent chromosomal abnormalities in children with AML include t(8;21)(q22;q22), inv(16)(p13.1q22) (together referred as core binding factor [CBF]-AML), t(15;17)(q22;q21)/PML-RARA, and 11q23/MLL-rearranged abnormalities (up to 25%), which together account for ∼ 50% of pediatric AML, a much higher frequency than in adults.12–15  Additional abnormalities that are more predominant in pediatric AML are, for example, t(1;22)(p13;q13)/RBM15(OTT)-MKL1(MAL).16,17  Further types are the cryptic abnormalities t(7;12)(q36;p13)/ETV6(TEL)-HLXB9(MNX1), which are strongly associated with a +19 and t(5;11)(q35;p15.5)/NUP98-NDS1, predominantly found in cytogenetically normal AML (CN-AML).18–22  However, both translocations have not yet been included in the WHO 2008 classification (Table 1).

Importantly, the 2008 WHO classification categorizes t(9;11)(p22;q23)/MLL-MLLT3(AF9) as an entity and recommends that partners in the variant MLL translocations should be identified. Frequently, 11q23/MLL rearrangements are complex and/or cryptic. For example, the t(10;11)(p12;q23)/MLL-MLLT10(AF10) or t(6;11)(q27;q23)/MLL-MLLT4(AF6) can generate the fusion via inversions, insertions, or translocations involving other chromosomes. The best method to evaluate these cases is the sequential G-banding to FISH with a MLL probe or RT-PCR, using specific primers.

Monosomy 7, monosomy 5/5q deletions, and aberrations of 12p are rare events (seen in 3%-5% of patients) that occur in nearly all subtypes of childhood AML.14,15,23  Monosomal karyotypes, which are associated with poor prognosis in adults, are extremely rare in children.24  Trisomies 8 und 21 are often associated with additional aberrations.14,15  Cytogenetic abnormalities correlate strongly with age: ∼ 50% of infants have MLL-rearranged AML, whereas CBF-AML occur typically in older children.25 

Recommendation

Routine evaluation should include the evaluation of prognostically relevant genetic aberrations by cytogenetics/FISH, including at least the following fusion genes at diagnosis: RUNX1-RUNX1T1, CBFB-MYH11, PML-RARA, and MLL rearrangements. Other rare fusion genes mentioned in Table 4 should be traced to determine adverse risk patients.

Table 4

Genetically defined prognostic groups in pediatric AML

Prognosis14,15,22,27 Genetics
Favorable t(8;21)(q22;q22)/RUNX1-RUNX1T1 
 inv(16)(p13.1q22) or t(16;16)(p13.1;q22)/CBFB-MYH11 
 t(15;17)(q22;q21)/PML-RARA* 
 Molecular (in CN-AML) 
 NPM1-mutated AML 
 CEBPA double mutation 
 t(1;11)(q21;q23)/MLL-MLLT11(AF1Q) 
 GATA1s 
Intermediate Cytogenetic abnormalities not classified as favorable or adverse§ 
Adverse −7, −5 or del(5q) 
 inv(3)(q21q26.2) or t(3;3)(q21;q26.2)/RPN1-MECOM(EVI1-MDS1-EAP) 
 t(6;9)(p23;q34)/DEK-NUP214 
 t(7;12)(q36;p13)/ETV6(TEL)-HLXB9(MNX1) 
 t(4;11)(q21;q23)/MLL-MLLT2(AF4) 
 t(6;11)(q27;q23)/MLL-MLLT4(AF6) 
 t(5;11)(q35;p15.5)/NUP98-NSD1 
 t(10;11)(p12;q23)/MLL-MLLT10(AF10) 
 complex karyotype# 
 WT1mut/FLT3-ITD** 
 t(9;22)(q34;q11.2)†† 
Prognosis14,15,22,27 Genetics
Favorable t(8;21)(q22;q22)/RUNX1-RUNX1T1 
 inv(16)(p13.1q22) or t(16;16)(p13.1;q22)/CBFB-MYH11 
 t(15;17)(q22;q21)/PML-RARA* 
 Molecular (in CN-AML) 
 NPM1-mutated AML 
 CEBPA double mutation 
 t(1;11)(q21;q23)/MLL-MLLT11(AF1Q) 
 GATA1s 
Intermediate Cytogenetic abnormalities not classified as favorable or adverse§ 
Adverse −7, −5 or del(5q) 
 inv(3)(q21q26.2) or t(3;3)(q21;q26.2)/RPN1-MECOM(EVI1-MDS1-EAP) 
 t(6;9)(p23;q34)/DEK-NUP214 
 t(7;12)(q36;p13)/ETV6(TEL)-HLXB9(MNX1) 
 t(4;11)(q21;q23)/MLL-MLLT2(AF4) 
 t(6;11)(q27;q23)/MLL-MLLT4(AF6) 
 t(5;11)(q35;p15.5)/NUP98-NSD1 
 t(10;11)(p12;q23)/MLL-MLLT10(AF10) 
 complex karyotype# 
 WT1mut/FLT3-ITD** 
 t(9;22)(q34;q11.2)†† 

Frequencies, response rates, and outcome measures should be reported by genetic group, and, if sufficient numbers are available, by specific subsets indicated.

*

t(15;17)/PML-RARA is treated separately from other AMLs.

In particular in DS patients and infants with acute megakaryoblastic leukemia, analysis of GATA1s mutations should be included. Identification of GATA1s-associated leukemia in trisomy 21 mosaicism can prevent overtreatment.

Includes all AMLs with normal karyotype, except for those included in the favorable subgroup; most of these cases are associated with poor prognosis, but they should be reported separately as they may respond differently to treatment.

§

For most abnormalities, adequate numbers have not been studied to draw firm conclusions on their prognostic significance.

Excluding recurrent genetic aberrations, as defined in the WHO 2008 classification.

Results in t(10;11)(p12;q23) are heterogeneous; therefore, intermediate prognosis may also be adequate.

#

Three or more chromosome abnormalities in the absence of one of the WHO-designated recurring translocations or inversions.

**

There are differences in the risk allocation of FLT3-ITD considering the allelic ratio.

††

t(9;22) is rare, but it is included because its poor prognostic impact is known.

Molecular genetics

Several gene mutations and aberrantly expressed genes have been recognized in pediatric AML, further contributing to the disease's heterogeneity. AML is thought to result from at least 2 classes of cooperating mutations as hypothesized by Gilliland and supported by animal models.26  These classes can be categorized as type I mutations inducing proliferation, such as abnormalities in tyrosine kinases, and type II mutations, inducing maturation arrest, comprising most of the translocations. The frequency and nonrandom associations of type I and type II mutations in pediatric AML (Figure 1; Table 1) differ from adults, highlighting the difference in AML biology over age groups.27  In CN-AML, several mutations, such as NPM1, FLT3, WT1, and biallelic CEPBA mutations, are clinically relevant and should be included in standard diagnostics.27–31  Interestingly, the frequency and distribution of the subtypes of NPM1 mutations differ between children and adults.29,32–34  Mutations in the WT1 gene are found mainly in CN-AML and are often associated with FLT3-ITD mutations.35–37  The frequency of activating mutations of tyrosine kinase receptor genes, such as FLT3 (predominantly in CN-AML, t(15;17)(q22;q21)/PML-RARA and t(5;11)(q35;p15.5)/NUP98-NSD1), increases with age.22,30,31,35  Point mutations in the activating loop domain of the FLT3 receptor (frequency 2%-8% in children) are mutually exclusive of FLT3-ITD mutations.27,30,38 

Figure 1

Pie chart illustrating the molecular and genetic aberrations as well as their nonrandom associations in pediatric AML. (A) Total group: Integrative analysis of recurrent cytogenetic aberrations (MLL-rearrangements, t(8;21), inv(16), t(15;17), t(7;12), and t(6;9), CN and other nonspecific cytogenetic subgroups and type I molecular aberrations (in FLT3-ITD, WT1, N-RAS, K-RAS, PTPN11, and c-KIT) based on 237 unselected de novo pediatric AML patients. Each sector indicates the percentage of patients harboring 1 or more of the aforementioned mutations. (B) CN group: Pie chart based on 40 cytogenetically normal de novo pediatric AML patients analyzed for the presence of type II molecular aberrations (in NUP98-NSD1, CEBPA dm, NPM1, MLL-PTD, and other molecular aberrations) and type I molecular aberrations (in FLT3-ITD, WT1, N-RAS, K-RAS, PTPN11, and c-KIT). Each sector indicates the percentage of patients harboring 1 or more of the aforementioned mutations. None indicates patients with only wild-type alleles of genes tested. Adapted from Hollink et al22  and Balgobind et al27  with permission.

Figure 1

Pie chart illustrating the molecular and genetic aberrations as well as their nonrandom associations in pediatric AML. (A) Total group: Integrative analysis of recurrent cytogenetic aberrations (MLL-rearrangements, t(8;21), inv(16), t(15;17), t(7;12), and t(6;9), CN and other nonspecific cytogenetic subgroups and type I molecular aberrations (in FLT3-ITD, WT1, N-RAS, K-RAS, PTPN11, and c-KIT) based on 237 unselected de novo pediatric AML patients. Each sector indicates the percentage of patients harboring 1 or more of the aforementioned mutations. (B) CN group: Pie chart based on 40 cytogenetically normal de novo pediatric AML patients analyzed for the presence of type II molecular aberrations (in NUP98-NSD1, CEBPA dm, NPM1, MLL-PTD, and other molecular aberrations) and type I molecular aberrations (in FLT3-ITD, WT1, N-RAS, K-RAS, PTPN11, and c-KIT). Each sector indicates the percentage of patients harboring 1 or more of the aforementioned mutations. None indicates patients with only wild-type alleles of genes tested. Adapted from Hollink et al22  and Balgobind et al27  with permission.

Close modal

Mutations in genes involved in the RAS-RAF-ERK signal transduction pathway (PTPN11, NF-1, N-RAS, K-RAS) occur in 5%-21% of children with AML, more frequently in those with CBF-AML, and in young children with MLL-rearranged AML (Figure 1).27,39–42 

C-KIT mutations occur in ∼ 25% of children with CBF-AML, but in only 5%-8% of those with other leukemia types.30,35,43,44 MLL-PTDs are rare in childhood AML.45 

In addition to mutations, aberrant expression levels of genes have recently been reported in both adults and children; however, the biologic and clinical relevance might differ. Whereas BAALC and ERG overexpression is associated with CN-AML, EVI1 expression, which in adults is associated with monosomy 7 and inv(3), rarely occurs in children but is mainly found in association with t(6;11)(q27;q23)/MLL-MLLT4(AF6) and FAB types M6/7.46,47  Recent whole-genome sequencing studies have identified that novel somatic mutations in TET2, IDH1, IDH2, and DNMT3A are highly prevalent in adult AML but not in childhood AML.27,48–52 

The detection of the different, prognostically relevant MLL fusions genes is also important; for further details concerning the impact of molecular aberrations on outcome in pediatric AML, see “Prognostic factors, Molecular genetics.”

Recommendation

Routine evaluation should include the evaluation of a prognostically relevant and potentially targetably selected set of molecular genetic markers FLT3-ITD, WT1, C-KIT, CEBPA (double mutation), NPM1, and further specific MLL-abnormalities with favorable or very poor prognosis (eg, MLL-AF1Q, AF6, AF10).

Genome-wide studies

Several gene expression profiling studies have been conducted in recent years in pediatric AML to study their diagnostic potential and to determine whether they can replace current labor-intensive diagnostic procedures that involve many different techniques. However, the value of gene expression profiling for routine diagnostics is currently limited.53 

Biobanking

Biobanking of primary AML blasts, DNA, and RNA is relevant for patient care as it allows confirmation of initial findings or adding new data if required (ie, in case of relapse). It is a prerequisite for translational research. Stored material can be used to identify and characterize new prognostic markers, validate methods such as MRD, and enable experimental research of biologic mechanism, subgroups, and leukemogenesis. To discriminate between germline and somatic genetic aberrations in pediatric AML, the panel suggests storing additional material, such as buccal swabs.

Recommendation

Biobanking of leukemic blasts, DNA, and RNA as well as germline material is advised for potentially required diagnostic procedures and translational research.

Collaborative research in childhood and adult AML has identified several prognostic factors. These factors have been variably used for risk stratification. The most relevant factors are genetic abnormalities and treatment response, with substantial differences between adult and childhood AML. In the AML-Berlin/Frankfurt/Muenster (BFM) studies, age could not be used as an independent prognostic factor in infants and adolescents, but this may not be the case for other study groups, which reflects treatment dependency.25,54  Very high blast counts at diagnosis are associated with an increased risk of early death and nonresponse, but not necessarily with disease-free survival.55 

Cytogenetics

As in adult AML, CBF-AML and t(15;17)(q22;q21) in children are highly predictive of a favorable outcome. Translocation t(1;11)(q21;q23)/MLL-MLLT11(AF1Q) is a newly described translocation associated with favorable outcome in childhood AML.56  However, prognosis of different MLL fusions is heterogeneous.56,57 

Cytogenetics indicating an adverse outcome include −7 (excluding recurrent genetic aberrations, as defined in the WHO 2008 classification3 ), t(6;11)(q27;q23)/MLL-MLLT4(AF6), t(10;11)(p12;q23)/MLL-MLLT10(AF10), t(7;12)(q36;p13)/ETV6(TEL)-HLXB9(MNX1), t(6;9) (p23;q34)/DEK-NUP214, and t(5;11)(q35;p15.5)/NUP98-NSD1 and other rare abnormalities, such as 12p (Table 4).14,15,22  Further, adverse cytogenetics described in adult AML, such as 5q−, inv(3)(q21q26.2) or t(3;3)(q21;q26.2)/RPN1-EVI1, are very rare in children.

Results of studies on complex karyotypes (> 3 chromosomal abnormalities, excluding recurrent changes) have been inconsistent, in part because of differences in definition.14,15  Intermediate-risk factors include normal and other karyotypes. However, CN-AML has been shown to be a heterogeneous disease and the clinical outcome highly dependent on the presence of additional molecular aberrations and cryptic translocations (see “Molecular genetics”). The prognostic value of rare recurrent chromosomal aberrations remains to be elucidated and can be a future risk-stratification tool for pediatric AML.

Molecular genetics

The recognition of novel molecular subgroups in pediatric AML (see “Diagnostic procedures and initial workup, Molecular genetics”; Figure 1) has led to stratified treatment by using the identified mutated genes as treatment targets or by identifying patients eligible for myeloablative therapy.35 

In CN-AML, single-gene mutations are of specific interest, especially the NPM1 and biallelic CEPBA mutations, as they are associated with favorable outcome.27–31  In contrast, a FLT3-ITD mutant to wild-type ratio (ITD allelic ratio) of > 0.4 has been associated with adverse outcome.30  Coincidentally occurring translocations, for example, cryptic translocations of t(5;11)(q35;p15.5)/NUP98-NDS1 or mutations such as WT1 or NPM1, can modify the prognostic relevance of the FLT3-ITD.22,37,58  The prognostic impact of other mutations mentioned in “Diagnostic procedures and initial workup, Molecular genetics” remains unclear.

Table 4 proposes genetically defined prognostic groups based on similar data obtained from large studies in Europe and the United States on the prognostic impact of mutations.14,15,22,27  These studies also represent the currently recommended standardized reporting for correlation of cytogenetic and molecular genetic data in pediatric AML.

Response and prognosis

Response to the first course of treatment and cytogenetics and molecular genetics are the 2 most important indicators of outcome. Both are independent prognostic factors and are usually essential elements of the risk group classification.59–61 

Most study groups evaluate treatment response morphologically in the bone marrow after the first (eg, on day 15 or day 28) and second induction courses. This may be challenging in hypoplastic bone marrows. Blast cell reduction until day 15 and treatment response after the first and second induction are strongly predictive of outcome.59–61 

Recommendation

Determination of treatment response after first and second induction courses is recommended for risk group stratification.

Monitoring of residual disease.

Residual disease can be monitored by morphology, immunophenotyping, and quantification of molecular aberrations and gene expression levels. Although some study groups have used MRD diagnostics for treatment stratification, its definitive clinical benefit is still under investigation and also probably treatment dependent.10,62  Depending on the method and the informative marker used, MRD monitoring can be applied in variable subsets of pediatric AML; however, a single approach may not meet the specific features of all patients.

Immunophenotyping.

MRD assessment by immunophenotyping can be done in up to 96% of children with AML. However, the heterogeneity of leukemia-associated immunophenotypes and frequent antigen shifts over time can limit the sensitivity and specificity of immunophenotypic detection of MRD. Whereas some groups failed to show an independent prognostic impact, other groups have used the technique for treatment intensification.10,62,63  Current technologic advances, such as ≥ 6-color flow cytometry, may overcome any limitations. However, method standardization and quality control are essential.

Fusion genes.

The high specificity and sensitivity (up to 10−5) of real-time quantitative PCR of AML fusion genes of RUNX1(AML1)-RUNX1T1(ETO), CBFB-MYH11, PML-RARA, and MLLT3(AF9)-MLL lend themselves to MRD monitoring but are applicable in only ∼ 35% of pediatric patients. The assessment of early response requires strict definition of time points because the MRD level may vary several logs in a short time frame.64  Low-level PCR positivity (< 10−4) of RUNX1(AML1)-RUNX1T1(ETO) or CBFB-MYH11 can be detected in patients in long-term remission; therefore, the evaluation of molecular relapse must consider the dynamics of increasing levels of fusion genes by quantitative PCR.65  Importantly, the kinetics of relapse differs between genetic subtypes with a median time from molecular to clinical relapse between 2 and 8 months.65  If confirmed to be of prognostic significance, increasing levels of MRD measured by fusion genes may indicate the need for early HSCT or restart of chemotherapy.

Mutation-specific MRD.

Although methods for MRD monitoring using specific mutations, such as NPM1, FLT3-ITD, or GATA1s, have been established in childhood AML, the clinical significance requires to be clarified.33,63,66  Additional candidate mutations may be WT1, C-KIT, N-/K-RAS, PTPN11, or FLT3-point mutations.

Recommendation

MRD measurement should be incorporated in the context of clinical trials and used, if appropriate, for treatment stratification.

Response assessment

The response criteria for adult AML are widely used in pediatric AML, with some minor modifications (Table 5).4 

Table 5

Response criteria in AML

CategoryDefinition
Complete remission (CR)* Bone marrow blasts < 5%; absence of blasts with Auer rods; absence of extramedullary disease; absolute neutrophil count > 1.0 × 109/L (1000/μL); platelet count > 80 × 109/L (80 000/μL); independence of red cell transfusions 
CR with incomplete recovery (CRi) All CR criteria except for residual neutropenia (< 1.0 × 109/L [1000/μL]) or thrombocytopenia (< 80 × 109/L [80 000/μL]) 
Treatment failure  
    Resistant disease (RD) Failure to achieve CR or CRi (general practice; phase 2/3 trials), or failure to achieve CR, CRi, or PR (phase 1 trials); only includes patients surviving ≥ 7 days after completion of initial treatment, with evidence of persistent leukemia by blood and/or bone marrow examination (this should be stated by bone marrow examination < day 42 and at the end of intensification courses) 
Early death 
    Death in aplasia Death occurring ≥ 7 days after completion of initial treatment while cytopenic; with an aplastic or hypoplastic bone marrow obtained within 7 days of death, without evidence of persistent leukemia (< day 42) 
    Death from indeterminate cause 
    Early bleeding/leukostasis Deaths occurring before completion of therapy, or < 7 days after its completion; or deaths occurring ≥ 7 days after completion of initial therapy with no blasts in the blood, but no bone marrow examination available (< day 42) 
Relapse Bone marrow blasts ≥ 5%; or reappearance of blasts in the blood; or development of extramedullary disease 
CategoryDefinition
Complete remission (CR)* Bone marrow blasts < 5%; absence of blasts with Auer rods; absence of extramedullary disease; absolute neutrophil count > 1.0 × 109/L (1000/μL); platelet count > 80 × 109/L (80 000/μL); independence of red cell transfusions 
CR with incomplete recovery (CRi) All CR criteria except for residual neutropenia (< 1.0 × 109/L [1000/μL]) or thrombocytopenia (< 80 × 109/L [80 000/μL]) 
Treatment failure  
    Resistant disease (RD) Failure to achieve CR or CRi (general practice; phase 2/3 trials), or failure to achieve CR, CRi, or PR (phase 1 trials); only includes patients surviving ≥ 7 days after completion of initial treatment, with evidence of persistent leukemia by blood and/or bone marrow examination (this should be stated by bone marrow examination < day 42 and at the end of intensification courses) 
Early death 
    Death in aplasia Death occurring ≥ 7 days after completion of initial treatment while cytopenic; with an aplastic or hypoplastic bone marrow obtained within 7 days of death, without evidence of persistent leukemia (< day 42) 
    Death from indeterminate cause 
    Early bleeding/leukostasis Deaths occurring before completion of therapy, or < 7 days after its completion; or deaths occurring ≥ 7 days after completion of initial therapy with no blasts in the blood, but no bone marrow examination available (< day 42) 
Relapse Bone marrow blasts ≥ 5%; or reappearance of blasts in the blood; or development of extramedullary disease 

Definitions of response criteria are based primarily on those given by Cheson et al2  and amended by Creutzig and Kaspers.225 Table 5 is analogous to the table of Döhner et al.4 

*

All criteria need to be fulfilled; marrow evaluation should be based on a count of 200 nucleated cells in an aspirate with spicules; if ambiguous, consider repeating examination after 5-7 days; flow cytometric evaluation may help to distinguish between persistent leukemia and regenerating normal marrow; a marrow biopsy should be performed in cases of dry tap, or if no spicules are obtained; no minimum duration of response required. The categories morphologic leukemia-free state, partial remission (PR), cytogenetic CR (CRc), and molecular CR (CRm) may be useful in the clinical development of novel agents in phase 1 or 2 clinical trials. For definitions, see Döhner et al.4 

The criterion of CRi is of value in protocols that use intensified induction or double-induction strategies, in which hematologic recovery is not awaited but intensive therapy will be continued. In such protocols, CR may even not be achieved in the course of the entire treatment plan. In these instances, the overall remission rate should include CR and CRi patients. Some patients may not achieve complete hematologic recovery, even with longer observation times. As treatment courses should be very condensed (with minimal delay) in pediatric patients, treatment is continued, even without complete platelet recovery. Therefore, the recommended cut-off value for platelets indicating CR is 80 × 109/L.

In patients with low blast percentages (5%-10%), a repeat marrow should be performed to confirm relapse. Appearance of new dysplastic changes should be closely monitored for emerging relapse. In a patient who has been recently treated, dysplasia or a transient increase in blasts may reflect a chemotherapy effect and recovery of hematopoiesis. Cytogenetics should be tested to distinguish true relapse from therapy-related MDS/AML.

Treatment failure

Early death can be defined more precisely using the time point of 42 days because response status is normally evaluated within 42 days. At later time points, death from nonresponse (relapse) or treatment-related mortality in remission should be distinguished as competing events.

General aspects of treatment

All children with AML should preferably be treated within the context of well-designed clinical trials to ensure highest quality and safety of diagnostics and management. AML treatment in childhood should be risk-adapted, based on biologic factors to avoid overtreatment in patients with favorable prognosis and to improve outcome in those with unfavorable prognosis (see “Prognostic factors”). The aim of therapy is to cure the child by eradicating the leukemic clone while avoiding side effects and late effects as much as possible. Intensive polychemotherapy based mainly on anthracyclines and purine analogues should be started as soon as AML is diagnosed. Hyperleukocytosis and APL should be regarded as medical emergencies because of their early hemorrhagic risk (see “APL” and “Hyperleukocytosis”).

After induction therapy, postremission treatment is necessary to maintain remission. Additional CNS-directed intrathecal therapy is routine. In general, maintenance therapy has not been proven effective in children and adults, except in patients with APL (see “APL”).4,67,68 

Several clinical trials suggest that increased intensity is associated with improved outcome in most AML subtypes, except APL, ML-DS, and inv(16). However, treatment-related toxicity, particularly cardiotoxicity, must be balanced against antileukemic efficacy to result in long-term cure (see “Induction”).

Improvements in supportive care have had a major impact on increased survival rates in childhood AML. Treatment intensity can be achieved either by high cumulative dosages of anthracyclines or purine analogues or intensively timed chemotherapy.69–71  The Medical Research Council (MRC) Study Group delivered relatively high doses of anthracyclines, whereas the Nordic Society of Pediatric Hematology and Oncology (NOPHO) Group, the Japanese Childhood AML Cooperative Study Group (JPLSG), and the St Jude AML Study Group have focused more on high-dose cytarabine (or other antimetabolites, such as cladribine). The AML-BFM study group has used both drugs in relatively high doses. Despite these different approaches, survival rates have been roughly similar. The number of treatment courses used by different groups has varied between 4 and 8, again with similar outcomes. Supplemental Table 1 (available on the Blood Web site; see the Supplemental Materials link at the top of the online article) gives an overview of the results of recent pediatric AML trials.

Recommendation

Children with AML should be treated within controlled clinical trials. Treatment of childhood AML requires an intensive anthracycline- and cytarabine-based therapy using at least 4 or 5 courses.

Induction

One or 2 courses of induction therapy are routinely used in children and adults. Standard induction therapy comprises 3 days of an anthracycline (eg, daunorubicin at least 60 mg/m2, idarubicin 10-12 mg/m2, or the anthracenedione mitoxantrone 10-12 mg/m2) and 7-10 days of cytarabine (100-200 mg/m2 continuously or twice daily intravenously; ie, “3 + 7” or “3 + 10”). With these regimens, > 85% of children and adolescents achieve complete remission (CR). Although a third drug, such as etoposide or 6-thioguanine, is commonly included in induction, their benefit has not been proven.72 

Anthracyclines.

Various anthracyclines have been evaluated in prospective randomized pediatric clinical trials. There is evidence that higher doses of anthracyclines improve outcome in children and adults.73,74  However, toxicity, especially acute and late cardiotoxicity, is dose related and limits the cumulative dose.75,76  Cumulative dosages > 300 mg/m2 have been associated with significant later cardiac toxicity.77  There is no safe dose, but the antileukemic benefit of anthracyclines is such that most groups use a relatively high dose. The cumulative dose should consider host factors, such as age and sex.78  To avoid high peak serum concentrations of anthracyclines, splitting the daily dose or using prolonged drug infusions has been proposed; however, results concerning the benefit of dose scheduling are conflicting; hence, there is no consensus on the best regimen.75,79–81  Anthracyclines with a low cardiac exposure, such as liposomal anthracyclines, may be of benefit.79,81 

Given an arbitrary conversion rate of 5:1 (daunorubicin 60 mg/m2 × 3 days: idarubicin 12 mg/m2 × 3 days), a better blast cell reduction at day 15 with comparable acute toxicity was achieved in the AML-BFM 93 trial by idarubicin (however, without an overall survival benefit).82  The randomized comparison between mitoxantrone (first induction 12 mg/m2, second induction 8 mg/m2, each × 3 days) and daunorubicin (50 mg/m2 × 3 days) in the MRC12 trial revealed similar results.83  The AML-BFM 2004 trial has shown that a liposomal formulation of daunorubicin allows the daunorubicin dose to be increased (80 mg/m2 × 3 days) without increasing acute and long-term cardiotoxicity thus far.

Cardioprotection with dexrazozane was another option to reduce cardiotoxicity during anthracycline exposure.84  However, the approved indications for this drug have been revised because of a possible higher rate of secondary malignancies in pediatric cancer patients (http://www.fda.gov/Drugs/DrugSafety/ucm263729).85 

Recommendation

In childhood AML, the cumulative dose and application modus of anthracyclines must be considered because the risk of acute and long-term cardiotoxicity depends on both dosage and patient conditions.

Dosage of cytarabine.

The use of high-dose cytarabine (HiDAC) in first induction did not improve the CR rate or survival in adults or children.4,10,86  HiDAC may benefit CBF-AML patients when given in consolidation. The results of the AML-BFM 98 and 2004 trials in patients with t(8;21) AML demonstrated a significant favorable impact of HiDAC plus mitoxantrone in second induction.87 

Additional agents.

Other drugs that have been used during induction include aclarubicin, amsacrine (adults), mitoxantrone (children and adults), and 2-chlorodeoxyadenosine (children).4,88–90  It is not clear whether these agents improve early treatment response, event-free survival, or overall survival compared with daunorubicin plus cytarabine at equivalent doses. Similarly, the benefit of gemtuzumab ozogamicin (GO), a calicheamicin conjugated to a CD33 antibody, has not been defined in children with de novo AML (see “Antibody-targeted drugs”).

Recommendation

One or 2 courses of induction therapy comprising 3 days of an anthracycline and 7-10 days of cytarabine should be applied.

Postremission strategies

Consolidation/intensification.

In most pediatric studies, 2 to 5 courses of chemotherapy with non–cross-resistant drug combinations similar to those given during induction are used to consolidate and maintain remission. There is no clear evidence that more than 3 consolidation courses (after 2 induction courses) are beneficial.72,83 

High-dose cytarabine.

The Cancer and Leukemia Group B (CALGB) study in adults showed that 4 courses of HiDAC (3 g/m2 per every 12 hours on days 1, 3, and 5) were superior to 4 courses of lower-dose (100 mg/m2 continuous intravenously on days 1-5) cytarabine.91  This benefit of cytarabine dose intensification, however, was restricted to patients with CBF-AML and, to a lesser extent, to patients with CN-AML.92 

Results of several pediatric trials (NOPHO AML 93, MRC AML 10, AML-BFM 2004, and AML99 of the JPLSG) and those of the adult CALGB study show that relapse rates can be reduced by introducing intensive chemotherapy courses that include HiDAC.92–97 

HSCT.

HSCT is used as postremission consolidation therapy, and both autologous and allogeneic HSCT have been studied.

Autologous HSCT.

Several trials and a meta-analysis of pediatric AML trials have found no benefit for auto-HSCT compared with nonmyeloablative chemotherapy in first CR.95,98–101  In the absence of benefit, the toxicity associated with the conditioning regimen in auto-HSCT further negates against auto-HSCT. However, there is a role for auto-HSCT in relapsed APL without detectable MRD.102 

Allogeneic HSCT.

The benefit of allo-HSCT as postremission consolidation treatment in first CR AML in children remains controversial. The significantly lower relapse risk associated with allo-HSCT compared with postremission chemotherapy has not consistently translated into an improvement in survival.103–105  Moreover, it has been calculated that at least 10 children must be transplanted to avoid 1 relapse.105  The risk-benefit for patients relies on intensity of prior chemotherapy, risk group allocation, CR status, donor type, predicted transplant-related mortality, and long-term toxicity. There is consensus that favorable-risk patients should not be transplanted in first CR, but for intermediate- and high-risk patients results are controversial, showing either no benefit in first CR or some benefit in intermediate- and high-risk patients.103,105,106 

A meta-analysis of the comparative outcomes by risk group for HSCT and chemotherapy for 1373 children enrolled in 4 cooperative group clinical trials (Pediatric Oncology Group [POG] 8821, CCG 2891, CCG 2961, and MRC AML10) showed a benefit for allo-HSCT in intermediate-risk patients only.103  The analysis was weakened because a large number of patients could not be stratified by risk group, and this resulted in a possible selection bias. In addition, in some of the studies, the results of the chemotherapy only groups were inferior to those achieved in other pediatric trials.

In pediatric AML, conditioning regimens with myeloablative chemotherapy are preferred to total body irradiation as the latter is associated with more late effects and an increased risk of secondary malignancies.107–109  However, the optimal conditioning regimen remains to be identified and standardized.110,111 

Pursuing a strategy of recommending sibling donors for standard-risk AML and reserving alternative donors for high-risk disease is in conflict with smaller family size and fewer children with matched siblings, and recently improved outcome with HSCT using unrelated matched donors.112  Whether HSCT from an alternative donor is superior to intensive chemotherapy alone as consolidation therapy in first CR remains to be determined. It has to be mentioned that high levels of MRD before HSCT have been associated with poorer outcome.113 

In summary, the role of HSCT in children with AML in first CR should be reassessed as the field evolves, particularly within specific risk groups. There is consensus, however, that HSCT should be offered to all children with relapsed AML in second CR. In addition, many groups offer HSCT to children with high-risk or refractory disease and exploit the graft-versus-leukemia effect.114  There is agreement that favorable-risk patients should not be offered HSCT.105  The role of allo-HSCT in intermediate- and high-risk patients may be redefined by improving risk stratification, including the identification of new prognostic markers and improvements in MRD monitoring.

Recommendation

Auto-HSCT is not recommended for children with AML in first CR. Allo-HSCT in first CR is not beneficial in childhood AML with favorable risk factors. In other risk groups, the benefit of allo-HSCT must be balanced against toxicity. Allo-HSCT in second CR is generally considered.

CNS-directed therapy

CNS involvement at diagnosis and at relapse is seen in 5%-10% of pediatric patients with AML. Factors associated with CNS leukemia include hyperleukocytosis, monocytic leukemia [FAB M4 or M5, including M4eo with inv(16)], MLL gene rearrangement, and younger age.115 

CNS treatment is given to all pediatric patients, including those who do not have detectable CNS involvement, assuming that systemic therapy has limited efficacy to eradicate hidden AML blasts in the CNS compartment. CNS treatment has varied from intrathecal chemotherapy (single-agent cytarabine or methotrexate, or triple cytarabine, methotrexate, and hydrocortisone) alone or given in combination with cranial radiotherapy. Cranial irradiation seems to be effective in preventing CNS leukemia; however, the risk of late toxicities and secondary malignancies, as well as the increasing use of HiDAC, which crosses the blood-brain barrier, and the lack of evidence of superiority for cranial irradiation, led to it being abandoned by all major study groups.116  The AML-BFM 87 trial was the only prospective study testing the benefit of cranial irradiation. Results including the nonrandomized patients showed an increased risk of CNS and/or bone marrow relapses in nonirradiated patients.117  However, other studies with comparable overall outcomes do not support a benefit from CNS irradiation. Pui and Howard recently reported that changing the intrathecal regimen from triple intrathecal therapy to cytarabine alone increased the CNS relapse rate.118  Finally, the optimal number of intrathecal treatments (range 4-12) remains unknown.

Recommendation

CNS treatment needs to consist of intrathecal treatments with cytarabine or methotrexate or in combination with steroids, although the optimal number of administrations is unknown.

Additional CNS-directed therapy in patients with CNS involvement.

CNS positivity (ie, CNS3 status) at the time of AML diagnosis necessitates intensified, CNS-directed therapy. Apparently, unlike in pediatric ALL, a low number of (sporadic) cerebrospinal fluid blasts (CNS2 status) does not influence the CNS relapse risk in pediatric AML.119  In contrast to ALL, CNS positivity is not a crucial factor within the AML risk group stratification because it does not affect overall survival.120  However, those with CNS involvement (as defined in “Diagnostic procedures and initial workup”) relapse more frequently in the CNS.119  Therefore, patients with CNS involvement require intensified intrathecal therapy to clear blasts from the CNS fluid. Although most study groups have added CNS irradiation to the regimen of these patients, recent observations suggest that frequent intrathecal chemotherapy combined with intensive systemic chemotherapy may yield similar results.119,121 

In infants, cranial irradiation should be avoided or delayed by bridging or substituting with intrathecal chemotherapy. In patients with CNS involvement undergoing HSCT, post-transplantation CNS therapy should be considered.122  There is no proven superior approach for patients with CNS relapse.123 

Hematopoietic growth factors as priming agents

Sensitization of leukemic cells with hematopoietic growth factors (priming), such as G-CSF and GM-CSF, has been studied predominantly in adults with the aim of increasing cytotoxicity of chemotherapy.4  However, it cannot be recommended as routine practice in either adults and children, as the data are conflicting, and recent studies raise doubt about the beneficial role of G-CSF.124–126 

Children under 2 years of age

The biology of AML in children younger than 2 years is different from that of older children. Morphologic, cytogenetic, molecular genetics, and initial response show that these patients generally have features of high-risk AML.54 MLL rearrangements are frequent (∼ 50%), and some rare aberrations t(7;12)(q36;p13)/t(7;12)(q32;p13), t(7;12)(q36;p12), and t(1;22)(p13;q13)/RBM15(OTT)-MKL1(MAL) are nearly exclusive to this age group. In contrast, CBF-AML and t(15;17) are rarely seen.

In principle, management of young children is not different from that of older children. However, immaturity of organs (lung, liver, brain) in infants younger than 1 year and differences in pharmacokinetic and pharmacodynamic profiles of certain drugs (eg, cytarabine) increase their susceptibility to toxicities. Drug dosage in infants is generally calculated according to body weight (mg/kg) rather than body surface. Pharmacokinetic analysis has shown that children younger than 2 years have a reduced cytarabine clearance, which prompted the AML-BFM study group to adjust the dose of HiDAC by age.127 

Compared with older children, acute toxicities in infants during induction chemotherapy are both more common and more severe, particularly sepsis, gut toxicity, and pulmonary toxicities.54  Acute and chronic cardiotoxicity is higher in infants than older children.78  Cranial irradiation should be avoided or postponed to avoid neurologic late effects (see “AML in Down syndrome and other genetic disorders”).

Congenital AML is rare and is curable when it occurs.54  A GATA1 mutation-associated leukemia based on an underlying constitutional (mosaicism) of trisomy 21 should be excluded because treatment is not required (see “AML in Down syndrome and other genetic disorders”). There are also rare reports of durable spontaneous remissions in newborns. Therefore, in clinically asymptomatic and stable newborns, intensive chemotherapy should be deferred until the hematologic course evolves or clinical symptoms requiring treatment develop.128 

Recommendation

Infants tolerate intensive therapy, including HSCT, but their increased susceptibility to toxicities requires complex, sophisticated therapy concepts comprising age-adapted drug dosages and high treatment expertise.

Myeloid sarcoma

Myeloid sarcoma (MS; also termed extramedullary AML, extramedullary myeloid tumor, and granulocytic sarcoma) is a rare manifestation of AML characterized by the occurrence of 1 or more myeloid tumor masses (or infiltrations) at an extramedullary site.129  MS mainly occurs concurrently with AML but sometimes presents before bone marrow involvement as the first manifestation. Usually, the skin is affected (leukemia cutis), but other organs [chloroma, eg, in orbits (associated with t(8;21)), lymph nodes, bone, soft tissue, kidneys, testes] may also be involved. The presence of < 20% malignant bone marrow blasts distinguishes MS from frank de novo AML.130 

Isolated primary MS and MS with a low blast count composes 2%-4% of all cases of childhood AML.131  MS is most frequent in young children (median age, 5 years), and its incidence decreases with age. MS can be misdiagnosed, primarily as non-Hodgkin lymphoma. Intensive AML-specific chemotherapy stratified according to general AML risk factors is recommended for all patients with MS.131  Local irradiation may be considered, but prospective studies are lacking because of the rarity of these manifestations.132 

Recommendation

MS requires systemic treatment similar to that given for de novo AML.

Mixed phenotype acute leukemia

The survival of children with MPAL is superior to that of adults.133  Several retrospective analyses show that ALL-directed therapy is very effective in patients with a dominant lymphoblastic phenotype.134,135 

Recommendation

MPAL should be treated according to the dominant lineage defined by genetics, morphology, cytochemistry, and immunophenotyping.

AML in Down Syndrome and other genetic disorders

Children with specific inborn diseases are at increased risk of developing AML, with DS being the most frequent genetic disorder associated with increased likelihood of AML.136 

Children with DS or DS mosaicism are at a 14- to 20-fold risk of developing acute leukemia. Approximately 5% of newborns with DS have transient leukemia (also referred to as transient abnormal myelopoiesis or transient myeloproliferative disorder), which usually disappears spontaneously.137  Within the first 4 years of life 10%-20% of these infants will develop myeloid leukemia with megakaryoblastic features (ML-DS). Transient leukemia and ML-DS are characterized by mutations in exon 2 of the hematologic transcription factor GATA1, which result in loss of the activation domain and leads to a truncated protein GATA1s.138,139  The leukemic blasts originate from fetal liver hematopoiesis.140  Although most newborns with DS experience spontaneous remission within 4-10 weeks, some have severe and life-threatening clinical symptoms associated with pancytopenia or organ infiltration. Hydrops fetalis is the most severe complication, and pleural effusions and ascites can be life-threatening in the unborn infant. Within the first weeks of life, some children develop fatal liver cirrhosis with hyperbilirubinemia (conjugated bilirubin > 250μmol/L).141  In patients with life-threatening symptoms resulting from hyperleukocytosis or organomegaly/organ dysfunction, intervention with exchange transfusion and/or low-dose cytarabine chemotherapy (1-1.5 mg/kg × 5-7 days) is usually applied.137,141 

ML-DS can be successfully treated with intensity-reduced chemotherapy and without HSCT, resulting in event-free survival and survival rates of > 85%.142–144  As children with DS are especially susceptible to chemotherapy and have a compromised immune system, side effects and infections are frequent and treatment-related deaths are as frequent as relapse.142,145,146 

Recommendation

Newborns with DS who have transient leukemia with severe clinical symptoms secondary to leukemic infiltrates should receive supportive care, and consideration should be given to the use of low-dose cytarabine.

ML-DS should receive reduced-intensive chemotherapy and particular attention given to the high risk of severe infections. HSCT is not indicated in first CR in ML-DS.

Patients with congenital syndromes, associated with increased chromosome fragility because of disturbed DNA-repair mechanisms (eg, Fanconi anemia and Bloom syndrome), as well as those with congenital diseases of the myelopoiesis (eg, Kostmann syndrome and Diamond-Blackfan anemia) are at a considerably increased risk of developing AML.147,148  Optimal therapy, including scheduling and intensity, has not yet been defined for these rare subgroups. In congenital neutropenia and Fanconi-anemia associated AML, less intensive therapy aimed at blast reduction followed by allo-HSCT may be successful. In Fanconi-anemia associated AML, cross-linking chemotherapy, high anthracycline dosages, and irradiation should be avoided.149 

Little is known about AML in familial cancer syndromes, such as those involving germline CEBPA mutations. The few reported cases of pediatric AML have been successfully treated with conventional chemotherapy and allo-HSCT in first CR.150 

Acute promyelocytic leukemia

APL is characterized by chromosomal rearrangements of 17q21 involving RARA, which is most commonly fused to the PML (promyelocytic leukemia) gene as a result of the t(15;17)(q22;q21) translocation. In < 5% of cases, RARA is fused to an alternative partner, causing a variable sensitivity to all-trans retinoic acid (ATRA). APL is considered a medical emergency because of the risk of life-threatening hemorrhage; therefore, treatment with ATRA should be initiated immediately.

In patients with high WBC counts (arbitrarily defined as > 10 × 109/L in children), the combined use of ATRA with chemotherapy decreases the incidence of the APL differentiation syndrome,102  which is characterized by fever, weight gain, respiratory distress, and pleural and pericardial effusions and occurs in ∼ 10% of children with APL treated with ATRA or arsenic trioxide. Pseudotumor cerebri during initial ATRA administration is common in children (11%) and can be treated with steroids.151,152 

In children, a dose of 25 mg/m2 per day ATRA appears to produce outcomes equal to the higher dose of 45 mg/m2 per day commonly used in adults with a better safety profile.152–155 

The use of ATRA in maintenance is generally recommended.102  Limited data obtained in children also support the use of ATRA in maintenance.155  Recent trials in adults suggest that subgroups of AML patients may not require (prolonged) maintenance treatment.156  This is also an important area for future clinical research in pediatric APL.

High cumulative dosages of anthracyclines combined with ATRA are very effective in treating APL.157  However, high doses of anthracyclines carry a risk of cardiotoxicity in children, and it would appear that a reduced cumulative dose of anthracyclines combined with HiDAC may be an equally successful approach.158 

An intergroup protocol for children with APL (International Consortium for childhood APL; ICC APL; www.clinicaltrials.gov; #NCT01226303) has recently been opened. Quantitative monitoring of MRD will direct treatment of molecular relapse to prevent frank relapse. Such treatment has been shown to be beneficial, although only in sequential clinical trials in adults.102,159,160 

Several studies in adults and children have demonstrated the effectiveness of ATRA combined with arsenic trioxid without conventional chemotherapy in refractory, relapsed, or newly diagnosed APL.161,162  However, more safety and efficacy data need to be generated before routinely introducing arsenic in newly diagnosed APL protocols in children.102 

Recommendations

In children, ATRA at 25 mg/m2 per day should already be started if APL is suspected, as it reduces the risk of fatal hemorrhage. It should be used throughout treatment. The intensive, risk-adapted chemotherapy regimen in APL should be based on anthracyclines, cytarabine, and ATRA to avoid excessive anthracycline exposure.

Monitoring to detect molecular relapse allows earlier treatment, which might improve outcome.

Therapy-related secondary AML (t-AML) is one of the most frequent secondary malignancies and generally follows the use of chemotherapy, including alkylating agents and topoisomerase II inhibitors, and/or radiotherapy.163 

The etiology and strong association with specific translocations involving 11q23 (MLL) and less often with 5q−/−5 and 7q−/−7 in children are similar to that seen in adults.164  As in adults, the prognosis of t-AML in children is generally poor, which is because of resistance and cumulative toxicities of previous chemotherapy.4,165  Results from small studies suggest that these patients benefit from AML-type induction courses (double induction), followed by allogeneic HSCT, if remission can be achieved.165 

Recommendations

Children with t-AML should participate in prospective AML trials that take into account prior chemotherapy. Allo-HSCT is recommended in first CR.

Approximately 5% of children with AML have refractory disease and 30% experience relapse.166  Bone marrow is the most common site of relapse, with the CNS being involved in up to 10% of cases (including combined relapses). Approximately 50% of patients have an early relapse, arbitrarily defined as within 1 year of initial diagnosis. In general, the prognosis in these children is poor.167,168 

Prognostic factors in relapsed AML

The international study Relapsed AML 2001/01 confirmed early treatment response (bone marrow evaluation shortly before the second reinduction course) as the most important prognostic factor.169  Duration of first CR and CBF-leukemia are also important prognostic factors in relapse.167,168,170,171 

Current treatment in relapsed AML

Different protocols and schedules have been used to induce a second remission, but currently antimetabolite (cytarabine, fludarabine) and anthracycline-based approaches are mainly used.171,172  With a realistic option for cure, reinduction should be offered to all children and adolescents with relapsed AML who can tolerate intensive treatment.167  Patients enrolled on the recently completed randomized international relapsed AML 2001/01 study comparing fludarabine/cytarabine/G-CSF with the addition of liposomal daunorubicin showed a second CR rate of 59% and 69%, respectively, translating to a survival rate of 38% for all patients.169 

As in first-line treatment, CNS-directed therapy with several administrations of intrathecal triple chemotherapy is usual. Allo-HSCT is recommended for all children who achieve CR, using either related or unrelated HLA-matched donors, although formal evidence is lacking. If necessary, time to transplantation may be bridged with consolidation chemotherapy. The use of haploidentical or very high-risk allo-HSCT must be balanced against the potential treatment related mortality and the possibility of survival in late relapse after treatment with chemotherapy only.173 

Patients who respond poorly to the first course of reinduction chemotherapy, who do not achieve a second CR and who relapse for the second time, can be offered more experimental therapy or palliation alone. Data on allo-HSCT in AML with blast persistence are scarce; however, survival rates are generally poor.167 

Other options include GO (see “Antibody-targeted drugs”) alone or in combination with other agents.174,175  More intensive approaches include clofarabine monotherapy or combined with cytarabine and/or liposomal daunorubicin/cyclophosphamide and have shown moderate survival rates (26% and 38%, respectively) in these heavily pretreated patients.176,177 

Recommendation

Children with relapsed AML should be treated with reinduction chemotherapy followed by allo-SCT.

The intensification of conventional chemotherapy along with improvements in supportive care has improved the prognosis in childhood AML. However, side effects and toxicity still remain a major concern. New compounds, such as epigenetically active agents, tyrosine kinase inhibitors, and antibody-mediated treatment, might be effective but less toxic approaches in AML.

Antibody-targeted drugs

Gemtuzumab ozogamicin.

Gemtuzumab ozogamicin (GO), a calicheamicin-conjugated CD33 antibody, has shown promising results in children with AML.175,178,179  GO causes considerable toxicity, especially myelosuppression with an extended thrombocytopenia, and veno-occlusive disease, but less frequently in children than in adults.175  Moreover, such toxicity is dose related and has been shown to be significantly less pronounced at GO doses of ≤ 6 mg/m2 in recent studies.180  Salvage studies demonstrated that the drug was beneficial in children who had poor chances of survival by conventional therapy.175,181,182 

In adults, study MRC AML15 showed that GO was beneficial in those with favorable cytogenetics and in part of patients in the intermediate-risk group; however, there was no benefit to those in the adverse-risk group.174  In addition, other recent international studies on adult AML (by the French and United Kingdom groups) confirmed a survival benefit for AML subgroups when GO was added to induction chemotherapy.180,183–185  Recent data from a prospective nonrandomized COG trial showed that GO did not cause increased toxicity, but data on efficacy are pending.186  However, as the approved indications for GO have been revised in the United States by the FDA request, it may not be easily available for use in pediatric AML outside of clinical trials.

Tyrosine kinase inhibitors

AML patients with activating FLT3 or KIT mutations are candidates for targeted therapy. Many FLT3-targeting tyrosine kinase inhibitors (sorafenib, CEP701, PKC412, AC220) have been on trial in children or used on a compassionate-use basis.187  A recent report from the St Jude Children's Research Hospital demonstrated the feasibility of combining sorafenib and conventional chemotherapy in childhood AML, with some evidence of efficacy limited to patients with FLT3-ITD.188 

Several aspects of supportive care deserve special consideration in children, such as approval and dosing of pharmaceutical agents and proxy-reporting of preferences and quality-of-life status. The improved outcome in children with AML over the last 10 years is probably associated with better supportive care strategies.

Hyperleukocytosis

Initial hyperleukocytosis (arbitrarily defined as WBC ≥ 100 × 109/L) is associated with a high risk of hemorrhage (mainly in the CNS) and leukostasis, which are often fatal. Patients with monocytic or myelomonocytic leukemia (FAB M4/M5) and hyperleukocytosis as well as APL patients have an increased risk of early death, and emergency strategies to reduce these leukemia-related complications are needed.189,190 

In the case of hyperleukocytosis, emergency care with intensive monitoring and careful hydration with concurrent administration of rasburicase may be necessary.191  If there are also symptomatic coagulopathy and/or leukostasis, exchange transfusion or leukapheresis can be life-saving. Initiation of chemotherapy should not be delayed. A controlled but effective cell reduction (eg, low-dose cytarabine; leukapheresis) together with enforced diuresis, or even hemodialysis, may prevent the severe tumor lysis syndrome.189,190 

Recommendation

For patients with initial hyperleukocytosis and symptomatic coagulopathy and/or leukostasis, emergency strategies should be initiated to reduce the risk of fatal hemorrhage and leukostasis.

Prophylactic anti-infectious treatment

Antifungal prophylaxis.

The incidence of invasive fungal infections in children with AML is ∼ 20%, which is similar to that in adults, for whom prophylaxis with an antifungal agent is recommended. In adults, prophylactic posaconazole is more effective at reducing invasive aspergillosis and thus reducing mortality than fluconazole or itraconazole.192  Unfortunately, posaconazole is not licensed for use in children younger than 13 years, and its dosage has not yet been defined for young children in the prophylactic setting; itraconazole, voriconazole, micafungin, or liposomal amphotericin B may be used instead. However, these compounds have limitations, such as variable absorption or multiple potential drug-drug interactions (eg, itraconazole and voriconazole), or can only be administered intravenously (eg, echinocandins and amphotericin B formulations).193  As in other childhood malignancies, cotrimoxazole prophylaxis should be used for Pneumocystis jirovecii.194,195 

Recommendations

Antifungal prophyaxis (including trimethoprim-sulfamethoxazole for prophylaxis of P jirovecii) should be administered to all children.

Antibiotic prophylaxis.

Up to 70% of children have a microbiologically documented bacterial infection per course of AML therapy, and bacterial infections remain a common cause of toxic death.196  Importantly, the cumulative incidence of bacteremia with viridans group streptococci is > 40% and is, in particular, associated with use of HiDAC and mucositis.197  However, there are insufficient data to recommend routine vancomycin prophylaxis in this setting.198  A recent Cochrane review analyzing pediatric and adult patients with various underlying malignancies suggested that antibiotic prophylaxis in afebrile neutropenic patients significantly reduced all-cause mortality, with the most significant reduction in mortality observed in trials assessing prophylaxis with quinolones.199  Therefore, according to current guidelines, fluoroquinolone prophylaxis might be considered for high-risk patients with expected prolonged and profound neutropenia (absolute neutrophil count < 100/μL for > 7 days).199–202  This potential benefit of quinolones must be weighed against an increase in antimicrobial resistance to fluoroquinolones, which are not licensed for use in children.

Recommendation

The use of fluoroquinolones may be considered as prophylaxis against Streptococcus viridans and gram-negative sepsis.

Hematopoietic growth factors

Although a recent survey demonstrated that there is a wide variation in the prophylactic use of G-CSF in children with AML,203  the routine use of hematopoietic growth factors, such as G-CSF or GM-CSF, is not recommended in this patient population. Although G-CSF significantly decreases the duration of neutropenia in induction therapy, it does not influence the incidence of febrile neutropenia or microbiologically documented infections or affect infection-associated mortality.204,205 

It has also been recently demonstrated that children who overexpress the differentiation-defective G-CSFR isoform IV and receive G-CSF have a high risk of relapse.124  However, in critically ill patients, shortening neutropenia by G-CSF may be an option.

Recommendation

Prophylactic hematopoietic growth factors, such as G-CSF, should not be used routinely.

Transfusion support is an important issue in childhood AML. Information regarding this topic is given in the supplemental Appendix (available on the Blood Web site; see the Supplemental Materials link at the top of the online article).

Diagnosis and treatment in pediatric AML has improved considerably during the past decades, with children currently achieving 5-year overall survival rates of ∼ 70%. This has resulted from the cooperative studies of national groups, leading to a stepwise improvement of treatment, including improved supportive care. The definition of risk groups has led to a more risk-adapted treatment, avoiding overtreatment in low-risk patients and including more intensive therapy in others. The current risk definition is mainly based on cytogenetics, a limited number of molecular markers, and the response to induction therapy. Future genomewide approaches, such as gene expression analysis, methylation array studies, genomic profiling, miRNA profiling, and whole-genome sequencing, will further expand the potential of diagnostics and promote the identification of genes and involved pathways, providing better insight into the leukemogenesis of pediatric AML. Knowledge of the biology may then translate into better treatment options and improved cure rates in conjunction with better risk-group adapted therapy and reduced late effects of therapy.

International cooperation is extremely important, as not only is AML rare but also a very heterogenous disease, and only international collaboration will provide adequate numbers of patients to power trial questions within subgroups as new targeted therapies become available to children.

Appendix

Panel

The panel included 21 representatives from national groups from Europe, the United States, and Japan collaborating in the AML committee of the International Berlin/Frankfurt/Muenster (I-BFM) Study Group who have clinical and research expertise in pediatric AML. Between May 2011 and February 2012, the panel met twice in person and communicated extensively via electronic discussion (mails and telephone conferences).

Participating groups

Participating groups included the following: AIDA, Associazione Italiana di Ematologia e Oncologia Pediatrica (Italy); BFM, Berlin/Frankfurt/Münster (Germany, Austria, Switzerland, Czech Republic); COG, Children's Oncology Group (United States); DCOG, Dutch Childhood Oncology Group (The Netherlands); JPLSG, Japan Pediatric Leukemia and Lymphoma Study Group (Japan); LAME, Leucémie Aiguë Myéloblastique Enfant (France); NCRI, National Cancer Research Institute (United Kingdom); NOPHO, Nordic Society of Pediatric Hematology and Oncology (Denmark, Finland, Island, Norway, Sweden); and St Jude AML Study Group (United States).

Scope of the review

The expert panel has developed diagnostic and treatment recommendations for AML in children and adolescents, motivated by recent recommendations from the adult AML panel.4  All the main pediatric AML study groups were included, and an extensive PubMed literature search (keywords = pediatric/childhood acute myeloid leukemia/AML, relapsed acute myeloid leukemia, diagnosis, risk stratification, therapy management, minimal residual disease, mutations AML, cytogenetic/karyotype AML) was performed. The majority of recommendations is based on well-planned but nonrandomized therapy optimizing studies often including randomized questions or on expert opinions with a consensus of the panel.

The online version of this article contains a data supplement.

The authors thank Hartmut Döhner (University Hospital of Ulm, Ulm, Germany) for valuable advice, constructive criticism, and extensive guidance while preparing the manuscript; Ursula Bernsmann (Medical School Hannover, Hannover, Germany) for help in preparing the manuscript; and Dr Vani Shanker (St Jude Children's Research Hospital, Memphis, TN) for editing the manuscript.

Contribution: U.C. assembled the sections; U.C., M.M.v.d.H.-E., D.R., G.J.L.K., B.G., and C.M.Z. wrote the final version of the manuscript; and all authors reviewed the literature, wrote first drafts of specific sections, and reviewed the final version of the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Ursula Creutzig, Hannover Medical School, Children's Hospital, Department of Paediatric Haematology and Oncology, Carl-Neuberg-Str 1, D-30625 Hannover, Germany; e-mail: ursula@creutzig.de.

1
Cheson
 
BD
Cassileth
 
PA
Head
 
DR
, et al. 
Report of the National Cancer Institute-sponsored workshop on definitions of diagnosis and response in acute myeloid leukemia.
J Clin Oncol
1990
, vol. 
8
 
5
(pg. 
813
-
819
)
2
Cheson
 
BD
Bennett
 
JM
Kopecky
 
KJ
, et al. 
Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia.
J Clin Oncol
2003
, vol. 
21
 
24
(pg. 
4642
-
4649
)
3
Swerdlow
 
SH
Campo
 
E
Harris
 
NL
, et al. 
WHO Classification of Tumors of Haemtopoetic and Lymphoid Tissues
2008
Lyon, France
International Agency for Research on Cancer
4
Döhner
 
H
Estey
 
EH
Amadori
 
S
, et al. 
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet.
Blood
2010
, vol. 
115
 
3
(pg. 
453
-
474
)
5
Vardiman
 
JW
Thiele
 
J
Arber
 
DA
, et al. 
The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes.
Blood
2009
, vol. 
114
 
5
(pg. 
937
-
951
)
6
Bene
 
MC
Castoldi
 
G
Knapp
 
W
, et al. 
Proposals for the immunological classification of acute leukemias: European Group for the Immunological Characterization of Leukemias (EGIL).
Leukemia
1995
, vol. 
9
 
10
(pg. 
1783
-
1786
)
7
Bennett
 
JM
Catovsky
 
D
Daniel
 
MT
, et al. 
Proposal for the recognition of minimally differentiated acute myeloid leukaemia (AML-M0).
Br J Haematol
1991
, vol. 
78
 (pg. 
325
-
329
)
8
Hasle
 
H
Niemeyer
 
CM
Chessells
 
JM
, et al. 
A pediatric approach to the WHO classification of myelodysplastic and myeloproliferative diseases.
Leukemia
2003
, vol. 
17
 
2
(pg. 
277
-
282
)
9
Bene
 
MC
Nebe
 
T
Bettelheim
 
P
, et al. 
Immunophenotyping of acute leukemia and lymphoproliferative disorders: a consensus proposal of the European LeukemiaNet Work Package 10.
Leukemia
2011
, vol. 
25
 
4
(pg. 
567
-
574
)
10
Rubnitz
 
JE
Inaba
 
H
Dahl
 
G
, et al. 
Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial.
Lancet Oncol
2010
, vol. 
11
 
6
(pg. 
543
-
552
)
11
Wood
 
BL
Arroz
 
M
Barnett
 
D
, et al. 
2006 Bethesda International Consensus recommendations on the immunophenotypic analysis of hematolymphoid neoplasia by flow cytometry: optimal reagents and reporting for the flow cytometric diagnosis of hematopoietic neoplasia.
Cytometry B Clin Cytom
2007
, vol. 
72
 
Suppl 1
(pg. 
S14
-
S22
)
12
Betts
 
DR
Ammann
 
RA
Hirt
 
A
, et al. 
The prognostic significance of cytogenetic aberrations in childhood acute myeloid leukaemia: a study of the Swiss Paediatric Oncology Group (SPOG).
Eur J Haematol
2007
, vol. 
78
 
6
(pg. 
468
-
476
)
13
Grimwade
 
D
The clinical significance of cytogenetic abnormalities in acute myeloid leukaemia.
Best Pract Res Clin Haematol
2001
, vol. 
14
 
3
(pg. 
497
-
529
)
14
Harrison
 
CJ
Hills
 
RK
Moorman
 
AV
, et al. 
Cytogenetics of childhood acute myeloid leukemia: United Kingdom Medical Research Council Treatment trials AML 10 and 12.
J Clin Oncol
2010
, vol. 
28
 
16
(pg. 
2674
-
2681
)
15
von Neuhoff
 
C
Reinhardt
 
D
Sander
 
A
, et al. 
Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98.
J Clin Oncol
2010
, vol. 
28
 
16
(pg. 
2682
-
2689
)
16
Mercher
 
T
Busson-Le Coniat
 
M
Nguyen
 
KF
, et al. 
Recurrence of OTT-MAL fusion in t(1;22) of infant AML-M7.
Genes Chromosomes Cancer
2002
, vol. 
33
 
1
(pg. 
22
-
28
)
17
Torres
 
L
Lisboa
 
S
Vieira
 
J
, et al. 
Acute megakaryoblastic leukemia with a four-way variant translocation originating the RBM15-MKL1 fusion gene.
Pediatr Blood Cancer
2011
, vol. 
56
 
5
(pg. 
846
-
849
)
18
Park
 
J
Kim
 
M
Lim
 
J
, et al. 
Three-way complex translocations in infant acute myeloid leukemia with t(7;12)(q36;p13): the incidence and correlation of a HLXB9 overexpression.
Cancer Genet Cytogenet
2009
, vol. 
191
 
2
(pg. 
102
-
105
)
19
Simmons
 
HM
Oseth
 
L
Nguyen
 
P
O'Leary
 
M
Conklin
 
KF
Hirsch
 
B
Cytogenetic and molecular heterogeneity of 7q36/12p13 rearrangements in childhood AML.
Leukemia
2002
, vol. 
16
 
12
(pg. 
2408
-
2416
)
20
Slater
 
RM
von Drunen
 
E
Kroes
 
WG
, et al. 
t(7;12)(q36;p13) and t(7;12)(q32;p13)-translocations involving ETV6 in children 18 months of age or younger with myeloid disorders.
Leukemia
2001
, vol. 
15
 
6
(pg. 
915
-
920
)
21
von Bergh
 
AR
van Drunen
 
E
van Wering
 
ER
, et al. 
High incidence of t(7;12)(q36;p13) in infant AML but not in infant ALL, with a dismal outcome and ectopic expression of HLXB9.
Genes Chromosomes Cancer
2006
, vol. 
45
 
8
(pg. 
731
-
739
)
22
Hollink
 
IH
Van den Heuvel-Eibrink
 
MM
Arentsen-Peters
 
ST
, et al. 
NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern.
Blood
2011
, vol. 
118
 
13
(pg. 
3645
-
3656
)
23
Hasle
 
H
Alonzo
 
TA
Auvrignon
 
A
, et al. 
Monosomy 7 and deletion 7q in children and adolescents with acute myeloid leukemia: an international retrospective study.
Blood
2007
, vol. 
109
 
11
(pg. 
4641
-
4647
)
24
Breems
 
DA
Van Putten
 
WL
De Greef
 
GE
, et al. 
Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype.
J Clin Oncol
2008
, vol. 
26
 
29
(pg. 
4791
-
4797
)
25
Creutzig
 
U
Buchner
 
T
Sauerland
 
MC
, et al. 
Significance of age in acute myeloid leukemia patients younger than 30 years.
Cancer
2008
, vol. 
112
 
3
(pg. 
562
-
571
)
26
Gilliland
 
DG
Molecular genetics of human leukemias: new insights into therapy.
Semin Hematol
2002
, vol. 
39
 
4 Suppl 3
(pg. 
6
-
11
)
27
Balgobind
 
BV
Hollink
 
IH
Arentsen-Peters
 
ST
, et al. 
Integrative analysis of type-I and type-II aberrations underscores the genetic heterogeneity of pediatric acute myeloid leukemia.
Haematologica
2011
, vol. 
96
 
10
(pg. 
1478
-
1487
)
28
Ho
 
PA
Alonzo
 
TA
Gerbing
 
RB
, et al. 
Prevalence and prognostic implications of CEBPA mutations in pediatric acute myeloid leukemia (AML): a report from the Children's Oncology Group.
Blood
2009
, vol. 
113
 
26
(pg. 
6558
-
6566
)
29
Hollink
 
IH
Zwaan
 
CM
Zimmermann
 
M
, et al. 
Favorable prognostic impact of NPM1 gene mutations in childhood acute myeloid leukemia, with emphasis on cytogenetically normal AML.
Leukemia
2009
, vol. 
23
 
2
(pg. 
262
-
270
)
30
Meshinchi
 
S
Alonzo
 
TA
Stirewalt
 
DL
, et al. 
Clinical implications of FLT3 mutations in pediatric AML.
Blood
2006
, vol. 
108
 
12
(pg. 
3654
-
3661
)
31
Pui
 
CH
Carroll
 
WL
Meshinchi
 
S
Arceci
 
RJ
Biology, risk stratification, and therapy of pediatric acute leukemias: an update.
J Clin Oncol
2011
, vol. 
29
 
5
(pg. 
551
-
565
)
32
Brown
 
P
McIntyre
 
E
Rau
 
R
, et al. 
The incidence and clinical significance of nucleophosmin mutations in childhood AML.
Blood
2007
, vol. 
110
 
3
(pg. 
979
-
985
)
33
Thiede
 
C
Creutzig
 
E
Reinhardt
 
D
Ehninger
 
G
Creutzig
 
U
Different types of NPM1 mutations in children and adults: evidence for an effect of patient age on the prevalence of the TCTG-tandem duplication in NPM1-exon 12.
Leukemia
2007
, vol. 
21
 
2
(pg. 
366
-
367
)
34
Cazzaniga
 
G
Dell'Oro
 
MG
Mecucci
 
C
, et al. 
Nucleophosmin mutations in childhood acute myelogenous leukemia with normal karyotype.
Blood
2005
, vol. 
106
 
4
(pg. 
1419
-
1422
)
35
Balgobind
 
BV
Van den Heuvel-Eibrink
 
MM
De Menezes
 
RX
, et al. 
Evaluation of gene expression signatures predictive of cytogenetic and molecular subtypes of pediatric acute myeloid leukemia.
Haematologica
2011
, vol. 
96
 
2
(pg. 
221
-
230
)
36
Ho
 
PA
Zeng
 
R
Alonzo
 
TA
, et al. 
Prevalence and prognostic implications of WT1 mutations in pediatric acute myeloid leukemia (AML): a report from the Children's Oncology Group.
Blood
2010
, vol. 
116
 
5
(pg. 
702
-
710
)
37
Hollink
 
IH
Van den Heuvel-Eibrink
 
MM
Zimmermann
 
M
, et al. 
Clinical relevance of Wilms tumor 1 gene mutations in childhood acute myeloid leukemia.
Blood
2009
, vol. 
113
 
23
(pg. 
5951
-
5960
)
38
Meshinchi
 
S
Appelbaum
 
FR
Structural and functional alterations of FLT3 in acute myeloid leukemia.
Clin Cancer Res
2009
, vol. 
15
 
13
(pg. 
4263
-
4269
)
39
Berman
 
JN
Gerbing
 
RB
Alonzo
 
TA
, et al. 
Prevalence and clinical implications of NRAS mutations in childhood AML: a report from the Children's Oncology Group.
Leukemia
2011
, vol. 
25
 
6
(pg. 
1039
-
1042
)
40
Boissel
 
N
Leroy
 
H
Brethon
 
B
, et al. 
Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML).
Leukemia
2006
, vol. 
20
 
6
(pg. 
965
-
970
)
41
Shih
 
LY
Liang
 
DC
Huang
 
CF
, et al. 
Cooperating mutations of receptor tyrosine kinases and Ras genes in childhood core-binding factor acute myeloid leukemia and a comparative analysis on paired diagnosis and relapse samples.
Leukemia
2008
, vol. 
22
 
2
(pg. 
303
-
307
)
42
Loh
 
ML
Reynolds
 
MG
Vattikuti
 
S
, et al. 
PTPN11 mutations in pediatric patients with acute myeloid leukemia: results from the Children's Cancer Group.
Leukemia
2004
, vol. 
18
 
11
(pg. 
1831
-
1834
)
43
Goemans
 
BF
Zwaan
 
CM
Miller
 
M
, et al. 
Mutations in KIT and RAS are frequent events in pediatric core-binding factor acute myeloid leukemia.
Leukemia
2005
, vol. 
19
 
9
(pg. 
1536
-
1542
)
44
Pollard
 
JA
Alonzo
 
TA
Gerbing
 
RB
, et al. 
Prevalence and prognostic significance of KIT mutations in pediatric patients with core binding factor AML enrolled on serial pediatric cooperative trials for de novo AML.
Blood
2010
, vol. 
115
 
12
(pg. 
2372
-
2379
)
45
Balgobind
 
BV
Hollink
 
IH
Reinhardt
 
D
, et al. 
Low frequency of MLL-partial tandem duplications in paediatric acute myeloid leukaemia using MLPA as a novel DNA screenings technique.
Eur J Cancer
2010
, vol. 
46
 
10
(pg. 
1892
-
1899
)
46
Balgobind
 
BV
Lugthart
 
S
Hollink
 
IH
, et al. 
EVI1 overexpression in distinct subtypes of pediatric acute myeloid leukemia.
Leukemia
2010
, vol. 
24
 
5
(pg. 
942
-
949
)
47
Staffas
 
A
Kanduri
 
M
Hovland
 
R
, et al. 
Presence of FLT3-ITD and high BAALC expression are independent prognostic markers in childhood acute myeloid leukemia.
Blood
2011
, vol. 
118
 
22
(pg. 
5905
-
5913
)
48
Damm
 
F
Thol
 
F
Hollink
 
I
, 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
, vol. 
25
 
11
(pg. 
1704
-
1710
)
49
Ho
 
PA
Kutny
 
MA
Alonzo
 
TA
, et al. 
Leukemic mutations in the methylation-associated genes DNMT3A and IDH2 are rare events in pediatric AML: a report from the Children's Oncology Group.
Pediatr Blood Cancer
2011
, vol. 
57
 
2
(pg. 
204
-
209
)
50
Hollink
 
IH
Feng
 
Q
Danen-van Oorschot
 
AA
, 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
, vol. 
26
 
2
(pg. 
371
-
373
)
51
Mardis
 
ER
Ding
 
L
Dooling
 
DJ
, et al. 
Recurring mutations found by sequencing an acute myeloid leukemia genome.
N Engl J Med
2009
, vol. 
361
 
11
(pg. 
1058
-
1066
)
52
Thol
 
F
Damm
 
F
Ludeking
 
A
, et al. 
Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia.
J Clin Oncol
2011
, vol. 
29
 
21
(pg. 
2889
-
2896
)
53
Ross
 
ME
Mahfouz
 
R
Onciu
 
M
, et al. 
Gene expression profiling of pediatric acute myelogenous leukemia.
Blood
2004
, vol. 
104
 
12
(pg. 
3679
-
3687
)
54
Creutzig
 
U
Zimmermann
 
M
Bourquin
 
J-P
, et al. 
Favorable outcome in infants with AML after intensive first- and second-line treatment: an AML-BFM study group report.
Leukemia
2012
, vol. 
26
 
4
(pg. 
654
-
661
)
55
Creutzig
 
U
Zimmermann
 
M
Ritter
 
J
, et al. 
Definition of a standard-risk group in children with AML.
Br J Haematol
1999
, vol. 
104
 
3
(pg. 
630
-
639
)
56
Balgobind
 
BV
Raimondi
 
SC
Harbott
 
J
, et al. 
Novel prognostic subgroups in childhood 11q23/MLL-rearranged acute myeloid leukemia: results of an international retrospective study.
Blood
2009
, vol. 
114
 
12
(pg. 
2489
-
2496
)
57
Coenen
 
EA
Zwaan
 
CM
Meyer
 
C
, et al. 
KIAA1524: a novel MLL translocation partner in acute myeloid leukemia.
Leuk Res
2011
, vol. 
35
 
1
(pg. 
133
-
135
)
58
Noronha
 
SA
Farrar
 
JE
Alonzo
 
TA
, et al. 
WT1 expression at diagnosis does not predict survival in pediatric AML: a report from the Children's Oncology Group.
Pediatr Blood Cancer
2009
, vol. 
53
 
6
(pg. 
1136
-
1139
)
59
Abrahamsson
 
J
Forestier
 
E
Heldrup
 
J
, et al. 
Response-guided induction therapy in pediatric acute myeloid leukemia with excellent remission rate.
J Clin Oncol
2011
, vol. 
29
 
3
(pg. 
310
-
315
)
60
Creutzig
 
U
Zimmermann
 
M
Ritter
 
J
, et al. 
Definition of a standard-risk group in children with AML.
Br J Haematol
1999
, vol. 
104
 (pg. 
630
-
639
)
61
Wheatley
 
K
Burnett
 
AK
Goldstone
 
AH
, et al. 
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
, vol. 
107
 
1
(pg. 
69
-
79
)
62
Langebrake
 
C
Creutzig
 
U
Dworzak
 
M
, et al. 
Residual disease monitoring in childhood acute myeloid leukemia by multiparameter flow cytometry: the MRD-AML-BFM Study Group.
J Clin Oncol
2006
, vol. 
24
 
22
(pg. 
3686
-
3692
)
63
Van der Velden
 
V
Sluijs-Geling
 
A
Gibson
 
BE
, 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
, vol. 
24
 
9
(pg. 
1599
-
1606
)
64
Viehmann
 
S
Teigler-Schlegel
 
A
Bruch
 
J
Langebrake
 
C
Reinhardt
 
D
Harbott
 
J
Monitoring of minimal residual disease (MRD) by real-time quantitative reverse transcription PCR (RQ-RT-PCR) in childhood acute myeloid leukemia with AML1/ETO rearrangement.
Leukemia
2003
, vol. 
17
 
6
(pg. 
1130
-
1136
)
65
Ommen
 
HB
Schnittger
 
S
Jovanovic
 
JV
, et al. 
Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias.
Blood
2010
, vol. 
115
 
2
(pg. 
198
-
205
)
66
Alford
 
KA
Reinhardt
 
K
Garnett
 
C
, et al. 
Analysis of GATA1 mutations in Down syndrome transient myeloproliferative disorder and myeloid leukemia.
Blood
2011
, vol. 
118
 
8
(pg. 
2222
-
2238
)
67
Wells
 
RJ
Woods
 
WG
Lampkin
 
BC
, et al. 
Impact of high-dose cytarabine and asparaginase intensification on childhood acute myeloid leukemia: a report from the Childrens Cancer Group.
J Clin Oncol
1993
, vol. 
11
 
3
(pg. 
538
-
545
)
68
Perel
 
Y
Auvrignon
 
A
Leblanc
 
T
, 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
, vol. 
20
 
12
(pg. 
2774
-
2782
)
69
Estey
 
E
Dohner
 
H
Acute myeloid leukaemia.
Lancet
2006
, vol. 
368
 
9550
(pg. 
1894
-
1907
)
70
Lowenberg
 
B
Griffin
 
JD
Tallman
 
MS
Acute myeloid leukemia and acute promyelocytic leukemia.
Hematology Am Soc Hematol Educ Program
2003
, vol. 
2003
 (pg. 
82
-
101
)
71
Woods
 
WG
Kobrinsky
 
N
Buckley
 
JD
, et al. 
Timed-sequential induction therapy improves postremission outcome in acute myeloid leukemia: a report from the Children's Cancer Group.
Blood
1996
, vol. 
87
 
12
(pg. 
4979
-
4989
)
72
Kaspers
 
GJ
Pediatric acute myeloid leukemia.
Expert Rev Anticancer Ther
2012
, vol. 
12
 
3
(pg. 
405
-
413
)
73
Fernandez
 
HF
Sun
 
Z
Yao
 
X
, et al. 
Anthracycline dose intensification in acute myeloid leukemia.
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1249
-
1259
)
74
Lowenberg
 
B
Ossenkoppele
 
GJ
van Putten
 
W
, et al. 
High-dose daunorubicin in older patients with acute myeloid leukemia.
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1235
-
1248
)
75
van Dalen
 
EC
van der Pal
 
HJ
Caron
 
HN
Kremer
 
LC
Different dosage schedules for reducing cardiotoxicity in cancer patients receiving anthracycline chemotherapy.
Cochrane Database Syst Rev
2009
4
pg. 
CD005008
 
76
van Dalen
 
EC
Michiels
 
EM
Caron
 
HN
Kremer
 
LC
Different anthracycline derivates for reducing cardiotoxicity in cancer patients.
Cochrane Database Syst Rev
2010
3
pg. 
CD005006
 
77
Kremer
 
LC
van der Pal
 
HJ
Offringa
 
M
van Dalen
 
EC
Voute
 
PA
Frequency and risk factors of subclinical cardiotoxicity after anthracycline therapy in children: a systematic review.
Ann Oncol
2002
, vol. 
13
 
6
(pg. 
819
-
829
)
78
Grenier
 
MA
Lipshultz
 
SE
Epidemiology of anthracycline cardiotoxicity in children and adults.
Semin Oncol
1998
, vol. 
25
 
4 Suppl 10
(pg. 
72
-
85
)
79
Bielack
 
SS
Erttmann
 
R
Kempf-Bielack
 
B
Winkler
 
K
Impact of scheduling on toxicity and clinical efficacy of doxorubicin: what do we know in the mid-nineties?
Eur J Cancer
1996
, vol. 
32A
 
10
(pg. 
1652
-
1660
)
80
Lipshultz
 
SE
Giantris
 
AL
Lipsitz
 
SR
, et al. 
Doxorubicin administration by continuous infusion is not cardioprotective: the Dana-Farber 91–01 Acute Lymphoblastic Leukemia protocol.
J Clin Oncol
2002
, vol. 
20
 
6
(pg. 
1677
-
1682
)
81
Smith
 
LA
Cornelius
 
VR
Plummer
 
CJ
, et al. 
Cardiotoxicity of anthracycline agents for the treatment of cancer: systematic review and meta-analysis of randomised controlled trials.
BMC Cancer
2010
, vol. 
10
 pg. 
337
 
82
Creutzig
 
U
Ritter
 
J
Zimmermann
 
M
, 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
, vol. 
15
 
3
(pg. 
348
-
354
)
83
Gibson
 
BE
Webb
 
DK
Howman
 
AJ
De Graaf
 
SS
Harrison
 
CJ
Wheatley
 
K
Results of a randomized trial in children with acute myeloid leukaemia: Medical Research Council AML12 trial.
Br J Haematol
2011
, vol. 
155
 
3
(pg. 
366
-
376
)
84
Lipshultz
 
SE
Alvarez
 
JA
Scully
 
RE
Anthracycline associated cardiotoxicity in survivors of childhood cancer.
Heart
2008
, vol. 
94
 
4
(pg. 
525
-
533
)
85
U.S. Food and Drug Administration
FDA statement on dexrazoxane.
Accessed July 20, 2011 
86
Burnett
 
AK
Hills
 
RK
Milligan
 
DW
, et al. 
Attempts to optimize induction and consolidation treatment in acute myeloid leukemia: results of the MRC AML12 trial.
J Clin Oncol
2010
, vol. 
28
 
4
(pg. 
586
-
595
)
87
Creutzig
 
U
Zimmermann
 
M
Bourquin
 
JP
, 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
, vol. 
118
 
20
(pg. 
5409
-
5415
)
88
Arlin
 
Z
Case
 
DC
Moore
 
J
, et al. 
Randomized multicenter trial of cytosine arabinoside with mitoxantrone or daunorubicin in previously untreated adult patients with acute nonlymphocytic leukemia (ANLL): Lederle Cooperative Group.
Leukemia
1990
, vol. 
4
 
3
(pg. 
177
-
183
)
89
Michel
 
G
Baruchel
 
A
Tabone
 
MD
, et al. 
Induction chemotherapy followed by allogeneic bone marrow transplantation or aggressive consolidation chemotherapy in childhood acute myeloblastic leukemia: a prospective study from the French Society of Pediatric Hematology and Immunology (SHIP).
Hematol Cell Ther
1996
, vol. 
38
 
2
(pg. 
169
-
176
)
90
Krance
 
RA
Hurwitz
 
CA
Head
 
DR
, et al. 
Experience with 2-chlorodeoxyadenosine in previously untreated children with newly diagnosed acute myeloid leukemia and myelodysplastic diseases.
J Clin Oncol
2001
, vol. 
19
 
11
(pg. 
2804
-
2811
)
91
Mayer
 
RJ
Davis
 
RB
Schiffer
 
CA
, et al. 
Intensive postremission chemotherapy in adults with acute myeloid leukemia: Cancer and Leukemia Group B.
N Engl J Med
1994
, vol. 
331
 
14
(pg. 
896
-
903
)
92
Bloomfield
 
CD
Lawrence
 
D
Byrd
 
JC
, et al. 
Frequency of prolonged remission duration after high-dose cytarabine intensification in acute myeloid leukemia varies by cytogenetic subtype.
Cancer Res
1998
, vol. 
58
 
18
(pg. 
4173
-
4179
)
93
Lie
 
SO
Abrahamsson
 
J
Clausen
 
N
, et al. 
Treatment stratification based on initial in vivo response in acute myeloid leukaemia in children without Down's syndrome: results of NOPHO-AML trials.
Br J Haematol
2003
, vol. 
122
 
2
(pg. 
217
-
225
)
94
Gibson
 
BE
Wheatley
 
K
Hann
 
IM
, et al. 
Treatment strategy and long-term results in paediatric patients treated in consecutive UK AML trials.
Leukemia
2005
, vol. 
19
 
12
(pg. 
2130
-
2138
)
95
Stevens
 
RF
Hann
 
IM
Wheatley
 
K
Gray
 
RG
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
, vol. 
101
 
1
(pg. 
130
-
140
)
96
Creutzig
 
U
Zimmermann
 
M
Dworzak
 
M
, et al. 
Study AML-BFM 2004: improved survival in childhood acute myeloid leukemia without increased toxicity [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
 
21
 
Abstract 181
97
Tsukimoto
 
I
Tawa
 
A
Horibe
 
K
, 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
, vol. 
27
 
24
(pg. 
4007
-
4013
)
98
Alonzo
 
TA
Wells
 
RJ
Woods
 
WG
, et al. 
Postremission therapy for children with acute myeloid leukemia: the Children's Cancer Group experience in the transplant era.
Leukemia
2005
, vol. 
19
 
6
(pg. 
965
-
970
)
99
Oliansky
 
DM
Rizzo
 
JD
Aplan
 
PD
, et al. 
The role of cytotoxic therapy with hematopoietic stem cell transplantation in the therapy of acute myeloid leukemia in children: an evidence-based review.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
1
(pg. 
1
-
25
)
100
Ravindranath
 
Y
Yeager
 
AM
Chang
 
MN
, et al. 
Autologous bone marrow transplantation versus intensive consolidation chemotherapy for acute myeloid leukemia in childhood: Pediatric Oncology Group.
N Engl J Med
1996
, vol. 
334
 
22
(pg. 
1428
-
1434
)
101
Woods
 
WG
Neudorf
 
S
Gold
 
S
, et al. 
A comparison of allogeneic bone marrow transplantation, autologous bone marrow transplantation, and aggressive chemotherapy in children with acute myeloid leukemia in remission.
Blood
2001
, vol. 
97
 
1
(pg. 
56
-
62
)
102
Sanz
 
MA
Grimwade
 
D
Tallman
 
MS
, et al. 
Management of acute promyelocytic leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet.
Blood
2009
, vol. 
113
 
9
(pg. 
1875
-
1891
)
103
Horan
 
JT
Alonzo
 
TA
Lyman
 
GH
, et al. 
Impact of disease risk on efficacy of matched related bone marrow transplantation for pediatric acute myeloid leukemia: the Children's Oncology Group.
J Clin Oncol
2008
, vol. 
26
 
35
(pg. 
5797
-
5801
)
104
Klingebiel
 
T
Reinhardt
 
D
Bader
 
P
Place of HSCT in treatment of childhood AML.
Bone Marrow Transplant
2008
, vol. 
42
 
Suppl 2
(pg. 
S7
-
S9
)
105
Niewerth
 
D
Creutzig
 
U
Bierings
 
MB
Kaspers
 
GJ
A review on allogeneic stem cell transplantation for newly diagnosed pediatric acute myeloid leukemia.
Blood
2010
, vol. 
116
 
13
(pg. 
2205
-
2214
)
106
Perel
 
Y
Auvrignon
 
A
Leblanc
 
T
, et al. 
Treatment of childhood acute myeloblastic leukemia: dose intensification improves outcome and maintenance therapy is of no benefit-multicenter studies of the French LAME (Leucemie Aigue Myeloblastique Enfant) Cooperative Group.
Leukemia
2005
, vol. 
19
 
12
(pg. 
2082
-
2089
)
107
Bhatia
 
S
Long-term health impacts of hematopoietic stem cell transplantation inform recommendations for follow-up.
Expert Rev Hematol
2011
, vol. 
4
 
4
(pg. 
437
-
452
)
108
Bresters
 
D
van Gils
 
IC
Kollen
 
WJ
, et al. 
High burden of late effects after haematopoietic stem cell transplantation in childhood: a single-centre study.
Bone Marrow Transplant
2010
, vol. 
45
 
1
(pg. 
79
-
85
)
109
Gupta
 
T
Kannan
 
S
Dantkale
 
V
Laskar
 
S
Cyclophosphamide plus total body irradiation compared with busulfan plus cyclophosphamide as a conditioning regimen prior to hematopoietic stem cell transplantation in patients with leukemia: a systematic review and meta-analysis.
Hematol Oncol Stem Cell Ther
2011
, vol. 
4
 
1
(pg. 
17
-
29
)
110
Appelbaum
 
FR
Optimising the conditioning regimen for acute myeloid leukaemia.
Best Pract Res Clin Haematol
2009
, vol. 
22
 
4
(pg. 
543
-
550
)
111
Socie
 
G
Clift
 
RA
Blaise
 
D
, et al. 
Busulfan plus cyclophosphamide compared with total-body irradiation plus cyclophosphamide before marrow transplantation for myeloid leukemia: long-term follow-up of 4 randomized studies.
Blood
2001
, vol. 
98
 
13
(pg. 
3569
-
3574
)
112
Appelbaum
 
FR
Hematopoietic cell transplantation from unrelated donors for treatment of patients with acute myeloid leukemia in first complete remission.
Best Pract Res Clin Haematol
2007
, vol. 
20
 
1
(pg. 
67
-
75
)
113
Walter
 
RB
Gooley
 
TA
Wood
 
BL
, et al. 
Impact of pretransplantation minimal residual disease, as detected by multiparametric flow cytometry, on outcome of myeloablative hematopoietic cell transplantation for acute myeloid leukemia.
J Clin Oncol
2011
, vol. 
29
 
9
(pg. 
1190
-
1197
)
114
Bonanomi
 
S
Connor
 
P
Webb
 
D
, et al. 
Successful outcome of allo-SCT in high-risk pediatric AML using chemotherapy-only conditioning and post transplant immunotherapy.
Bone Marrow Transplant
2008
, vol. 
42
 
4
(pg. 
253
-
257
)
115
Creutzig
 
U
Zimmermann
 
M
Bourquin
 
JP
, et al. 
CNS irradiation in pediatric acute myleoid leukemia: equal results by 12 or 18 Gy in studies AML-BFM98 and 2004.
Pediatr Blood Cancer
2011
, vol. 
57
 
6
(pg. 
986
-
992
)
116
Dahl
 
GV
Simone
 
JV
Hustu
 
HO
Mason
 
C
Preventive central nervous system irradiation in children with acute nonlymphocytic leukemia.
Cancer
1978
, vol. 
42
 
5
(pg. 
2187
-
2192
)
117
Creutzig
 
U
Ritter
 
J
Zimmermann
 
M
Schellong
 
G
Does cranial irradiation reduce the risk for bone marrow relapse in acute myelogenous leukemia? Unexpected results of the Childhood Acute Myelogenous Leukemia Study BFM-87.
J Clin Oncol
1993
, vol. 
11
 
2
(pg. 
279
-
286
)
118
Pui
 
CH
Howard
 
SC
Current management and challenges of malignant disease in the CNS in paediatric leukaemia.
Lancet Oncol
2008
, vol. 
9
 
3
(pg. 
257
-
268
)
119
Johnston
 
DL
Alonzo
 
TA
Gerbing
 
RB
Lange
 
BJ
Woods
 
WG
The presence of central nervous system disease at diagnosis in pediatric acute myeloid leukemia does not affect survival: a Children's Oncology Group study.
Pediatr Blood Cancer
2010
, vol. 
55
 
3
(pg. 
414
-
420
)
120
Pession
 
A
The open issue of central nervous system disease in pediatric acute myeloid leukemia.
Pediatr Blood Cancer
2010
, vol. 
55
 
3
(pg. 
399
-
400
)
121
Abbott
 
BL
Rubnitz
 
JE
Tong
 
X
, et al. 
Clinical significance of central nervous system involvement at diagnosis of pediatric acute myeloid leukemia: a single institution's experience.
Leukemia
2003
, vol. 
17
 
11
(pg. 
2090
-
2096
)
122
Mayadev
 
JS
Douglas
 
JG
Storer
 
BE
Appelbaum
 
FR
Storb
 
R
Impact of cranial irradiation added to intrathecal conditioning in hematopoietic cell transplantation in adult acute myeloid leukemia with central nervous system involvement.
Int J Radiat Oncol Biol Phys
2011
, vol. 
80
 
1
(pg. 
193
-
198
)
123
Johnston
 
DL
Alonzo
 
TA
Gerbing
 
RB
Lange
 
BJ
Woods
 
WG
Risk factors and therapy for isolated central nervous system relapse of pediatric acute myeloid leukemia.
J Clin Oncol
2005
, vol. 
23
 
36
(pg. 
9172
-
9178
)
124
Ehlers
 
S
Herbst
 
C
Zimmermann
 
M
, et al. 
Granulocyte colony-stimulating factor (G-CSF) treatment of childhood acute myeloid leukemias that overexpress the differentiation-defective G-CSF receptor isoform IV is associated with a higher incidence of relapse.
J Clin Oncol
2010
, vol. 
28
 
15
(pg. 
2591
-
2597
)
125
Lee
 
YJ
Moon
 
JH
Kim
 
JG
, et al. 
Prospective randomization trial of G-CSF-primed induction regimen versus standard regimen in patients with AML.
Chonnam Med J
2011
, vol. 
47
 
2
(pg. 
80
-
84
)
126
Lowenberg
 
B
van Putten
 
W
Theobald
 
M
, et al. 
Effect of priming with granulocyte colony-stimulating factor on the outcome of chemotherapy for acute myeloid leukemia.
N Engl J Med
2003
, vol. 
349
 
8
(pg. 
743
-
752
)
127
Periclou
 
AP
Avramis
 
VI
NONMEM population pharmacokinetic studies of cytosine arabinoside after high-dose and after loading bolus followed by continuous infusion of the drug in pediatric patients with leukemias.
Cancer Chemother Pharmacol
1996
, vol. 
39
 
1
(pg. 
42
-
50
)
128
van den Berg
 
H
Hopman
 
AH
Kraakman
 
KC
de Jong
 
D
Spontaneous remission in congenital leukemia is not related to (mosaic) trisomy 21: case presentation and literature review.
Pediatr Hematol Oncol
2004
, vol. 
21
 
2
(pg. 
135
-
144
)
129
Campidelli
 
C
Agostinelli
 
C
Stitson
 
R
Pileri
 
SA
Myeloid sarcoma: extramedullary manifestation of myeloid disorders.
Am J Clin Pathol
2009
, vol. 
132
 
3
(pg. 
426
-
437
)
130
Harris
 
NL
Jaffe
 
ES
Diebold
 
J
, et al. 
World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting–Airlie House, Virginia, November 1997.
J Clin Oncol
1999
, vol. 
17
 
12
(pg. 
3835
-
3849
)
131
Reinhardt
 
D
Creutzig
 
U
Isolated myelosarcoma in children-update and review.
Leuk Lymphoma
2002
, vol. 
43
 
3
(pg. 
565
-
574
)
132
Felice
 
MS
Zubizarreta
 
PA
Alfaro
 
EM
, et al. 
Good outcome of children with acute myeloid leukemia and t(8;21)(q22;q22), even when associated with granulocytic sarcoma: a report from a single institution in Argentina.
Cancer
2000
, vol. 
88
 
8
(pg. 
1939
-
1944
)
133
Matutes
 
E
Pickl
 
WF
Van't Veer
 
M
, et al. 
Mixed-phenotype acute leukemia: clinical and laboratory features and outcome in 100 patients defined according to the WHO 2008 classification.
Blood
2011
, vol. 
117
 
11
(pg. 
3163
-
3171
)
134
Gerr
 
H
Zimmermann
 
M
Schrappe
 
M
, et al. 
Acute leukaemias of ambiguous lineage in children: characterization, prognosis and therapy recommendations.
Br J Haematol
2010
, vol. 
149
 
1
(pg. 
84
-
92
)
135
Mejstrikova
 
E
Volejnikova
 
J
Fronkova
 
E
, et al. 
Prognosis of children with mixed phenotype acute leukemia treated on the basis of consistent immunophenotypic criteria.
Haematologica
2010
, vol. 
95
 
6
(pg. 
928
-
935
)
136
Miller
 
DR
Miller
 
LP
Acute lymphoblastic leukemia in children: an update of clinical, biological, and therapeutic aspects.
Crit Rev Oncol Hematol
1990
, vol. 
10
 
2
(pg. 
131
-
164
)
137
Gamis
 
AS
Alonzo
 
TA
Gerbing
 
RB
, et al. 
Natural history of transient myeloproliferative disorder clinically diagnosed in Down syndrome neonates: a report from the Children's Oncology Group Study A2971.
Blood
2011
, vol. 
118
 
26
(pg. 
6752
-
6759
)
138
Hitzler
 
JK
Cheung
 
J
Li
 
Y
Scherer
 
SW
Zipursky
 
A
GATA1 mutations in transient leukemia and acute megakaryoblastic leukemia of Down syndrome.
Blood
2003
, vol. 
101
 
11
(pg. 
4301
-
4304
)
139
Wechsler
 
J
Greene
 
M
McDevitt
 
MA
, et al. 
Acquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome.
Nat Genet
2002
, vol. 
32
 
1
(pg. 
148
-
152
)
140
Li
 
Z
Godinho
 
FJ
Klusmann
 
JH
Garriga-Canut
 
M
Yu
 
C
Orkin
 
SH
Developmental stage-selective effect of somatically mutated leukemogenic transcription factor GATA1.
Nat Genet
2005
, vol. 
37
 
6
(pg. 
613
-
619
)
141
Klusmann
 
JH
Creutzig
 
U
Zimmermann
 
M
, et al. 
Treatment and prognostic impact of transient leukemia in neonates with Down's syndrome.
Blood
2008
, vol. 
111
 
6
(pg. 
2991
-
2998
)
142
Creutzig
 
U
Reinhardt
 
D
Diekamp
 
S
Dworzak
 
M
Stary
 
J
Zimmermann
 
M
AML patients with Down syndrome have a high cure rate with AML-BFM therapy with reduced dose intensity.
Leukemia
2005
, vol. 
19
 
8
(pg. 
1355
-
1360
)
143
Kudo
 
K
Hama
 
A
Kojima
 
S
, et al. 
Mosaic Down syndrome-associated acute myeloid leukemia does not require high-dose cytarabine treatment for induction and consolidation therapy.
Int J Hematol
2010
, vol. 
91
 
4
(pg. 
630
-
635
)
144
Kudo
 
K
Kojima
 
S
Tabuchi
 
K
, et al. 
Prospective study of a pirarubicin, intermediate-dose cytarabine, and etoposide regimen in children with Down syndrome and acute myeloid leukemia: the Japanese Childhood AML Cooperative Study Group.
J Clin Oncol
2007
, vol. 
25
 
34
(pg. 
5442
-
5447
)
145
Zwaan
 
CM
Kaspers
 
GJ
Pieters
 
R
, et al. 
Different drug sensitivity profiles of acute myeloid and lymphoblastic leukemia and normal peripheral blood mononuclear cells in children with and without Down syndrome.
Blood
2002
, vol. 
99
 
1
(pg. 
245
-
251
)
146
Taub
 
JW
Ge
 
Y
Down syndrome, drug metabolism and chromosome 21.
Pediatr Blood Cancer
2005
, vol. 
44
 
1
(pg. 
33
-
39
)
147
Li
 
FP
Bader
 
JL
Nathan
 
DG
Oski
 
FA
Epidemiology of cancer in childhood.
Hematology of Infancy and Childhood
1993
Philadelphia, PA
Saunders
(pg. 
1102
-
1119
)
148
Belson
 
M
Kingsley
 
B
Holmes
 
A
Risk factors for acute leukemia in children: a review.
Environ Health Perspect
2007
, vol. 
115
 
1
(pg. 
138
-
145
)
149
Mehta
 
PA
Ileri
 
T
Harris
 
RE
, et al. 
Chemotherapy for myeloid malignancy in children with Fanconi anemia.
Pediatr Blood Cancer
2007
, vol. 
48
 
7
(pg. 
668
-
672
)
150
Stelljes
 
M
Corbacioglu
 
A
Schlenk
 
RF
, et al. 
Allogeneic stem cell transplant to eliminate germline mutations in the gene for CCAAT-enhancer-binding protein alpha from hematopoietic cells in a family with AML.
Leukemia
2011
, vol. 
25
 
7
(pg. 
1209
-
1210
)
151
Gregory
 
J
Feusner
 
J
Acute promyelocytic leukemia in childhood.
Curr Oncol Rep
2009
, vol. 
11
 
6
(pg. 
439
-
445
)
152
Mann
 
G
Reinhardt
 
D
Ritter
 
J
, et al. 
Treatment with all-trans retinoic acid in acute promyelocytic leukemia reduces early deaths in children.
Ann Hematol
2001
, vol. 
80
 
7
(pg. 
417
-
422
)
153
de Botton
 
S
Coiteux
 
V
Chevret
 
S
, et al. 
Outcome of childhood acute promyelocytic leukemia with all-trans-retinoic acid and chemotherapy.
J Clin Oncol
2004
, vol. 
22
 
8
(pg. 
1404
-
1412
)
154
Ortega
 
JJ
Madero
 
L
Martin
 
G
, et al. 
Treatment with all-trans retinoic acid and anthracycline monochemotherapy for children with acute promyelocytic leukemia: a multicenter study by the PETHEMA Group.
J Clin Oncol
2005
, vol. 
23
 
30
(pg. 
7632
-
7640
)
155
Testi
 
AM
Biondi
 
A
Lo
 
CF
, et al. 
GIMEMA-AIEOPAIDA protocol for the treatment of newly diagnosed acute promyelocytic leukemia (APL) in children.
Blood
2005
, vol. 
106
 
2
(pg. 
447
-
453
)
156
Asou
 
N
Kishimoto
 
Y
Kiyoi
 
H
, et al. 
A randomized study with or without intensified maintenance chemotherapy in patients with acute promyelocytic leukemia who have become negative for PML-RARalpha transcript after consolidation therapy: the Japan Adult Leukemia Study Group (JALSG) APL97 study.
Blood
2007
, vol. 
110
 
1
(pg. 
59
-
66
)
157
Sanz
 
MA
Martin
 
G
Rayon
 
C
, et al. 
A modified AIDA protocol with anthracycline-based consolidation results in high antileukemic efficacy and reduced toxicity in newly diagnosed PML/RARalpha-positive acute promyelocytic leukemia: PETHEMA group.
Blood
1999
, vol. 
94
 
9
(pg. 
3015
-
3021
)
158
Creutzig
 
U
Zimmermann
 
M
Dworzak
 
M
, et al. 
Favourable outcome of patients with childhood acute promyelocytic leukaemia after treatment with reduced cumulative anthracycline doses.
Br J Haematol
2010
, vol. 
149
 
3
(pg. 
399
-
409
)
159
Lo-Coco
 
F
Ammatuna
 
E
Front line clinical trials and minimal residual disease monitoring in acute promyelocytic leukemia.
Curr Top Microbiol Immunol
2007
, vol. 
313
 (pg. 
145
-
156
)
160
Grimwade
 
D
Jovanovic
 
JV
Hills
 
RK
, 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
, vol. 
27
 
22
(pg. 
3650
-
3658
)
161
Zhou
 
J
Zhang
 
Y
Li
 
J
, et al. 
Single-agent arsenic trioxide in the treatment of children with newly diagnosed acute promyelocytic leukemia.
Blood
2010
, vol. 
115
 
9
(pg. 
1697
-
1702
)
162
Estey
 
E
Garcia-Manero
 
G
Ferrajoli
 
A
, et al. 
Use of all-trans retinoic acid plus arsenic trioxide as an alternative to chemotherapy in untreated acute promyelocytic leukemia.
Blood
2006
, vol. 
107
 
9
(pg. 
3469
-
3473
)
163
Larson
 
RA
Etiology and management of therapy-related myeloid leukemia.
Hematology Am Soc Hematol Educ Program
2007
, vol. 
2007
 (pg. 
453
-
459
)
164
Pui
 
CH
Raimondi
 
SC
Head
 
DR
, et al. 
Characterization of childhood acute leukemia with multiple myeloid and lymphoid markers at diagnosis and at relapse.
Blood
1991
, vol. 
78
 
5
(pg. 
1327
-
1337
)
165
Aguilera
 
DG
Vaklavas
 
C
Tsimberidou
 
AM
Wen
 
S
Medeiros
 
LJ
Corey
 
SJ
Pediatric therapy-related myelodysplastic syndrome/acute myeloid leukemia: the M. D. Anderson Cancer Center experience.
J Pediatr Hematol Oncol
2009
, vol. 
31
 
11
(pg. 
803
-
811
)
166
Kaspers
 
GJ
Creutzig
 
U
Pediatric acute myeloid leukemia: international progress and future directions.
Leukemia
2005
, vol. 
19
 
12
(pg. 
2025
-
2029
)
167
Sander
 
A
Zimmermann
 
M
Dworzak
 
M
, et al. 
Consequent and intensified relapse therapy improved survival in pediatric AML: results of relapse treatment in 379 patients of three consecutive AML-BFM trials.
Leukemia
2010
, vol. 
24
 
8
(pg. 
1422
-
1428
)
168
Stahnke
 
K
Boos
 
J
Bender-Gotze
 
C
Ritter
 
J
Zimmermann
 
M
Creutzig
 
U
Duration of first remission predicts remission rates and long-term survival in children with relapsed acute myelogenous leukemia.
Leukemia
1998
, vol. 
12
 
10
(pg. 
1534
-
1538
)
169
Kaspers
 
GJL
Zimmermann
 
M
Reinhardt
 
D
, et al. 
Addition of liposomal daunorubicin (DaunoXome) to FLAG significantly improves treatment response in pediatric relapsed AML: final results from the International Randomised Phase III Study Relapsed AML 2001/01 [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
 
Abstract 18
170
Abrahamsson
 
J
Clausen
 
N
Gustafsson
 
G
, et al. 
Improved outcome after relapse in children with acute myeloid leukaemia.
Br J Haematol
2007
, vol. 
136
 
2
(pg. 
229
-
236
)
171
Webb
 
DK
Wheatley
 
K
Harrison
 
G
Stevens
 
RF
Hann
 
IM
Outcome for children with relapsed acute myeloid leukaemia following initial therapy in the Medical Research Council (MRC) AML 10 trial: MRC Childhood Leukaemia Working Party.
Leukemia
1999
, vol. 
13
 
1
(pg. 
25
-
31
)
172
Gandhi
 
V
Estey
 
E
Keating
 
MJ
Plunkett
 
W
Fludarabine potentiates metabolism of cytarabine in patients with acute myelogenous leukemia during therapy.
J Clin Oncol
1993
, vol. 
11
 
1
(pg. 
116
-
124
)
173
Goemans
 
BF
Tamminga
 
RY
Corbijn
 
CM
Hahlen
 
K
Kaspers
 
GJ
Outcome for children with relapsed acute myeloid leukemia in The Netherlands following initial treatment between 1980 and 1998: survival after chemotherapy only?
Haematologica
2008
, vol. 
93
 
9
(pg. 
1418
-
1420
)
174
Burnett
 
AK
Hills
 
RK
Milligan
 
D
, et al. 
Identification of patients with acute myeloblastic leukemia who benefit from the addition of gemtuzumab ozogamicin: results of the MRC AML15 trial.
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
369
-
377
)
175
Zwaan
 
CM
Reinhardt
 
D
Zimmerman
 
M
, et al. 
Salvage treatment for children with refractory first or second relapse of acute myeloid leukaemia with gemtuzumab ozogamicin: results of a phase II study.
Br J Haematol
2010
, vol. 
148
 
5
(pg. 
768
-
776
)
176
Jeha
 
S
Razzouk
 
B
Rytting
 
M
, et al. 
Phase II study of clofarabine in pediatric patients with refractory or relapsed acute myeloid leukemia.
J Clin Oncol
2009
, vol. 
27
 
26
(pg. 
4392
-
4397
)
177
Hijiya
 
N
Gaynon
 
P
Barry
 
E
, et al. 
A multi-center phase I study of clofarabine, etoposide and cyclophosphamide in combination in pediatric patients with refractory or relapsed acute leukemia.
Leukemia
2009
, vol. 
23
 
12
(pg. 
2259
-
2264
)
178
Sievers
 
EL
Larson
 
RA
Stadtmauer
 
EA
, et al. 
Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse.
J Clin Oncol
2001
, vol. 
19
 
13
(pg. 
3244
-
3254
)
179
Zwaan
 
CM
Reinhardt
 
D
Corbacioglu
 
S
, et al. 
Gemtuzumab ozogamicin: first clinical experiences in children with relapsed/refractory acute myeloid leukemia treated on compassionate-use basis.
Blood
2003
, vol. 
101
 
10
(pg. 
3868
-
3871
)
180
Delaunay
 
J
Recher
 
C
Pigneux
 
A
, et al. 
Addition of gemtuzumab ozogamycin to chemotherapy improves event-free survival but not overall survival of AML patients with intermediate cytogenetics not eligible for allogeneic rransplantation: results of the GOELAMS AML 2006 IR Study [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
 
Abstract 79
181
Brethon
 
B
Yakouben
 
K
Oudot
 
C
, et al. 
Efficacy of fractionated gemtuzumab ozogamicin combined with cytarabine in advanced childhood myeloid leukaemia.
Br J Haematol
2008
, vol. 
143
 
4
(pg. 
541
-
547
)
182
Reinhardt
 
D
Diekamp
 
S
Fleischhack
 
G
, et al. 
Gemtuzumab ozogamicin (Mylotarg) in children with refractory or relapsed acute myeloid leukemia.
Onkologie
2004
, vol. 
27
 
3
(pg. 
269
-
272
)
183
Burnett
 
AK
Hills
 
RK
Hunter
 
AE
, et al. 
The addition of gemtuzumab ozogamicin to intensive chemotherapy in older patients with AML produces a significant improvement in overall survival: results of the UK NCRI AML16 Randomized Trial [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
 
Abstract 582
184
Castaigne
 
S
Pautas
 
C
Terre
 
C
, et al. 
Fractionated doses of gemtuzumab ozogamicin (GO) combined to standard chemotherapy (CT) improve event-free and overall survival in newly-diagnosed de novo AML patients aged 50-70 years old: a prospective randomized phase 3 trial from the Acute Leukemia French Association (ALFA) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
 
Abstract 6
185
Castaigne
 
S
Pautas
 
C
Terre
 
C
, et al. 
Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study.
Lancet
2012
, vol. 
379
 
9825
(pg. 
1508
-
1516
)
186
Cooper
 
TM
Franklin
 
J
Gerbing
 
RB
, et al. 
AAML03P1, a pilot study of the safety of gemtuzumab ozogamicin in combination with chemotherapy for newly diagnosed childhood acute myeloid leukemia: a report from the Children's Oncology Group.
Cancer
2012
, vol. 
118
 
3
(pg. 
761
-
769
)
187
Fathi
 
A
Levis
 
M
FLT3 inhibitors: a story of the old and the new.
Curr Opin Hematol
2011
, vol. 
18
 
2
(pg. 
71
-
76
)
188
Inaba
 
H
Rubnitz
 
JE
Coustan-Smith
 
E
, 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
, vol. 
29
 
24
(pg. 
3293
-
3300
)
189
Creutzig
 
U
Ritter
 
J
Budde
 
M
Sutor
 
A
Schellong
 
G
Early deaths due to hemorrhage and leukostasis in childhood acute myelogenous leukemia: associations with hyperleukocytosis and acute monocytic leukemia.
Cancer
1987
, vol. 
60
 
12
(pg. 
3071
-
3079
)
190
Inaba
 
H
Fan
 
Y
Pounds
 
S
, et al. 
Clinical and biologic features and treatment outcome of children with newly diagnosed acute myeloid leukemia and hyperleukocytosis.
Cancer
2008
, vol. 
113
 
3
(pg. 
522
-
529
)
191
Malaguarnera
 
G
Giordano
 
M
Malaguarnera
 
M
Rasburicase for the treatment of tumor lysis in hematological malignancies.
Expert Rev Hematol
2012
, vol. 
5
 
1
(pg. 
27
-
38
)
192
Cornely
 
OA
Maertens
 
J
Winston
 
DJ
, et al. 
Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia.
N Engl J Med
2007
, vol. 
356
 
4
(pg. 
348
-
359
)
193
Lehrnbecher
 
T
Groll
 
AH
Invasive fungal infections in the pediatric population.
Expert Rev Anti Infect Ther
2011
, vol. 
9
 
3
(pg. 
275
-
278
)
194
Hughes
 
WT
Kuhn
 
S
Chaudhary
 
S
, et al. 
Successful chemoprophylaxis for Pneumocystis carinii pneumonitis.
N Engl J Med
1977
, vol. 
297
 
26
(pg. 
1419
-
1426
)
195
Hughes
 
WT
Rivera
 
GK
Schell
 
MJ
Thornton
 
D
Lott
 
L
Successful intermittent chemoprophylaxis for Pneumocystis carinii pneumonitis.
N Engl J Med
1987
, vol. 
316
 
26
(pg. 
1627
-
1632
)
196
Sung
 
L
Nathan
 
PC
Alibhai
 
SM
Tomlinson
 
GA
Beyene
 
J
Meta-analysis: effect of prophylactic hematopoietic colony-stimulating factors on mortality and outcomes of infection.
Ann Intern Med
2007
, vol. 
147
 
6
(pg. 
400
-
411
)
197
Gamis
 
AS
Howells
 
WB
DeSwarte-Wallace
 
J
Feusner
 
JH
Buckley
 
JD
Woods
 
WG
Alpha hemolytic streptococcal infection during intensive treatment for acute myeloid leukemia: a report from the Children's Cancer Group Study CCG-2891.
J Clin Oncol
2000
, vol. 
18
 
9
(pg. 
1845
-
1855
)
198
Kurt
 
B
Flynn
 
P
Shenep
 
JL
, et al. 
Prophylactic antibiotics reduce morbidity due to septicemia during intensive treatment for pediatric acute myeloid leukemia.
Cancer
2008
, vol. 
113
 
2
(pg. 
376
-
382
)
199
Gafter-Gvili
 
A
Fraser
 
A
Paul
 
M
, et al. 
Antibiotic prophylaxis for bacterial infections in afebrile neutropenic patients following chemotherapy.
Cochrane Database Syst Rev
2012
, vol. 
1
 pg. 
CD004386
 
200
Freifeld
 
AG
Bow
 
EJ
Sepkowitz
 
KA
, et al. 
Clinical practice guideline for the use of antimicrobial agents in neutropenic patients with cancer: 2010 update by the Infectious Diseases Society of America.
Clin Infect Dis
2011
, vol. 
52
 
4
(pg. 
e56
-
e93
)
201
van de Wetering
 
MD
de Witte
 
MA
Kremer
 
LC
Offringa
 
M
Scholten
 
RJ
Caron
 
HN
Efficacy of oral prophylactic antibiotics in neutropenic afebrile oncology patients: a systematic review of randomised controlled trials.
Eur J Cancer
2005
, vol. 
41
 
10
(pg. 
1372
-
1382
)
202
Gafter-Gvili
 
A
Fraser
 
A
Paul
 
M
Leibovici
 
L
Meta-analysis: antibiotic prophylaxis reduces mortality in neutropenic patients.
Ann Intern Med
2005
, vol. 
142
 
12
(pg. 
979
-
995
)
203
Lehrnbecher
 
T
Ethier
 
MC
Zaoutis
 
T
, et al. 
International variations in infection supportive care practices for paediatric patients with acute myeloid leukaemia.
Br J Haematol
2009
, vol. 
147
 
1
(pg. 
125
-
128
)
204
Lehrnbecher
 
T
Zimmermann
 
M
Reinhardt
 
D
Dworzak
 
M
Stary
 
J
Creutzig
 
U
Prophylactic human granulocyte colony-stimulating factor after induction therapy in pediatric acute myeloid leukemia.
Blood
2007
, vol. 
109
 
3
(pg. 
936
-
943
)
205
Schaison
 
G
Eden
 
OB
Henze
 
G
, et al. 
Recommendations on the use of colony-stimulating factors in children: conclusions of a European panel.
Eur J Pediatr
1998
, vol. 
157
 
12
(pg. 
955
-
966
)
206
Dores
 
GM
Devesa
 
SS
Curtis
 
RE
Linet
 
MS
Morton
 
LM
Acute leukemia incidence and patient survival among children and adults in the United States, 2001-2007.
Blood
2012
, vol. 
119
 
1
(pg. 
34
-
43
)
207
Strahm
 
B
Nollke
 
P
Zecca
 
M
, et al. 
Hematopoietic stem cell transplantation for advanced myelodysplastic syndrome in children: results of the EWOG-MDS 98 study.
Leukemia
2011
, vol. 
25
 
3
(pg. 
455
-
462
)
208
Wandt
 
H
Schakel
 
U
Kroschinsky
 
F
, et al. 
MLD according to the WHO classification in AML has no correlation with age and no independent prognostic relevance as analyzed in 1766 patients.
Blood
2008
, vol. 
111
 
4
(pg. 
1855
-
1861
)
209
Grimwade
 
D
Walker
 
H
Oliver
 
F
, et al. 
What happens subsequently in AML when cytogenetic abnormalities persist at bone marrow harvest? Results of the 10th UK MRC AML trial. Medical Research Council Leukaemia Working Parties.
Bone Marrow Transplant
1997
, vol. 
19
 
11
(pg. 
1117
-
1123
)
210
Smith
 
ML
Hills
 
RK
Grimwade
 
D
Independent prognostic variables in acute myeloid leukaemia.
Blood Rev
2011
, vol. 
25
 
1
(pg. 
39
-
51
)
211
Dohner
 
K
Dohner
 
H
Molecular characterization of acute myeloid leukemia.
Haematologica
2008
, vol. 
93
 
7
(pg. 
976
-
982
)
212
Haferlach
 
T
Bacher
 
U
Haferlach
 
C
Kern
 
W
Schnittger
 
S
Insight into the molecular pathogenesis of myeloid malignancies.
Curr Opin Hematol
2007
, vol. 
14
 
2
(pg. 
90
-
97
)
213
Schlenk
 
RF
Ganser
 
A
Dohner
 
K
Prognostic and predictive effect of molecular and cytogenetic aberrations in acute myeloid leukemia.
ASCO Educ Book
2010
(pg. 
228
-
232
)
214
Dohner
 
H
Gaidzik
 
VI
Impact of genetic features on treatment decisions in AML.
Hematology Am Soc Hematol Educ Program
2011
, vol. 
2011
 
1
(pg. 
36
-
42
)
215
de Jonge
 
HJ
Valk
 
PJ
de Bont
 
ES
, et al. 
Prognostic impact of white blood cell count in intermediate risk acute myeloid leukemia: relevance of mutated NPM1 and FLT3-ITD.
Haematologica
2011
, vol. 
96
 
9
(pg. 
1310
-
1317
)
216
Tyner
 
JW
Walters
 
DK
Willis
 
SG
, et al. 
RNAi screening of the tyrosine kinome identifies therapeutic targets in acute myeloid leukemia.
Blood
2008
, vol. 
111
 
4
(pg. 
2238
-
2245
)
217
Shimada
 
A
Taki
 
T
Kubota
 
C
, et al. 
N822 mutation of KIT gene was frequent in pediatric acute myeloid leukemia patients with t(8;21) in Japan: a study of the Japanese childhood AML cooperative study group.
Leukemia
2007
, vol. 
21
 
10
(pg. 
2218
-
2219
)
218
Delhommeau
 
F
Dupont
 
S
Della Valle
 
V
, et al. 
Mutation in TET2 in myeloid cancers.
N Engl J Med
2009
, vol. 
360
 
22
(pg. 
2289
-
2301
)
219
Ley
 
TJ
Ding
 
L
Walter
 
MJ
, et al. 
DNMT3A mutations in acute myeloid leukemia.
N Engl J Med
2010
, vol. 
363
 
25
(pg. 
2424
-
2433
)
220
Haferlach
 
T
Schoch
 
C
Loffler
 
H
, et al. 
Morphologic dysplasia in de novo acute myeloid leukemia (AML) is related to unfavorable cytogenetics but has no independent prognostic relevance under the conditions of intensive induction therapy: results of a multiparameter analysis from the German AML Cooperative Group studies.
J Clin Oncol
2003
, vol. 
21
 
2
(pg. 
256
-
265
)
221
Weinberg
 
OK
Seetharam
 
M
Ren
 
L
, et al. 
Clinical characterization of acute myeloid leukemia with myelodysplasia-related changes as defined by the 2008 WHO classification system.
Blood
2009
, vol. 
113
 
9
(pg. 
1906
-
1908
)
222
Miesner
 
M
Haferlach
 
C
Bacher
 
U
, et al. 
Multilineage dysplasia (MLD) in acute myeloid leukemia (AML) correlates with MDS-related cytogenetic abnormalities and a prior history of MDS or MDS/MPN but has no independent prognostic relevance: a comparison of 408 cases classified as “AML not otherwise specified” (AML-NOS) or “AML with myelodysplasia-related changes” (AML-MRC).
Blood
2010
, vol. 
116
 
15
(pg. 
2742
-
2751
)
223
Klusmann
 
JH
Godinho
 
FJ
Heitmann
 
K
, et al. 
Developmental stage-specific interplay of GATA1 and IGF signaling in fetal megakaryopoiesis and leukemogenesis.
Genes Dev
2010
, vol. 
24
 
15
(pg. 
1659
-
1672
)
224
Grimwade
 
D
Walker
 
H
Oliver
 
F
, et al. 
The importance of diagnostic cytogenetics on outcome in AML: analysis of 1612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children's Leukaemia Working Parties.
Blood
1998
, vol. 
92
 
7
(pg. 
2322
-
2333
)
225
Creutzig
 
U
Kaspers
 
GJ
Revised recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia.
J Clin Oncol
2004
, vol. 
22
 
16
(pg. 
3432
-
3433
)

Author notes

*

U.C. and M.M.v.d.H.-E. contributed equally to this study.

G.J.L.K. and D.R. contributed equally to this study.

Sign in via your Institution