ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Acute myeloid leukemia in children and adolescents

Acute myeloid leukemia in children and adolescents
Literature review current through: Jan 2024.
This topic last updated: Oct 26, 2023.

INTRODUCTION — Acute leukemia is the most common cancer in children, and it corresponds to one-third of all childhood malignancies. Acute myeloid leukemia (AML) accounts for approximately 15 percent of childhood leukemia, while acute lymphoblastic leukemia/lymphoblastic lymphoma (ALL/LBL) corresponds to 80 percent of pediatric acute leukemia.

Treatment in clinical trials, risk stratification, intensification of therapy, and improved supportive care have contributed to improved survival for children and adolescents with AML, but outcomes remain inferior to those with ALL/LBL.

This topic reviews AML in children and adolescents.

Treatment of childhood ALL/LBL is discussed separately. (See "Treatment of acute lymphoblastic leukemia/lymphoma in children and adolescents".)

CLINICAL PRESENTATION — The most common presenting symptoms of AML in children and adolescents are findings associated with the leukemic burden, associated cytopenias, or disease complications. The clinical presentation of AML may be indistinguishable from that of acute lymphoblastic leukemia/lymphoblastic lymphoma.

Patients can present with fever, malaise, musculoskeletal pains, lymphadenopathy, or hepatosplenomegaly. They may have clinical manifestations of cytopenias, including anemia (eg, pallor, fatigue, dyspnea), thrombocytopenia (eg, unusual bleeding or bruising), and/or neutropenia (eg, infection).

In some children, AML is first suspected because of laboratory abnormalities, such as circulating myeloblasts, electrolyte derangements, or liver dysfunction.

Less often, children present with symptoms related to complications of AML, such as involvement of the central nervous system (eg, headache, lethargy, mental status changes, cranial nerve palsies) or other extramedullary sites. Some children have respiratory or neurologic distress due to leukostasis (extreme levels of circulating myeloblasts). Disseminated intravascular coagulation can be present, especially in patients with acute promyelocytic leukemia.

EVALUATION — Evaluation of AML in children and adolescents includes history and physical examination, laboratory tests, bone marrow examination, and other clinical studies.

Clinical/laboratory testing

Clinical – History should evaluate the child for symptoms, described above, related to tumor burden, cytopenias, and extramedullary involvement. (See 'Clinical presentation' above.)

Family history should determine if relatives have conditions consistent with a familial/inherited basis for the leukemia. Examples include other leukemias, myelodysplastic syndromes/neoplasms (MDS), or multiple cancers; suspicious abnormalities of hair, skin, nails, or organ dysfunction (eg, lung or liver); deafness; and short stature, as discussed separately. (See "Familial disorders of acute leukemia and myelodysplastic syndromes".)

Physical examination should evaluate the child for hepatosplenomegaly, bleeding/bruising, findings that may reflect an inherited/familial condition, or involvement of the central nervous system (CNS) or other extramedullary sites.

Laboratory

Hematology – Blood counts and coagulation tests:

-Complete blood count with leukocyte differential count

-Prothrombin time, activated partial thromboplastin time, fibrinogen

Chemistries – Electrolytes, glucose, kidney function, liver function tests, including lactate dehydrogenase, calcium, magnesium, phosphorus, uric acid, albumin

Other clinical studies

Lumbar puncture – All children with suspected AML should have a lumbar puncture (LP) soon after diagnosis to examine cerebrospinal fluid, which should be analyzed by cell count, protein, glucose, and cytology (examination of stained cytospin slides). (See "Lumbar puncture in children".)

Leukemic involvement of the CNS is generally defined as >5 x 106 leukocytes/L cerebrospinal fluid including blasts, in a nonbloody specimen.

Imaging – Imaging is performed, as clinically indicated, to assess leukemic involvement of extramedullary sites.

-Central nervous system – For children with unexplained neurologic abnormalities, computed tomography (CT) and/or magnetic resonance imaging (MRI) should be performed to assess the patient for mass lesions or meningeal involvement.

-Other extramedullary sites – Suspected extramedullary involvement by AML should be evaluated by CT, positron emission tomography, and/or MRI, as clinically indicated.

Pathology — Bone marrow, peripheral blood, and suspected sites of extramedullary involvement should be evaluated for blasts. Analysis includes morphology, immunophenotype, cytogenetics, and molecular features, which are essential for distinguishing AML from ALL and other disorders.

Characteristic pathologic findings in childhood AML include:

Morphology – Myeloblasts are immature cells with large nuclei, usually with prominent nucleoli, and a variable amount of pale blue cytoplasm. Myeloid blasts typically have more abundant cytoplasm than lymphoid blasts and may have granules or Auer rods (picture 1).

Immunophenotype – The immunophenotype of the blasts is determined by flow cytometry and/or immunohistochemistry.

AML blasts express myeloid markers, such as CD11b, CD34, CD33, CD45, CD64, CD65, CD117, myeloperoxidase (MPO), and lysozyme.

Cytogenetics – Cytogenetic abnormalities are found in three-quarters of pediatric AML cases [1]. Many of these abnormalities have prognostic significance, as discussed below. (See 'Cytogenetics' below.)

Although many cytogenetic abnormalities associated with pediatric AML are detected by conventional karyotype analysis, some require fluorescence in situ hybridization (FISH) or molecular techniques for detection.

In addition to conventional karyotyping, FISH should evaluate the specimen for t(8;21), inv(16), t(15;17), and 11q23 translocations. (See "Acute myeloid leukemia: Cytogenetic abnormalities", section on 'Recurrent translocations'.)

Mutation analysis Pediatric AML is genetically heterogeneous, and the molecular profiles differ from those of adult AML. Certain mutations or genetic profiles have prognostic or therapeutic implications, as discussed below. (See 'Molecular features' below.)

Deoxyribonucleic acid (DNA) sequencing and/or polymerase chain reaction (PCR) should be used to identify mutations of NPM1, CEBPA, FLT3, KIT, WT1, and/or cryptic fusions that are not detected by conventional cytogenetics [2,3]. (See "Acute myeloid leukemia: Molecular genetics", section on 'Gene mutations'.)

DIAGNOSIS AND CLASSIFICATION — AML should be considered in a child or adolescent with unexplained cytopenias or associated symptoms, organomegaly, or blasts in peripheral blood. (See 'Clinical presentation' above.)

Diagnosis — Diagnosis of AML requires pathologic confirmation in bone marrow, with or without analysis of blood and/or sites of extramedullary involvement.

Diagnosis is based on morphology (microscopy) and immunophenotyping that reveal the myeloid lineage of blasts, along with cytogenetic and molecular analysis. Details of the diagnostic criteria for AML are presented separately. (See "Clinical manifestations, pathologic features, and diagnosis of acute myeloid leukemia", section on 'Diagnosis'.)

One of the following systems should be used to diagnose and classify AML:

International Consensus Classification (ICC) of myeloid neoplasms and acute leukemias [4]

The 5th edition of the World Health Organization (WHO5) of hematolymphoid tumors: myeloid and histiocytic/dendritic neoplasms [5]

Both ICC and WHO5 classify hematopoietic neoplasms by integrating morphology (eg, cytology, histology, tissue architecture) and cellular lineage (by immunophenotype) with cytogenetic and molecular features. Use of either ICC or WHO5 is acceptable and should replace outdated classification systems, such as the FAB (French-American-British) classification system [6] or the revised 4th edition of the WHO classification system [7]. Details of the ICC and WHO5 classification systems are presented separately. (See "Classification of hematopoietic neoplasms".)

Recognition of inherited/familial AML has important implications for management. Familial AML is demonstrated by personal and family history and genetic testing of the blasts and nonhematopoietic tissue to demonstrate the presence of an implicated gene variant/mutation in the germline. (See "Familial disorders of acute leukemia and myelodysplastic syndromes".)

Differential diagnosis — AML must be distinguished from other types of leukemia and hematologic malignancies with blood/marrow involvement and from other conditions associated with cytopenias and/or circulating immature cells.

Acute lymphoblastic leukemia – Acute lymphoblastic leukemia (ALL) manifests similar clinical findings, but it is considerably more common than AML in children.

Clinical findings are like those of AML, but ALL is more likely to present with extramedullary disease, such as a mediastinal mass. ALL blasts are generally smaller, have less conspicuous nucleoli, less cytoplasm, and lack intracytoplasmic granules and Auer rods.

Immunophenotyping (eg, using flow cytometry) is essential for distinguishing the lymphoid blasts of ALL from the myeloid blasts of AML. Diagnosis of ALL in children is discussed separately. (See "Overview of the clinical presentation and diagnosis of acute lymphoblastic leukemia/lymphoma in children".)

Other acute leukemias – A variety of childhood leukemias share clinical or morphologic similarities with AML. Examples include:

Mixed phenotype acute leukemia and acute undifferentiated leukemia are distinguished from AML by immunophenotype, as discussed separately. (See "Mixed phenotype acute leukemia".)

Juvenile myelomonocytic leukemia can present with organomegaly, but the malignant cells are not as immature and exhibit features of the monocytic lineage. (See "Juvenile myelomonocytic leukemia".)

Chronic myeloid leukemia – Accelerated phase or blast crisis of chronic myeloid leukemia (CML) typically manifests splenomegaly and/or constitutional symptoms, with an expanded population of circulating myeloid cells at various stages of differentiation. However, malignant cells of CML exhibit the characteristic t(9;22) chromosomal rearrangement (Philadelphia chromosome) and BCR::ABL1 rearrangement, which distinguish them from AML blasts. (See "Clinical manifestations and diagnosis of chronic myeloid leukemia".)

Myelodysplastic syndromes/neoplasms – Myelodysplastic syndromes/neoplasms (MDS) and AML are considered by many experts to be components of a spectrum of myeloid malignancies with related clinical features and certain aspects of natural history. MDS may present with cytopenias with circulating myeloid cells with aberrant morphology, including myeloblasts. MDS is distinguished from AML by fewer blasts in blood and marrow and by genetic abnormalities that differ from, but may overlap with, findings of AML.

For cases with 10 to 20 percent myeloid blasts, the ICC does not distinguish between MDS and AML; such cases are labeled MDS/AML [4]. The WHO5 describes such cases as MDS with increased blasts [5]. Details of diagnosis and classification of MDS and AML are discussed separately. (See "Classification of hematopoietic neoplasms", section on 'Myeloid neoplasms'.)

Leukemoid reaction – A leukemoid reaction describes a high leukocyte count (eg, ≥50,000/microL) with neutrophilia and prominent left shift, usually in response to infection. However, myeloblasts and other very immature cells are scant or absent, and a leukemoid reaction can be distinguished by morphology. Bone marrow examination and/or cytogenetic testing is rarely needed to distinguish a leukemoid reaction from AML.

Aplastic anemia – Aplastic anemia is a bone marrow failure state that presents with variable levels of pancytopenia and related symptoms. Aplastic anemia can be caused by drugs, infections, inherited conditions, and acquired/autoimmune damage to hematopoietic stem and progenitor cells. Bone marrow examination demonstrates profound hypoplasia of hematopoietic elements, and the marrow space is composed primarily of fat cells and marrow stroma. (See "Aplastic anemia: Pathogenesis, clinical manifestations, and diagnosis".)

Evaluation and diagnosis of various forms of familial/inherited aplastic anemia are discussed separately. (See "Clinical manifestations and diagnosis of Fanconi anemia" and "Shwachman-Diamond syndrome" and "Dyskeratosis congenita and other telomere biology disorders".)

For children with Down syndrome (DS)/trisomy 21, it is important to distinguish transient abnormal myelopoiesis (TAM) and myeloid leukemia associated with DS (ML-DS) from AML. TAM and ML-DS have very different natural histories and require management that is distinct from that of AML. (See "Transient abnormal myelopoiesis (TAM) of Down syndrome (DS)" and "Myeloid leukemia associated with Down syndrome (ML-DS)".)

Classification — Cases of pediatric AML should be classified using either the ICC or the WHO5 system. Both systems assign AML subtypes according to morphologic, cytogenetic, and molecular features.

Defining genetic abnormalities – ICC and WHO5 apply similar diagnostic criteria and labels for cases of AML with defining cytogenetic and molecular findings.

Details of classification of AML subtypes with defining genetic abnormalities are presented separately. (See "Acute myeloid leukemia: Classification", section on 'AML with defining genetic abnormalities'.)

Other cases of AML – For cases without AML-defining genetic abnormalities, ICC and WHO5 differ in the labels, diagnostic criteria, and assignment to disease categories. Differences between ICC and WHO5 that affect classification of pediatric AML include:

AML without a defining genetic abnormality – ICC defines many cases without defining genetic abnormalities as AML, not otherwise specified [4], while WHO5 categorizes them according to the degree of blast differentiation [5].

AML associated with germline or inherited gene variants – AML associated with germline or inherited gene variants is described differently:

-International Consensus Classification – Cases of AML in association with a germline predisposition constitute a separate category, Hematologic neoplasms with germline predisposition; cases are further defined according to association with abnormalities in multiple organ systems, a constitutional platelet disorder, or no abnormalities in multiple organ systems.

-The 5th edition of the World Health Organization – Cases of AML in association with a germline predisposition are included in the category, Secondary myeloid neoplasms, which also includes therapy-related AML. Cases are assigned subtypes according to associated findings, as described with ICC classification above.

Therapy-related AML – For AML that arises in a patient with prior exposure to cytotoxic agents, ionizing radiation, or immune interventions, "Therapy-related" is added as a qualifier following the diagnosis of AML in ICC. In WHO5, such cases are included in a category of Secondary myeloid neoplasms (which also includes AML in association with a germline predisposition).

Myeloid sarcoma – This is a distinct subtype of AML in the ICC system, whereas it is described as a tissue-based manifestation of AML in WHO5.

Other details of differences between the ICC and WHO5 classification systems are presented separately. (See "Acute myeloid leukemia: Classification", section on 'Other subtypes'.)

PROGNOSTIC FACTORS — Prognosis in pediatric AML is informed by cytogenetic and molecular features of the blasts. For children whose AML has no prognostically important genetic features, outcomes are associated with the response to induction therapy. These prognostic features are important components of risk stratification for treatment of pediatric AML, as discussed below. (See 'Risk stratification' below.)

Cytogenetics

Favorable – AML with favorable cytogenetics accounts for approximately 20 to 30 percent of pediatric AML. Favorable cytogenetic features are:

t(8;21)(q22;q22); RUNX1::RUNX1T1

Core-binding factor AML: inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB::MYH11

Treatment on contemporary protocols with chemotherapy alone results in approximately 80 percent long-term overall survival (OS) and 70 percent relapse-free survival (RFS) [8-10].

High risk – Children with adverse cytogenetic features account for approximately 15 percent of pediatric AML [8].

Patients with high-risk cytogenetic features include those that harbor any of the following cytogenetic abnormalities (and lack any of the favorable features described above) [3,11-13]:

Monosomy 7

Monosomy 5 or del(5q)

Abnormalities of 3q: inv(3)(q21q26.2) and t(3;3)(q21;q26.2); RPN1::EVI1

t(6;9)(p23;q34)

Complex karyotype (≥3 cytogenetic abnormalities)

Children and adolescents harboring these unfavorable features have <50 percent long-term OS; some features are associated with <20 percent OS [3]. Trisomies are common events in pediatric AML (often involving chromosome 8, 19, or 21) and can occur alone or in conjunction with other karyotypic abnormalities.

Cytogenetic abnormalities associated with AML are discussed in more detail separately. (See "Acute myeloid leukemia: Cytogenetic abnormalities".)

Molecular features — Certain mutations are predictive of relapse and associated with prognosis in pediatric AML.

Mutation testing is an essential component of the diagnostic/classification workup and subsequent risk stratification. Next generation sequencing (NGS) and other emerging technologies can detect cryptic fusion genes, copy number alternations (focal gains or losses of genomic regions), loss of heterozygosity, and AML subtypes [14-16].

Molecular features that are associated with prognosis in pediatric AML include:

Low risk

CEBPA – Mutations involving CEBPA occur in approximately 5 percent of pediatric AML. Children with CEBPA mutations affecting the bZip domain have a more favorable prognosis, with approximately 70 percent long-term OS following treatment with chemotherapy alone [17,18]. (See "Acute myeloid leukemia: Risk factors and prognosis".)

NPM1 – Mutations involving NPM1 are present in approximately 8 to 10 percent of pediatric AML [19]. Only cases of AML with mutated NPM1 without associated FLT3-ITD (internal tandem repeat) are associated with a favorable prognosis; in such patients, long-term OS is approximately 70 percent with chemotherapy alone. (See "Acute myeloid leukemia: Risk factors and prognosis" and "Acute myeloid leukemia: Molecular genetics", section on 'NPM1 mutations'.)

High risk

FLT3 mutation – Activating mutations in the FLT3 gene are among the most common recurring somatic mutations in AML. Of these, FLT3-ITD is the most common, and it occurs in 10 to 15 percent of pediatric AML [20,21]. The prevalence of FLT3-ITD increases with age, occurring in 15 to 25 percent of adolescents and young adults. Point mutations of FLT3 (ie, activating loop mutations) occur in 5 to 7 percent of childhood AML and have no clear prognostic impact [22]. (See "Acute myeloid leukemia: Molecular genetics", section on 'FLT3'.)

Patients with mutations involving FLT3-ITD with a high allelic ratio (HAR; of mutant to wild-type FLT3) have a poor prognosis. Cooperative groups use different thresholds, but it is generally accepted that an allelic ratio greater than 0.4 to 0.5 is a marker of poor prognosis [22,23]. Patients with HAR FLT3-ITD are more likely to be refractory to induction chemotherapy and are at a greater risk for relapse [20]. Children with HAR FLT3-ITD mutations have a 20 to 30 percent OS with contemporary chemotherapy regimens alone, but the OS is 50 to 60 percent when FLT3 inhibitors and/or consolidation allogeneic hematopoietic cell transplantation (HCT) are employed [22,23]. (See 'Postremission management' below.)

KMT2A rearrangements – Translocations involving KMT2A (previously called MLL) at the 11q23 locus are present in 15 to 20 percent of pediatric AML cases. Rearrangements involving KMT2A are most prevalent in young children, especially those <2 years [24].

There are many fusion partners of KMT2A, and prognosis may vary according to the fusion partner. Chromosomal rearrangements, including t(6;11)(q27;q23), t(10;11)(p11.2;q23.3), t(10;11)(p12;q23.3), and t(11;19)(q23;p13.3) have been associated with a poor prognosis [13,25].

The most common rearrangement in pediatric AML involving KMT2A is t(9;11)(p22;q23), which accounts for one-half of cases with KMT2A abnormalities, but it is not associated with adverse outcomes [25]. The t(1;11)(q21;23) rearrangement has been associated with a more favorable outcome relative to other KMT2A fusions [13,25].

NUP abnormalities

-t(6;9)(p23;q34)/DEK::NUP214.

-NUP98 fusions – NUP98::NSD1 fusions, which commonly occur with FLT3-ITD mutations, are associated with a poor prognosis in children and adults [13,26-28].

CBFA2T3A::GLIS2

Other recurring mutations have been identified in pediatric AML (eg, WT1, KIT, NRAS, and KRAS), but prognostic significance has not been confirmed. Their association of these mutations with additional cytogenetic and molecular aberrations suggests they may serve as cooperating mutations in leukemogenesis [29-32]. (See "Acute myeloid leukemia: Molecular genetics", section on 'Gene mutations' and "Acute myeloid leukemia: Risk factors and prognosis".)

Treatment response — Children with AML that does not harbor prognostically significant cytogenetic or molecular abnormalities are classified as intermediate risk. For these patients, the most important prognostic factor is the response to induction therapy, including the assessment of measurable residual disease (MRD) [33-35]:

Measurable residual disease negative – Children with intermediate-risk AML with negative MRD (eg, <0.1 percent by flow cytometry) experience approximately 65 percent RFS [34].

Measurable residual disease positive – Children with detectable MRD following induction have an inferior prognosis, with approximately 60 percent RFS [34].

The prognostic significance of persistent MRD for children with favorable or unfavorable cytogenetic or molecular features is less certain [34,36].

Detection of MRD by the expression of leukemia-associated genes or disease-specific mutations is not routinely used but can provide sensitive detection of a low level of residual leukemia and may inform prognostic and therapeutic decisions [37,38]. (See "Acute myeloid leukemia: Induction therapy in medically fit adults", section on 'Remission criteria'.)

RISK STRATIFICATION — Risk in cases of pediatric AML is based on prognostic factors, including cytogenetic/molecular features and/or response to induction chemotherapy, as described above. (See 'Prognostic factors' above.)

There is general agreement about what constitutes low-risk genetic features in pediatric AML. However, cooperative groups assign cases to high-risk disease using different genetic features and/or definitions for treatment response.

Favorable

t(8;21)(q22;q22); RUNX1::RUNX1T1

inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB::MYH11

Mutated NPM1 without FLT3-ITD (internal tandem duplication; normal karyotype)

Mutated bZip CEBPA (normal karyotype)

Intermediate – Cases that do not harbor cytogenetic or molecular abnormalities that classify them as favorable or adverse risk are categorized as intermediate risk.

For cases of intermediate risk, prognosis is primarily driven by response to induction therapy. (See 'Treatment response' above.)

Adverse

Cytogenetic features (see 'Cytogenetics' above)

-t(6;9)(p23;q34)/DEK::NUP214

-Abnormalities of 3q

-Monosomy 5 or del(5q)

-Monosomy 7

-Complex karyotype

Molecular features (see 'Molecular features' above)

-FLT3-ITD

-NUP98 fusions

-Certain KMT2A rearrangements

-CBFA2T3A::GLIS2

Immunophenotype – RAM immunophenotype (CD56 high, dim or negative CD45 and CD38, and negative HLA-DR expression) [15]

TREATMENT — Children and adolescents with AML should be treated in the context of a current clinical protocol, whenever possible. When that is not possible, treatment should adhere to a contemporary pediatric AML protocol in consultation with experts in the management of pediatric leukemia.

Our approach to the treatment of pediatric AML is consistent with recommendations from international expert panels [39-41].

Information and instructions for referring a patient to an appropriate research center can be obtained from the United States National Institutes of Health.

Induction therapy — Induction therapy with an anthracycline (or mitoxantrone) plus cytarabine is standard therapy in children and adolescents with AML. The doses and schedule of administration and the decision to add a third agent should be guided by the chosen protocol.

Induction therapy is intensive chemotherapy that seeks to achieve a complete remission (CR). Although achieving CR represents the reduction of the leukemic burden by orders of magnitude, it is generally not sufficient to cure AML in children and adolescents. For most patients, the achievement of CR is followed by consolidation therapy, as described below. (See 'Postremission management' below.)

Administration – Standard induction chemotherapy comprises an anthracycline (eg, daunorubicin, idarubicin) or mitoxantrone plus extended exposure to cytarabine. This generally involves two courses of induction chemotherapy. In some protocols, a third drug is included in induction therapy; examples of third agents include etoposide, 6-thioguanine, gemtuzumab ozogamicin (CD33-directed immunoconjugate), or a kinase inhibitor.

For patients with FLT3-ITD (internal tandem repeat) or other FLT3 mutations, a kinase inhibitor, such as sorafenib, may be added to induction therapy [42,43]. Midostaurin, sorafenib, quizartinib, and gilteritinib are approved by the US Food and Drug Administration (FDA) for the treatment of adults with FLT3-mutated AML. Although various kinase inhibitors are undergoing evaluation in children, none is currently approved for treatment in children. Treatment with a kinase inhibitor is typically continued through postremission management.

Adverse effects – Toxicity of induction therapy is substantial. Prolonged and profound cytopenias are experienced by all patients and may be complicated by infections from neutropenia, bleeding from thrombocytopenia, and anemia that requires transfusion support.

Monitoring of the child during induction therapy and supportive/adjunctive care for treatment-related adverse effects (AEs) are discussed below. (See 'Adjunctive/supportive care' below.)

Response assessment – Bone marrow examination is repeated as blood counts recover (typically ≥4 weeks) to assess treatment response and detection of measurable residual disease (MRD).

Remission criteria and response to therapy are described separately. (See "Acute myeloid leukemia: Induction therapy in medically fit adults", section on 'Remission status'.)

Patients with CR proceed to postremission management, as described below. (See 'Postremission management' below.)

Patients who do not achieve CR are managed as refractory AML. (See 'Relapsed/refractory AML' below.)

Outcomes – Most contemporary pediatric AML induction regimens are associated with CR in approximately three-quarters of children and adolescents, but the rate may be affected by cytogenetic and molecular features.

Anthracycline – No specific anthracycline/anthracenedione is clearly superior in this setting [44].

Daunorubicin, idarubicin, and mitoxantrone have been used for induction therapy for pediatric AML. No specific agent has emerged as clearly superior, based on studies by the Berlin-Frankfurt-Munster (BFM) group, Medical Research Council (MRC), EORTC (European Organisation for Research and Treatment of Cancer), and LAME (Leucemie Aique Myeloide Enfant) [8,45-47].

Children are generally at greater risk for anthracycline-induced cardiotoxicity than adults [48], but the risk of cardiac toxicity at increasing cumulative doses of anthracyclines in children is not well defined.

Cytarabine – No specific cytarabine regimen is proven superior in children.

Studies that used various cytarabine regimens have provided mixed results [49-51], but it is not possible to draw a firm conclusion about the benefit of high-dose versus low-dose cytarabine because of other protocol differences.

Third agent – There is no conclusive evidence of a survival benefit from the addition of a third agent to induction therapy for pediatric AML.

-A trial that randomly assigned 149 children (1 to 18 years) to two cycles of daunorubicin plus cytarabine, with or without etoposide, reported no difference in five-year overall survival (OS), five-year event-free survival, or toxicity [52].

-In a phase 3 trial for pediatric AML, the addition of gemtuzumab ozogamicin was associated with a lower rate of relapse, but there was no difference in OS compared with chemotherapy only [9].

Gemtuzumab ozogamicin is approved by the FDA for the treatment of newly diagnosed AML in children.

Central nervous system management — For all children and adolescents with AML, we suggest the administration of intrathecal (IT) chemotherapy as either prophylaxis or treatment based on neurologic evaluation that includes an initial diagnostic lumbar puncture (LP). Central nervous system (CNS)-directed therapy is generally continued through postremission management.

All patients should undergo an initial diagnostic LP to identify leukemic involvement of the CNS. It is acceptable to delay the initial LP for several days after initiation of therapy to reduce potential leukemic contamination of the CNS and to lessen the risk of bleeding, especially in patients with a coagulopathy at diagnosis. Children with unexplained neurologic abnormalities should also undergo imaging, as discussed above. (See 'Clinical/laboratory testing' above.)

CNS involvement is defined by cytologic confirmation of leukemia cells in the cerebrospinal fluid, clinical signs of CNS leukemia (eg, facial nerve palsy, brain/eye involvement), or a tumor mass/meningeal involvement detected by imaging.

All children and adolescents with AML receive CNS-directed therapy, as follows.

Prophylaxis – Patients with no evidence of CNS involvement receive CNS prophylaxis, but the method varies between protocols.

Prophylaxis reduces the risk of CNS relapse. In general, either single-agent IT cytarabine or triple IT therapy (cytarabine, methotrexate, hydrocortisone) is given. Most regimens provide at least one dose of prophylaxis with each course of therapy, but the optimal number of prophylactic IT treatments is uncertain.

When CNS prophylaxis is not used, CNS involvement at relapse may be as high as 20 percent [1] compared with <10 percent of patients who received prophylaxis [53].

Cranial radiation therapy (RT) should not be used for CNS prophylaxis in children and adolescents because of an increased risk for late AEs, including cognitive deficits, endocrine deficits, and secondary malignancies [54-56], as described below. (See 'Late effects' below.)

Treatment of central nervous system involvement – Patients with CNS involvement require augmented CNS-directed therapy, but no studies have defined an optimal method or established a consensus.

Management of CNS disease should be guided by the chosen treatment regimen. Most approaches deliver frequent IT chemotherapy combined with intensive systemic chemotherapy (eg, high-dose cytarabine [HiDAC]) [57,58]. Avoidance of cranial RT should be considered to lessen late AEs.

CNS involvement is generally found in 5 to 10 percent of children with AML at diagnosis [57], but some studies have estimated the risk in newly diagnosed pediatric AML to be as high as 30 percent [59]. Risk factors associated with CNS involvement include age <2 years; high white blood cell (WBC) count at presentation; hepatosplenomegaly; and t(8;21), inv(16), or t(16;16) [57,58].

CNS involvement at diagnosis is not an adverse prognostic factor for children with AML, and it is not associated with inferior OS when it is treated with intensified CNS-directed therapy [57].

Postremission management — For patients who achieve CR with induction therapy, postremission management is given to deepen the remission, reduce relapses, and achieve long-term survival and cure.

Consolidation therapy is the mainstay of postremission care; there is only limited evidence to support a role for maintenance therapy in childhood AML.

Consolidation therapy — The choice of consolidation varies with prognostic features and details of the chosen protocol. Risk stratification for children with AML is discussed above. (See 'Risk stratification' above.)

Favorable risk – For children with favorable prognosis AML, we suggest HiDAC-based consolidation chemotherapy rather than allogeneic hematopoietic cell transplantation (HCT), observation, or autologous HCT.

At least one-third of children who achieve first CR (CR1) will relapse without consolidation therapy, and there is no proven benefit for autologous HCT. Most experts agree that children with favorable features should receive consolidation chemotherapy rather than HCT in CR1 because transplant-associated AEs outweigh the good prognosis for these children.

Intermediate/higher risk – HiDAC or allogeneic HCT are reasonable options for children with intermediate-risk or high-risk features.

There is no consensus about which children should receive consolidation chemotherapy versus allogeneic HCT [60,61]. The choice is guided by prognostic risk factors (ie, genetic features of the blasts and/or response to induction therapy) and is defined by the chosen treatment protocol. The decision to choose allogeneic HCT must weigh the reduced rate of relapse against morbidity and mortality associated with transplantation.

No trials have randomly assigned children in CR1 to allogeneic HCT versus HiDAC-based consolidation. Most studies that compared approaches used biological randomization (ie, children who had a matched sibling donor [MSD] received transplantation, while those without an MSD received consolidation chemotherapy). Because of evolving transplantation techniques and different criteria for selecting consolidation therapy, it is difficult to compare outcomes across studies.

In AML99, 240 children with AML were stratified for postremission management based on age, WBC count at diagnosis, cytogenetics, and treatment response [62]. CR was achieved in 95 percent of children and the five-year OS was 75 percent. Children in CR1 were stratified into low-risk (112 children), intermediate-risk (92 children), and high-risk (23 children) groups. Those with low-risk AML or intermediate-risk AML without an MSD received consolidation chemotherapy, while children with intermediate-risk disease with an MSD and all children with high-risk disease were offered allogeneic HCT. There was no difference in OS for children with intermediate-risk AML who received consolidation chemotherapy versus transplantation. At five years, the OS for low-, intermediate-, and high-risk groups were 86, 72, and 57 percent, respectively.

Consolidation chemotherapy — Consolidation chemotherapy is generally based on HiDAC, with or without other agents, but the preferred dose and number of cycles of HiDAC differs between cooperative trial groups.

In AML99, for children with low-risk and intermediate-risk AML who received consolidation chemotherapy, rates of five-year OS were 86 and 72 percent, respectively [62]. The corresponding rates of five-year disease-free survival (DFS) were 71 and 60 percent. There was no difference in outcomes for children with intermediate-risk disease who received chemotherapy versus transplantation, according to the availability of an MSD. (See 'Consolidation therapy' above.)

Other studies have also shown that HiDAC consolidation therapy is associated with good outcomes for most children with AML [40,63].

No benefit was associated with autologous HCT compared with consolidation chemotherapy for children with AML who achieved remission [64-66].

Allogeneic hematopoietic cell transplantation — Allogeneic HCT is primarily offered for children who have AML with adverse pathologic features or limited treatment response. (See 'Risk stratification' above.)

There is broad support for allogeneic HCT for children with high-risk features and positive MRD after induction, but the benefit in specific genetic subsets of AML is not known. A survival benefit with allogeneic HCT must be balanced against transplantation-related mortality (TRM) and long-term AEs, including graft-versus-host disease (GVHD) [60].

Some studies suggested that allogeneic HCT in CR1 for patients with high-risk AML was associated with improved outcomes [45,60,67], while others reported no benefit compared with induction therapy alone [68,69]. Historical comparisons of outcomes are difficult because of evolving transplantation techniques and decreasing TRM.

Several studies indicated that allogeneic HCT in CR1 was associated with improved survival in children with high allelic ratio (HAR) FLT3/ITD [23,70,71].

A meta-analysis reported that allocation to HCT was associated with a reduced risk of relapse and an improved DFS and OS [72]. The graft-versus-leukemia effect is more evident in AML than in acute lymphoblastic leukemia [73].

MSDs are a preferred donor source, but matched unrelated donor grafts are currently associated with similar outcomes [61,74]. Donor selection is discussed separately. (See "Donor selection for hematopoietic cell transplantation".)

Maintenance therapy — We suggest not administering maintenance therapy, based on the absence of a proven benefit.

Results of studies that evaluated maintenance therapy have been mixed.

A trial (LAME 89/91) that randomly assigned children to 18 months of maintenance therapy (6-mercaptopurine and subcutaneous cytarabine) reported no benefit from maintenance therapy [75]. Maintenance therapy was associated with inferior OS (58 versus 81 percent) and did not reduce relapses. Furthermore, children who did not receive maintenance therapy had a higher likelihood of achieving a second CR than those who received maintenance therapy [76].

In another trial, among 215 children who were randomly assigned to 18 months of maintenance therapy (thioguanine, vincristine, cytarabine, azacitidine, and cyclophosphamide) versus no maintenance therapy, there was a trend toward inferior five-year OS in children who received maintenance therapy (46 versus 68 percent) and a lower rate of salvage from relapse if maintenance therapy had been given [77].

Maintenance therapy with sorafenib was associated with improved outcomes for children with HAR FLT3-mutated AML in a phase 1/2 study (AAML1031), but the interpretation of outcomes was confounded by a higher rate of allogeneic HCT in children who received sorafenib [42]. Multivariable analysis that accounted for rates of HCT and favorable co-occurring mutations reported that those who did not receive sorafenib had a higher rate of relapse (hazard ratio [HR] 3.03 [95% CI 1.31-7.04]).

Maintenance therapy using interleukin 2 failed to show a benefit in DFS [78].

Results from a series of studies by the BFM-AML group, which routinely administered one year of maintenance therapy to children with AML, were not better than results from other groups [8,44,46,62,79,80].

ADJUNCTIVE/SUPPORTIVE CARE — Children and adolescents with AML are at risk for complications of the underlying AML and its treatment. Supportive/adjunctive care is an essential aspect of management.

Monitoring — The child should be examined at least daily during induction therapy and consolidation therapy for infusion reactions, tumor lysis syndrome (TLS; especially those with high blast counts), nausea/vomiting, mucositis, diarrhea, infections, and other complications of therapy. Careful attention is paid to fluid balance and daily weights, and diuretics may be necessary.

It is especially important to be vigilant for complications in children. Depending on age and development, children may have a limited ability to describe their symptoms, and clinical findings of infection may be blunted or absent during periods of profound neutropenia.

Laboratory studies

Hematology – Monitor blood counts daily until recovery of white blood cell (WBC) count to ≥500/microL and platelet transfusion independence is achieved. The frequency of complete blood counts can then be reduced (eg, to every other day), as clinically appropriate.

Coagulation – Prothrombin time and partial thromboplastin time should be monitored at least weekly or more frequently, if clinically indicated. Fibrinogen should be measured early in remission induction because disseminated intravascular coagulation (DIC) can be triggered by chemotherapy. In patients with clinical or laboratory evidence of DIC, serum fibrinogen, fibrin degradation products (FDPs), and/or other tests should be monitored at least daily until DIC has resolved.

Chemistries – Chemistry profile, including electrolytes, blood urea nitrogen (BUN), creatinine, uric acid, calcium, phosphorus, and liver function tests, including lactate dehydrogenase, should be performed at least daily until the risk of TLS has passed. The frequency of testing can then be adjusted as clinically appropriate.

Close monitoring of BUN and creatinine may be required throughout the hospitalization for patients who are receiving nephrotoxic agents, such as certain antibiotics. Elevated serum lysozyme levels may lead to renal tubule damage/dysfunction in patients with monocytic/monoblastic leukemia.

Symptom management

Nausea and vomiting – Effective management of nausea and vomiting enhances patient comfort, improves oral hydration and nutritional status, and can reduce the risk of gastrointestinal (GI) bleeding or a Mallory-Weiss tear from forceful vomiting. Management of severe nausea and vomiting is described separately. (See "Approach to the infant or child with nausea and vomiting".)

Mucositis – Induction chemotherapy can damage GI epithelia and cause painful oral mucositis and diarrhea. Pain may also limit the ability to take oral fluids and nutrition, exacerbating fluid imbalances and impaired nutrition.

Breakdown of the mucosal barrier predisposes to viral, bacterial, and fungal (mostly Candida albicans) superinfection, particularly as the hematologic nadir is reached. Management of chemotherapy-induced mucositis and diarrhea is presented separately. (See "Oral toxicity associated with systemic anticancer therapy", section on 'Treatment of established mucositis'.)

Cytopenias — Profound and prolonged cytopenias are universal with intensive remission induction therapy, and transfusion of red blood cells (RBCs) and platelets should be provided as needed. Granulocyte colony-stimulating factor (G-CSF; filgrastim) and other myeloid growth factors are not routinely administered.

Transfusions – All patients should receive leukoreduced blood products to decrease febrile nonhemolytic transfusion reactions, alloimmunization, and other complications. Irradiated blood products are required for individuals who may be candidates for hematopoietic cell transplantation (HCT) to prevent transfusion-associated graft-versus-host disease (ta-GVHD). For cytomegalovirus (CMV)-negative patients who are candidates for HCT, blood products should be leukoreduced or from CMV-negative donors.

There is no consensus threshold for transfusion of RBCs or platelets. We generally transfuse RBCs when the child has anemia-associated symptoms (eg, profound fatigue, dyspnea) and aim to maintain the hemoglobin ≥7 g/dL, but this may vary with age, symptoms, comorbid conditions, and institutional approach.

We transfuse platelets prophylactically for patients with platelet counts <10,000/microL or for overt bleeding (eg, oral purpura).

Cytokines – There is no demonstrated role for prophylactic administration of G-CSF or other cytokines in this setting [81-83].

Infections/fever — Bacteremia is a leading cause of morbidity and mortality in children who are treated with intensive chemotherapy for AML. Prevention and effective management of bacterial, viral, and fungal infections is critical for optimizing outcomes in these patients.

The infectious risks are due, in part, to alterations in mucosal barriers and the prolonged absence of neutrophils. Adolescents and young adults may be at a higher risk for treatment-related toxicity compared with younger children [84,85].

Fever – Fever or other infectious findings in a neutropenic child require prompt evaluation and administration of empiric, broad-spectrum parenteral antibiotics. The choice of antibiotics should be tailored to the most likely organisms and institutional drug resistance patterns.

Initial evaluation of new or prolonged fevers in the setting of neutropenia should include CT of the chest, abdomen, pelvis, and sinuses [86]. If a patient has additional symptoms that involve extremities or the central nervous system (CNS), appropriate imaging and diagnostic testing, including examination of the cerebrospinal fluid, should be obtained. Among patients with radiographic lung findings suspicious for fungal infection, additional diagnostic testing may include bronchial alveolar lavage or biopsy of suspicious lesions [87].

Management of febrile neutropenia in children is discussed separately. (See "Fever in children with chemotherapy-induced neutropenia".)

Bacterial infections – Viridans group streptococci and gram-negative bacteria are common pathogens in this setting. Viridans group streptococci can cause severe sepsis and viridans streptococcal shock syndrome (SVS), resulting in hemodynamic instability and acute respiratory distress syndrome. The risk of infection with viridans group streptococci is more pronounced in patients treated with high-dose cytarabine (HiDAC) and in those with prolonged neutropenia [88]. Among children treated for AML, as many as 25 percent develop SVS [89]. Prompt recognition of the potential for SVS is critical as rapid progression can be fatal, and appropriate intravenous antibiotics should be started immediately [90].

Fungal infections – Severe fungal infections, including those caused by yeasts (eg, Candida spp) and invasive molds (eg, Aspergillus spp), pose a significant risk throughout AML therapy and for those undergoing allogeneic HCT [91]. New or prolonged fevers in the setting of neutropenia should prompt suspicion of fungal infection, and empiric antifungal therapy should be administered immediately for presumed or documented fungal infections; treatment should not be delayed for the completion of the diagnostic workup [87,92,93]. (See "Fever in children with chemotherapy-induced neutropenia".)

Prophylactic antimicrobials – Antibiotic prophylaxis can reduce the risk of bacteremia during intensive chemotherapy treatment, but there is no consensus for use and/or choice of prophylactic antibiotics, antifungals, and antiviral agents during remission induction therapy.

Some institutions administer prophylactic fluoroquinolones and/or antifungals, but the potential benefit varies according to the local flora and results of surveillance cultures and must be weighed against the risk of selecting for drug-resistant organisms.

Antibacterial prophylaxis – Quinolone prophylaxis has not been shown to reduce severe infectious complications (eg, sepsis) or improve survival; thus, the benefits must be balanced against potential toxicity and institutional resistance patterns to avoid the emergence of antibiotic resistance.

A phase 3 trial that evaluated levofloxacin prophylaxis versus no prophylaxis was terminated early based on efficacy of levofloxacin [81]. Among 195 children (age 6 months to 21 years) with AML or relapsed acute lymphoblastic leukemia (ALL) who were receiving two cycles of intensive chemotherapy, levofloxacin reduced bacteremia (21.9 versus 43.4 percent; risk difference, 21.6 percent [95% CI 8.8-34.4 percent]). However, for children with AML, the reduction of bacteremia was modest (23.4 versus 29.7 percent), and there was no benefit in a cohort of children undergoing allogeneic HCT. Levofloxacin was also associated with fewer episodes of fever and neutropenia (71.2 versus 82.1 percent), but there was no difference in severe infections, invasive fungal disease, Clostridioides difficile-associated diarrhea, or musculoskeletal toxic effects between trial arms.

Antifungal prophylaxis – Antifungal prophylaxis is effective at reducing invasive fungal infections and should be incorporated into the management of children and adolescents receiving therapy for AML [83,94].

Antifungals used for prophylaxis include fluconazole, which has activity against Candida spp, [95] and voriconazole, posaconazole, and itraconazole, which have activity against both Candida spp and molds, such as Aspergillus spp [96-99]. The choice of regimen depends upon the most likely pathogen, and prophylaxis regimens vary among centers.

Consultation with Infectious Disease specialists should be considered in cases of presumed or documented infections. (See "Treatment and prevention of invasive aspergillosis".)

Other infection prevention measures – Even though most infections during induction therapy are due to endogenous flora, precautions should be taken to limit exposure to exogenous pathogens.

Careful hand hygiene, prohibition against sick visitors, and other precautions are used to limit infections. Most methods to reduce the risk of infection have not been rigorously tested, but they are applied because they are relatively simple and may reduce exposure to potential pathogens.

Other complications

Hyperleukocytosis/leukostasis – WBC count >100,000/microL (>100 x 109/L) is described as hyperleukocytosis. Although the threshold is arbitrary, there is an increased risk for complications including leukostasis, DIC, and TLS.

The risk of morbidity and mortality from these complications is highest at the time of diagnosis and within days of initiating therapy [100]. In pediatric AML, higher WBC counts are more common in patients presenting with FLT3-ITD (internal tandem repeat), MLL rearrangements, myelomonocytic phenotypes, and core-binding factor-associated AML [22,100,101]. Hyperleukocytosis is more common with AML compared with acute lymphoblastic leukemia/lymphoblastic lymphoma (LBL), and complications can occur with lower WBC counts in AML [102].

Leukostasis – Hyperleukocytosis may lead to tissue damage from blast infiltration and leukostasis, resulting in hemorrhagic and thromboembolic events. Leukostasis is an oncologic emergency and can be life threatening, especially the pulmonary and neurologic complications.

The clinical presentation of leukostasis varies according to the affected organs. As an example, lung involvement may manifest as dyspnea, hypoxemia, or respiratory failure, while neurologic involvement may manifest as somnolence, coma, or focal neurologic deficits.

The most effective means of reducing the WBC count is prompt diagnosis and initiation of chemotherapy [103,104]. Leukapheresis or exchange transfusion can lower the WBC count temporarily, but these procedures pose significant risk, are temporary, and can exacerbate thrombocytopenia and the risk of DIC [105].

Diagnosis and management of leukostasis is discussed separately. (See "Hyperleukocytosis and leukostasis in hematologic malignancies".)

Tumor lysis syndrome – Supportive management should include vigorous hydration to reduce the risk of tumor lysis and decrease blood viscosity.

TLS may manifest as hyperphosphatemia, hypocalcemia (caused by precipitation of calcium phosphate), hyperuricemia, hyperkalemia, and acute renal failure. Rapid leukemic cell lysis after chemotherapy can cause overproduction and overexcretion of uric acid. Precipitation of uric acid in renal tubules can lead to oliguric or anuric renal failure. Administration of prophylactic medications to prevent complications of TLS (eg, allopurinol, rasburicase) and careful monitoring of renal function is essential. (See "Tumor lysis syndrome: Prevention and treatment".)

Disseminated intravascular coagulation – Acute DIC occurs most commonly in patients presenting with acute promyelocytic leukemia (APL), but it may also occur with AML with monoblastic differentiation or other subtypes.

If APL is suspected, all-trans retinoic acid should be started immediately (ie, even before confirmation of the diagnosis of APL). Supportive care of patients with AML-associated DIC should include standard management with appropriate blood products (eg, platelets and coagulation factors), which is essential to minimize bleeding risk. (See "Disseminated intravascular coagulation in infants and children".)

SPECIAL POPULATIONS

Down syndrome and AML — Children with Down syndrome (DS; trisomy 21) have a 10 to 20 times greater risk of developing AML compared with children without DS.

Children with myeloid leukemia associated with DS (ML-DS) cannot tolerate the toxic effects of the intensive pediatric AML regimens and must receive distinctive treatment that should be given in a center with specialized expertise. Children with DS have high rates of infection and cardiotoxicity as complications of intensive chemotherapy [106,107].

Children with DS also have specific myeloid cell disorders that must be distinguished from AML:

Transient abnormal myelopoiesis – Approximately 10 percent of infants with DS (or trisomy 21) manifest transient abnormal myelopoiesis (TAM), which presents with peripheral leukocytosis and circulating cells that resemble megakaryoblasts. Complications of TAM include pericardial and pleural effusions, coagulopathy, hydrops fetalis, hepatic dysfunction, and hepatic fibrosis [108-110].

In most affected children, TAM resolves spontaneously without any treatment [110]. Approximately one-quarter of infants with TAM require chemotherapy to ameliorate respiratory compromise, liver dysfunction, or hyperviscosity associated with hyperleukocytosis [110]. Diagnosis, management, and monitoring of infants with TAM are discussed separately. (See "Transient abnormal myelopoiesis (TAM) of Down syndrome (DS)".)

Myeloid leukemia of Down syndrome – Some children with DS later develop ML-DS, which resembles acute megakaryoblastic leukemia. ML-DS has a good prognosis, unique biologic characteristics (eg, trisomy 21 plus mutated GATA1), and requires distinctive treatment that should be administered in a center with specialized expertise.

Children with ML-DS have superior outcomes compared with children without DS who develop AML. ML-DS blasts are extremely sensitive to cytarabine and daunorubicin [111,112], and children with DS-AML can experience excellent outcomes without the intensive therapy that is used to cure pediatric AML in other children [113-117].

Diagnosis and management of ML-DS is discussed separately. (See "Myeloid leukemia associated with Down syndrome (ML-DS)".)

Myeloid sarcoma — Myeloid sarcoma (chloroma) refers to deposits of myeloid blasts outside the bone marrow that may cause destruction or compression in normal tissue. Management of myeloid sarcoma is like that for other cases of AML.

Myeloid sarcoma most often affects the central nervous system (CNS), skin, orbit, and bone. The incidence of myeloid sarcoma in pediatric AML is approximately 10 percent and is more common in patients with the following features [118,119]:

Younger age

High white blood cell (WBC) count at diagnosis

t(8;21)

Myelomonocytic morphology

In children, the prognostic impact of myeloid sarcoma appears to depend upon the site of involvement [57,118,119]. Children with myeloid sarcoma involving the CNS or orbit have improved outcomes compared with those without CNS/orbit involvement and compared with patients without chloroma. By contrast, children with chloromatous skin involvement have higher rates of relapse.

Extramedullary relapses are more common among children with extramedullary disease at diagnosis. Focal radiation may provide symptomatic benefit, but it does not appear to offer any additional survival benefit compared with chemotherapy in patients with myeloid sarcoma [118,120].

Acute promyelocytic leukemia — Acute promyelocytic leukemia (APL) is a rare subtype of AML that is characterized by maturation arrest at the promyelocyte stage, t(15;17)(q22;q21) chromosomal translocation, and PML::RARA rearrangement. Rare cases are associated with other genetic rearrangements.

Many patients with APL present with disseminated intravascular coagulation (DIC), which requires urgent and aggressive management. All-trans retinoic acid should be started quickly when APL is suspected [121].

Treatment of APL is risk stratified based on the presenting WBC count [122]:

Standard risk – WBC <10 x 109/L (<10,000/microL) at presentation

High risk – WBC ≥10 x 109/L

Patients with a high WBC count at presentation experience higher rates of relapse and are especially susceptible to early mortality due to DIC.

All-trans retinoic acid, which targets the PML::RARA fusion protein and induces differentiation of APL blasts, is a critical component of APL management [123]. Treatment using an anthracycline, all-trans retinoic acid, and arsenic trioxide is associated with excellent outcomes [124-127]. A prospective study reported that for children with standard-risk APL who were treated with all-trans retinoic acid and arsenic trioxide, outcomes were similar to those using chemotherapy-containing regimens [128]. Patients with high-risk APL treated with an anthracycline, all-trans retinoic acid, and arsenic trioxide for induction therapy, followed by consolidation therapy using all-trans retinoic acid and arsenic trioxide, had outcomes comparable with regimens that used more anthracycline and other conventional chemotherapies.

Management of APL in children is like that in adults, as discussed separately. (See "Initial treatment of acute promyelocytic leukemia in adults".)

LATE EFFECTS — Potential late treatment-related adverse effects include late onset cardiotoxicity, central nervous system (CNS) impairment, decreased linear growth, infertility, cataracts, secondary cancers, and impaired health status due to such factors as neurocognitive dysfunction, depression, fatigue, and anxiety. (See "Assuring quality of care for cancer survivors: The survivorship care plan" and "Overview of cancer survivorship in adolescents and young adults".)

The occurrence of specific complications depends on the patient's age and the type and intensity of therapy with which they were treated. Specific long-term follow-up guidelines after treatment of childhood cancer have been published by the Children's Oncology Group.

Cardiac – Anthracyclines carry a risk of both early and late onset cardiotoxicity, which in severe cases results in heart failure. The cumulative anthracycline dose is the most important risk factor for late cardiac effects.

Late cardiac effects are seen in 3 to 5 percent of patients who receive 300 to 440 mg/m2 cumulative anthracycline dosing with contemporary treatment protocols for pediatric AML [129,130]. There is no dose that is recognized to be safe, but cumulative doses of >300 mg/m2 are associated with higher rates of late cardiac compromise; this risk rises even more steeply with >400 mg/m2 [131,132].

Children and adolescents treated for AML should receive long-term follow-up with routine cardiac monitoring.

Second cancers – Survivors of childhood AML are at risk for secondary neoplasms, mainly solid tumors.

Long-term follow-up data report a prevalence of approximately 1.7 percent [133]. This risk is increased in patients who receive radiation, which may be used in hematopoietic cell transplantation (HCT) conditioning [134].

Endocrine – The intensity of therapy for AML, especially radiation therapy for HCT, puts survivors at risk for thyroid, gonadal, and other endocrine dysfunction [133-135].

Other – Children who receive total body irradiation or cranial irradiation are at risk for growth defects and neurocognitive dysfunction; this is more pronounced in very young children who receive radiation [136].

Further details of late effects of treatment of pediatric leukemia are described separately. (See "Acute lymphoblastic leukemia/lymphoblastic lymphoma: Outcomes and late effects of treatment in children and adolescents".)

RELAPSED/REFRACTORY AML — Approximately one-third of children with AML experience relapse. The prognosis for these children is generally poor and is especially grave for those patients with high-risk disease features or who previously received allogeneic hematopoietic cell transplantation (HCT).

We strongly encourage enrollment in a clinical trial.

Time to relapse is an important prognostic factor, with survival <20 percent for patients relapsing within one year of achieving first complete remission (CR) [137].

Salvage therapy – There is no consensus regimen for relapsed or refractory (r/r) pediatric AML. Reinduction regimens vary but commonly include cytarabine, with or without an anthracycline. Use of an anthracycline depends on cardiac function at relapse and the cumulative anthracycline dose with initial therapy.

High-dose cytarabine – Regimens have combined high-dose cytarabine (HiDAC) with other agents, such as fludarabine and clofarabine, and anthracyclines when appropriate [53,138,139].

CPX-351 – Treatment of 32 children with CPX-351 (liposomal daunorubicin-cytarabine) was associated with 54 percent CR and 26 percent with CR with incomplete hematologic recovery, most of whom had no detectable measurable residual disease (MRD) by flow cytometry [140]. Treatment was associated with grade ≥3 fever/neutropenia, infection, and rash in 45, 47, and 40 percent, respectively.

CPX-351 is approved by the US Food and Drug Administration (FDA) for children ≥1 year with newly diagnosed, therapy-related AML or AML with myelodysplasia-related changes [141-144].

Gemtuzumab ozogamicin – This CD33-directed immunoconjugate has shown responses in relapsed CD33-positive AML [145,146].

Gemtuzumab ozogamicin is approved by the FDA for treatment of r/r CD33-positive AML in patients ≥2 years.

Venetoclax – The BCL-2 inhibitor, venetoclax, has activity in children with r/r AML. A study of venetoclax combined with cytarabine (with or without idarubicin) reported a 70 percent overall response rate, with many patients being MRD negative [147].

Achievement of a second CR (CR2) is the most important prognostic factor in long-term survival, and patients who have no evidence of MRD following second-line therapy have the best outcomes [138].

Consolidation – Consolidation with allogeneic HCT is the only approach that offers children with r/r AML who achieve CR2 a chance for long-term survival.

Most study groups advocate HCT in CR2 for patients who were not previously transplanted [53,148-150]. Investigational approaches with a clinical trial should be considered for patients who relapse after having previously undergone HCT.

Central nervous system relapse – Central nervous system (CNS) relapse can occur either as an isolated relapse or as part of a combined relapse that usually involves the bone marrow. In one study, nearly 10 percent of patients had CNS involvement at relapse [53].

CNS relapse is managed as discussed above. (See 'Central nervous system management' above.)

Other treatments that are under investigation for r/r AML include epigenetic modifiers, targeted inhibitors, monoclonal antibody-based agents, chimeric antigen receptor T cells, novel T cell engagers (eg, dual affinity targeting receptors), and natural killer cell strategies [151-158].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Acute myeloid leukemia".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient education" and the keyword(s) of interest.)

Basics topics (see "Patient education: Leukemia in children (The Basics)" and "Patient education: Acute myeloid leukemia (AML) (The Basics)")

SUMMARY AND RECOMMENDATIONS

Description – Acute myeloid leukemia (AML) accounts for 15 percent of childhood acute leukemia. Pediatric AML is less common and has an inferior prognosis compared with childhood acute lymphoblastic leukemia (ALL).

Clinical presentation – Fever, malaise, musculoskeletal pains, lymphadenopathy, hepatosplenomegaly, bleeding, and/or cytopenias are common. Some present with disseminated intravascular coagulation, neurologic abnormalities, hyperleukocytosis, or tumor lysis syndrome. (See 'Clinical presentation' above.)

Evaluation – Clinical, laboratory, and pathologic evaluation is described above. (See 'Evaluation' above.)

All children and adolescents should have a lumbar puncture (LP) at diagnosis to assess central nervous system (CNS) involvement.

Diagnosis – AML should be considered in children with unexplained cytopenias or associated symptoms, organomegaly, or circulating blasts.

Diagnosis – Based on morphology and immunophenotype of myeloid blasts in bone marrow, blood, and/or extramedullary sites. (See 'Diagnosis' above.)

Differential diagnosis – ALL and other leukemias and other causes of cytopenias (eg, aplastic anemia) or circulating immature cells should be excluded. (See 'Differential diagnosis' above.)

Classification – Classified according to the International Consensus Classification (ICC) or World Health Organization 5th edition (WHO5) as a subtype within one of the following large categories (see 'Classification' above):

-AML with defining genetic features

-Other subtypes (defined by ICC and WHO5 criteria)

Prognosis – Informed by cytogenetic/molecular features and treatment response. (See 'Prognostic factors' above.)

Risk stratification – Stratified as low, intermediate, or high risk, based on genetic features and initial treatment response. (See 'Risk stratification' above.)

Treatment – Participation in a clinical trial is encouraged. Care should adhere to a contemporary pediatric treatment protocol. Treatment comprises:

Induction therapy – Induction therapy with an anthracycline (or mitoxantrone) plus cytarabine is standard therapy; choice of agents, protocol for administration, and addition of a third agent is guided by the chosen protocol. (See 'Induction therapy' above.)

Central nervous system management – All patients should have an initial diagnostic LP. (See 'Clinical/laboratory testing' above.)

-No central nervous system disease – We suggest intrathecal (IT) cytarabine or triple IT therapy (cytarabine, methotrexate, hydrocortisone) prophylaxis (Grade 2C). (See 'Central nervous system management' above.)

-Central nervous system involvement – There is no established consensus. Patients are generally treated with more intensive IT and systemic chemotherapy, with or without cranial radiation therapy, as guided by the chosen treatment protocol.

Consolidation – Varies with prognostic features and protocol:

-Favorable risk – We suggest high-dose cytarabine (HiDAC)-based consolidation chemotherapy rather than allogeneic hematopoietic cell transplantation (HCT), observation, or autologous HCT (Grade 2C). (See 'Consolidation therapy' above.)

-Intermediate/higher risk – HiDAC or allogeneic HCT are reasonable options.

Maintenance – We suggest not administering maintenance therapy (Grade 2C). (See 'Maintenance therapy' above.)

Adjunctive/supportive care – Monitoring and management of symptoms, cytopenias, and complications are essential aspects of care, as described above. (See 'Adjunctive/supportive care' above.)

Special populations – Children with Down syndrome, myeloid sarcoma (chloroma), and acute promyelocytic leukemia require distinctive management. (See 'Special populations' above.)

Late toxicity – Treatment may be associated with late cardiotoxicity, second cancers, and other late effects. (See 'Late effects' above.)

Relapsed/refractory AML – Prognosis of relapsed or refractory AML is poor. There is no consensus for salvage induction therapy.

For patients with complete remission after salvage therapy, allogeneic HCT (if eligible) or a clinical trial should be considered. (See 'Relapsed/refractory AML' above.)

  1. Pui CH, Schrappe M, Ribeiro RC, Niemeyer CM. Childhood and adolescent lymphoid and myeloid leukemia. Hematology Am Soc Hematol Educ Program 2004; :118.
  2. Bolouri H, Farrar JE, Triche T Jr, et al. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat Med 2018; 24:103.
  3. Tarlock K, Zhong S, He Y, et al. Distinct age-associated molecular profiles in acute myeloid leukemia defined by comprehensive clinical genomic profiling. Oncotarget 2018; 9:26417.
  4. Arber DA, Orazi A, Hasserjian RP, et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: integrating morphologic, clinical, and genomic data. Blood 2022; 140:1200.
  5. Khoury JD, Solary E, Abla O, et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022; 36:1703.
  6. Bennett JM, Catovsky D, Daniel MT, et al. Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-American-British Cooperative Group. Ann Intern Med 1985; 103:620.
  7. World health organization classification of tumours of haematopoietic and lymphoid tissues, revised 4th edition, Swerdlow SH, Campo E, Harris NL, et al. (Eds), IARC, Lyon 2017.
  8. Gibson BE, Webb DK, Howman AJ, et al. Results of a randomized trial in children with Acute Myeloid Leukaemia: medical research council AML12 trial. Br J Haematol 2011; 155:366.
  9. Gamis AS, Alonzo TA, Meshinchi S, et al. Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531. J Clin Oncol 2014; 32:3021.
  10. Klein K, Kaspers G, Harrison CJ, et al. Clinical Impact of Additional Cytogenetic Aberrations, cKIT and RAS Mutations, and Treatment Elements in Pediatric t(8;21)-AML: Results From an International Retrospective Study by the International Berlin-Frankfurt-Münster Study Group. J Clin Oncol 2015; 33:4247.
  11. Tarlock K, Alonzo TA, Moraleda PP, et al. Acute myeloid leukaemia (AML) with t(6;9)(p23;q34) is associated with poor outcome in childhood AML regardless of FLT3-ITD status: a report from the Children's Oncology Group. Br J Haematol 2014; 166:254.
  12. Inaba H, Zhou Y, Abla O, et al. Heterogeneous cytogenetic subgroups and outcomes in childhood acute megakaryoblastic leukemia: a retrospective international study. Blood 2015; 126:1575.
  13. Cooper TM, Ries RE, Alonzo TA, et al. Revised risk stratification criteria for children with newly diagnosed acute myeloid leukemia: a report from the Children’s Oncology Group. Blood 2017; 130:Abstract 407.
  14. Vujkovic M, Attiyeh EF, Ries RE, et al. Genomic architecture and treatment outcome in pediatric acute myeloid leukemia: a Children's Oncology Group report. Blood 2017; 129:3051.
  15. Eidenschink Brodersen L, Alonzo TA, Menssen AJ, et al. A recurrent immunophenotype at diagnosis independently identifies high-risk pediatric acute myeloid leukemia: a report from Children's Oncology Group. Leukemia 2016; 30:2077.
  16. Gronseth CM, McElhone SE, Storer BE, et al. Prognostic significance of acquired copy-neutral loss of heterozygosity in acute myeloid leukemia. Cancer 2015; 121:2900.
  17. 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; 113:6558.
  18. Tarlock K, Lamble AJ, Wang YC, et al. CEBPA-bZip mutations are associated with favorable prognosis in de novo AML: a report from the Children's Oncology Group. Blood 2021; 138:1137.
  19. Brown P, McIntyre E, Rau R, et al. The incidence and clinical significance of nucleophosmin mutations in childhood AML. Blood 2007; 110:979.
  20. Meshinchi S, Woods WG, Stirewalt DL, et al. Prevalence and prognostic significance of Flt3 internal tandem duplication in pediatric acute myeloid leukemia. Blood 2001; 97:89.
  21. Zwaan CM, Meshinchi S, Radich JP, et al. FLT3 internal tandem duplication in 234 children with acute myeloid leukemia: prognostic significance and relation to cellular drug resistance. Blood 2003; 102:2387.
  22. Meshinchi S, Alonzo TA, Stirewalt DL, et al. Clinical implications of FLT3 mutations in pediatric AML. Blood 2006; 108:3654.
  23. Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood 2014; 124:3441.
  24. Creutzig U, Büchner T, Sauerland MC, et al. Significance of age in acute myeloid leukemia patients younger than 30 years: a common analysis of the pediatric trials AML-BFM 93/98 and the adult trials AMLCG 92/99 and AMLSG HD93/98A. Cancer 2008; 112:562.
  25. Balgobind BV, Zwaan CM, Pieters R, Van den Heuvel-Eibrink MM. The heterogeneity of pediatric MLL-rearranged acute myeloid leukemia. Leukemia 2011; 25:1239.
  26. Niktoreh N, Walter C, Zimmermann M, et al. Mutated WT1, FLT3-ITD, and NUP98-NSD1 Fusion in Various Combinations Define a Poor Prognostic Group in Pediatric Acute Myeloid Leukemia. J Oncol 2019; 2019:1609128.
  27. Struski S, Lagarde S, Bories P, et al. NUP98 is rearranged in 3.8% of pediatric AML forming a clinical and molecular homogenous group with a poor prognosis. Leukemia 2017; 31:565.
  28. 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; 118:3645.
  29. 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; 115:2372.
  30. 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; 116:702.
  31. 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; 113:5951.
  32. 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; 96:1478.
  33. van der Velden VH, van der 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; 24:1599.
  34. Loken MR, Alonzo TA, Pardo L, et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: a report from Children's Oncology Group. Blood 2012; 120:1581.
  35. Buldini B, Rizzati F, Masetti R, et al. Prognostic significance of flow-cytometry evaluation of minimal residual disease in children with acute myeloid leukaemia treated according to the AIEOP-AML 2002/01 study protocol. Br J Haematol 2017; 177:116.
  36. MRD-AML-BFM Study Group, Langebrake C, Creutzig U, et al. Residual disease monitoring in childhood acute myeloid leukemia by multiparameter flow cytometry: the MRD-AML-BFM Study Group. J Clin Oncol 2006; 24:3686.
  37. Steinbach D, Bader P, Willasch A, et al. Prospective validation of a new method of monitoring minimal residual disease in childhood acute myelogenous leukemia. Clin Cancer Res 2015; 21:1353.
  38. Rizzari C, Cazzaniga G, Coliva T, et al. Predictive factors of relapse and survival in childhood acute myeloid leukemia: role of minimal residual disease. Expert Rev Anticancer Ther 2011; 11:1391.
  39. Creutzig U, van den Heuvel-Eibrink MM, Gibson B, et al. Diagnosis and management of acute myeloid leukemia in children and adolescents: recommendations from an international expert panel. Blood 2012; 120:3187.
  40. 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; 19:2130.
  41. 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; 26:5797.
  42. Pollard JA, Alonzo TA, Gerbing R, et al. Sorafenib in Combination With Standard Chemotherapy for Children With High Allelic Ratio FLT3/ITD+ Acute Myeloid Leukemia: A Report From the Children's Oncology Group Protocol AAML1031. J Clin Oncol 2022; 40:2023.
  43. Tarlock K, Chang B, Cooper T, et al. Sorafenib treatment following hematopoietic stem cell transplant in pediatric FLT3/ITD acute myeloid leukemia. Pediatr Blood Cancer 2015; 62:1048.
  44. Zwaan CM, Kolb EA, Reinhardt D, et al. Collaborative Efforts Driving Progress in Pediatric Acute Myeloid Leukemia. J Clin Oncol 2015; 33:2949.
  45. Creutzig U, Zimmermann M, Ritter J, et al. Treatment strategies and long-term results in paediatric patients treated in four consecutive AML-BFM trials. Leukemia 2005; 19:2030.
  46. Creutzig U, Zimmermann M, Bourquin JP, et al. Randomized trial comparing liposomal daunorubicin with idarubicin as induction for pediatric acute myeloid leukemia: results from Study AML-BFM 2004. Blood 2013; 122:37.
  47. Mandelli F, Vignetti M, Suciu S, et al. Daunorubicin versus mitoxantrone versus idarubicin as induction and consolidation chemotherapy for adults with acute myeloid leukemia: the EORTC and GIMEMA Groups Study AML-10. J Clin Oncol 2009; 27:5397.
  48. Armenian SH, Hudson MM, Mulder RL, et al. Recommendations for cardiomyopathy surveillance for survivors of childhood cancer: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group. Lancet Oncol 2015; 16:e123.
  49. 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; 11:543.
  50. Ravindranath Y, Chang M, Steuber CP, et al. Pediatric Oncology Group (POG) studies of acute myeloid leukemia (AML): a review of four consecutive childhood AML trials conducted between 1981 and 2000. Leukemia 2005; 19:2101.
  51. 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; 118:5409.
  52. Radhakrishnan V, Bakhshi S, Kayal S, et al. Two-drug versus three-drug induction chemotherapy in pediatric acute myeloid leukemia: a randomized controlled trial. Blood Cancer J 2022; 12:131.
  53. Kaspers GJ, Zimmermann M, Reinhardt D, et al. Improved outcome in pediatric relapsed acute myeloid leukemia: results of a randomized trial on liposomal daunorubicin by the International BFM Study Group. J Clin Oncol 2013; 31:599.
  54. Dahl GV, Simone JV, Hustu HO, Mason C. Preventive central nervous system irradiation in children with acute nonlymphocytic leukemia. Cancer 1978; 42:2187.
  55. Chow EJ, Liu W, Srivastava K, et al. Differential effects of radiotherapy on growth and endocrine function among acute leukemia survivors: a childhood cancer survivor study report. Pediatr Blood Cancer 2013; 60:110.
  56. Kadan-Lottick NS, Zeltzer LK, Liu Q, et al. Neurocognitive functioning in adult survivors of childhood non-central nervous system cancers. J Natl Cancer Inst 2010; 102:881.
  57. Johnston DL, Alonzo TA, Gerbing RB, et al. Superior outcome of pediatric acute myeloid leukemia patients with orbital and CNS myeloid sarcoma: a report from the Children's Oncology Group. Pediatr Blood Cancer 2012; 58:519.
  58. 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; 17:2090.
  59. Kaspers GJ, Creutzig U. Pediatric acute myeloid leukemia: international progress and future directions. Leukemia 2005; 19:2025.
  60. Niewerth D, Creutzig U, Bierings MB, Kaspers GJ. A review on allogeneic stem cell transplantation for newly diagnosed pediatric acute myeloid leukemia. Blood 2010; 116:2205.
  61. Hasle H. A critical review of which children with acute myeloid leukaemia need stem cell procedures. Br J Haematol 2014; 166:23.
  62. 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; 27:4007.
  63. 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; 122:217.
  64. 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; 97:56.
  65. 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; 334:1428.
  66. 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; 13:1.
  67. Lange BJ, Smith FO, Feusner J, et al. Outcomes in CCG-2961, a children's oncology group phase 3 trial for untreated pediatric acute myeloid leukemia: a report from the children's oncology group. Blood 2008; 111:1044.
  68. Kelly MJ, Horan JT, Alonzo TA, et al. Comparable survival for pediatric acute myeloid leukemia with poor-risk cytogenetics following chemotherapy, matched related donor, or unrelated donor transplantation. Pediatr Blood Cancer 2014; 61:269.
  69. Klusmann JH, Reinhardt D, Zimmermann M, et al. The role of matched sibling donor allogeneic stem cell transplantation in pediatric high-risk acute myeloid leukemia: results from the AML-BFM 98 study. Haematologica 2012; 97:21.
  70. Meshinchi S, Arceci RJ, Sanders JE, et al. Role of allogeneic stem cell transplantation in FLT3/ITD-positive AML. Blood 2006; 108:400; author reply 400.
  71. Schechter T, Gassas A, Chen H, et al. The outcome of allogeneic hematopoietic cell transplantation for children with FMS-like tyrosine kinase 3 internal tandem duplication-positive acute myelogenous leukemia. Biol Blood Marrow Transplant 2015; 21:172.
  72. Bleakley M, Lau L, Shaw PJ, Kaufman A. Bone marrow transplantation for paediatric AML in first remission: a systematic review and meta-analysis. Bone Marrow Transplant 2002; 29:843.
  73. Leung W, Pui CH, Coustan-Smith E, et al. Detectable minimal residual disease before hematopoietic cell transplantation is prognostic but does not preclude cure for children with very-high-risk leukemia. Blood 2012; 120:468.
  74. Moore J, Nivison-Smith I, Goh K, et al. Equivalent survival for sibling and unrelated donor allogeneic stem cell transplantation for acute myelogenous leukemia. Biol Blood Marrow Transplant 2007; 13:601.
  75. 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 (Leucémie Aiguë Myéloblastique Enfant) Cooperative Group. Leukemia 2005; 19:2082.
  76. Aladjidi N, Auvrignon A, Leblanc T, et al. Outcome in children with relapsed acute myeloid leukemia after initial treatment with the French Leucemie Aique Myeloide Enfant (LAME) 89/91 protocol of the French Society of Pediatric Hematology and Immunology. J Clin Oncol 2003; 21:4377.
  77. Wells RJ, Woods WG, Buckley JD, et al. Treatment of newly diagnosed children and adolescents with acute myeloid leukemia: a Childrens Cancer Group study. J Clin Oncol 1994; 12:2367.
  78. Lange BJ, Yang RK, Gan J, et al. Soluble interleukin-2 receptor α activation in a Children's Oncology Group randomized trial of interleukin-2 therapy for pediatric acute myeloid leukemia. Pediatr Blood Cancer 2011; 57:398.
  79. Rubnitz JE, Razzouk BI, Lensing S, et al. Prognostic factors and outcome of recurrence in childhood acute myeloid leukemia. Cancer 2007; 109:157.
  80. Hasle H, Abrahamsson J, Forestier E, et al. Gemtuzumab ozogamicin as postconsolidation therapy does not prevent relapse in children with AML: results from NOPHO-AML 2004. Blood 2012; 120:978.
  81. Alexander S, Fisher BT, Gaur AH, et al. Effect of Levofloxacin Prophylaxis on Bacteremia in Children With Acute Leukemia or Undergoing Hematopoietic Stem Cell Transplantation: A Randomized Clinical Trial. JAMA 2018; 320:995.
  82. Creutzig U, Zimmermann M, Lehrnbecher T, et al. Less toxicity by optimizing chemotherapy, but not by addition of granulocyte colony-stimulating factor in children and adolescents with acute myeloid leukemia: results of AML-BFM 98. J Clin Oncol 2006; 24:4499.
  83. Lehrnbecher T, Sung L. Anti-infective prophylaxis in pediatric patients with acute myeloid leukemia. Expert Rev Hematol 2014; 7:819.
  84. Canner J, Alonzo TA, Franklin J, et al. Differences in outcomes of newly diagnosed acute myeloid leukemia for adolescent/young adult and younger patients: a report from the Children's Oncology Group. Cancer 2013; 119:4162.
  85. Rubnitz JE, Pounds S, Cao X, et al. Treatment outcome in older patients with childhood acute myeloid leukemia. Cancer 2012; 118:6253.
  86. Barton CD, Waugh LK, Nielsen MJ, Paulus S. Febrile neutropenia in children treated for malignancy. J Infect 2015; 71 Suppl 1:S27.
  87. Patterson TF, Thompson GR 3rd, Denning DW, et al. Practice Guidelines for the Diagnosis and Management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin Infect Dis 2016; 63:e1.
  88. Feusner JH, Hastings CA. Infections in children with acute myelogenous leukemia. Concepts of management and prevention. J Pediatr Hematol Oncol 1995; 17:234.
  89. Lewis V, Yanofsky R, Mitchell D, et al. Predictors and outcomes of viridans group streptococcal infections in pediatric acute myeloid leukemia: from the Canadian infections in AML research group. Pediatr Infect Dis J 2014; 33:126.
  90. Okamoto Y, Ribeiro RC, Srivastava DK, et al. Viridans streptococcal sepsis: clinical features and complications in childhood acute myeloid leukemia. J Pediatr Hematol Oncol 2003; 25:696.
  91. Creutzig U, Zimmermann M, Reinhardt D, et al. Early deaths and treatment-related mortality in children undergoing therapy for acute myeloid leukemia: analysis of the multicenter clinical trials AML-BFM 93 and AML-BFM 98. J Clin Oncol 2004; 22:4384.
  92. Yamaguchi M, Kurokawa T, Ishiyama K, et al. Efficacy and safety of micafungin as an empirical therapy for invasive fungal infections in patients with hematologic disorders: a multicenter, prospective study. Ann Hematol 2011; 90:1209.
  93. Emiroglu M. Micafungin use in children. Expert Rev Anti Infect Ther 2011; 9:821.
  94. Fisher BT, Kavcic M, Li Y, et al. Antifungal prophylaxis associated with decreased induction mortality rates and resources utilized in children with new-onset acute myeloid leukemia. Clin Infect Dis 2014; 58:502.
  95. Rotstein C, Bow EJ, Laverdiere M, et al. Randomized placebo-controlled trial of fluconazole prophylaxis for neutropenic cancer patients: benefit based on purpose and intensity of cytotoxic therapy. The Canadian Fluconazole Prophylaxis Study Group. Clin Infect Dis 1999; 28:331.
  96. Mehta AK, Langston AA. Use of posaconazole in the treatment of invasive fungal  infections. Expert Rev Hematol 2009; 2:619.
  97. Cornely OA, Maertens J, Winston DJ, et al. Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia. N Engl J Med 2007; 356:348.
  98. Pagano L, Caira M, Candoni A, et al. Evaluation of the practice of antifungal prophylaxis use in patients with newly diagnosed acute myeloid leukemia: results from the SEIFEM 2010-B registry. Clin Infect Dis 2012; 55:1515.
  99. Glasmacher A, Prentice A, Gorschlüter M, et al. Itraconazole prevents invasive fungal infections in neutropenic patients treated for hematologic malignancies: evidence from a meta-analysis of 3,597 patients. J Clin Oncol 2003; 21:4615.
  100. Creutzig U, Ritter J, Budde M, et al. Early deaths due to hemorrhage and leukostasis in childhood acute myelogenous leukemia. Associations with hyperleukocytosis and acute monocytic leukemia. Cancer 1987; 60:3071.
  101. 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; 28:2682.
  102. Bunin NJ, Pui CH. Differing complications of hyperleukocytosis in children with acute lymphoblastic or acute nonlymphoblastic leukemia. J Clin Oncol 1985; 3:1590.
  103. Ganzel C, Becker J, Mintz PD, et al. Hyperleukocytosis, leukostasis and leukapheresis: practice management. Blood Rev 2012; 26:117.
  104. Christakopoulos GE, Walker KN, Smith J, et al. Clinical characteristics and outcomes of children with newly diagnosed acute myeloid leukemia and hyperleukocytosis managed with different cytoreductive methods. Cancer 2023; 129:1873.
  105. Porcu P, Farag S, Marcucci G, et al. Leukocytoreduction for acute leukemia. Ther Apher 2002; 6:15.
  106. Lange BJ, Kobrinsky N, Barnard DR, et al. Distinctive demography, biology, and outcome of acute myeloid leukemia and myelodysplastic syndrome in children with Down syndrome: Children's Cancer Group Studies 2861 and 2891. Blood 1998; 91:608.
  107. O'Brien MM, Taub JW, Chang MN, et al. Cardiomyopathy in children with Down syndrome treated for acute myeloid leukemia: a report from the Children's Oncology Group Study POG 9421. J Clin Oncol 2008; 26:414.
  108. Klusmann JH, Creutzig U, Zimmermann M, et al. Treatment and prognostic impact of transient leukemia in neonates with Down syndrome. Blood 2008; 111:2991.
  109. Al-Kasim F, Doyle JJ, Massey GV, et al. Incidence and treatment of potentially lethal diseases in transient leukemia of Down syndrome: Pediatric Oncology Group Study. J Pediatr Hematol Oncol 2002; 24:9.
  110. 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; 118:6752.
  111. Taub JW, Huang X, Matherly LH, et al. Expression of chromosome 21-localized genes in acute myeloid leukemia: differences between Down syndrome and non-Down syndrome blast cells and relationship to in vitro sensitivity to cytosine arabinoside and daunorubicin. Blood 1999; 94:1393.
  112. Ge Y, Jensen TL, Stout ML, et al. The role of cytidine deaminase and GATA1 mutations in the increased cytosine arabinoside sensitivity of Down syndrome myeloblasts and leukemia cell lines. Cancer Res 2004; 64:728.
  113. 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; 101:130.
  114. Ravindranath Y, Abella E, Krischer JP, et al. Acute myeloid leukemia (AML) in Down's syndrome is highly responsive to chemotherapy: experience on Pediatric Oncology Group AML Study 8498. Blood 1992; 80:2210.
  115. Creutzig U, Reinhardt D, Diekamp S, et al. AML patients with Down syndrome have a high cure rate with AML-BFM therapy with reduced dose intensity. Leukemia 2005; 19:1355.
  116. Zeller B, Gustafsson G, Forestier E, et al. Acute leukaemia in children with Down syndrome: a population-based Nordic study. Br J Haematol 2005; 128:797.
  117. Sorrell AD, Alonzo TA, Hilden JM, et al. Favorable survival maintained in children who have myeloid leukemia associated with Down syndrome using reduced-dose chemotherapy on Children's Oncology Group trial A2971: a report from the Children's Oncology Group. Cancer 2012; 118:4806.
  118. Dusenbery KE, Howells WB, Arthur DC, et al. Extramedullary leukemia in children with newly diagnosed acute myeloid leukemia: a report from the Children's Cancer Group. J Pediatr Hematol Oncol 2003; 25:760.
  119. Shimizu H, Saitoh T, Hatsumi N, et al. Clinical significance of granulocytic sarcoma in adult patients with acute myeloid leukemia. Cancer Sci 2012; 103:1513.
  120. Hall MD, Chen YJ, Schultheiss TE, et al. Treatment outcomes for patients with chloroma receiving radiation therapy. J Med Imaging Radiat Oncol 2014; 58:523.
  121. Breen KA, Grimwade D, Hunt BJ. The pathogenesis and management of the coagulopathy of acute promyelocytic leukaemia. Br J Haematol 2012; 156:24.
  122. Sanz MA, Lo Coco F, Martín G, et al. Definition of relapse risk and role of nonanthracycline drugs for consolidation in patients with acute promyelocytic leukemia: a joint study of the PETHEMA and GIMEMA cooperative groups. Blood 2000; 96:1247.
  123. Petrie K, Zelent A, Waxman S. Differentiation therapy of acute myeloid leukemia: past, present and future. Curr Opin Hematol 2009; 16:84.
  124. Avvisati G, Lo-Coco F, Paoloni FP, et al. AIDA 0493 protocol for newly diagnosed acute promyelocytic leukemia: very long-term results and role of maintenance. Blood 2011; 117:4716.
  125. Powell BL, Moser B, Stock W, et al. Arsenic trioxide improves event-free and overall survival for adults with acute promyelocytic leukemia: North American Leukemia Intergroup Study C9710. Blood 2010; 116:3751.
  126. Abla O, Ribeiro RC. How I treat children and adolescents with acute promyelocytic leukaemia. Br J Haematol 2014; 164:24.
  127. Testi AM, Pession A, Diverio D, et al. Risk-adapted treatment of acute promyelocytic leukemia: results from the International Consortium for Childhood APL. Blood 2018; 132:405.
  128. Kutny MA, Alonzo TA, Abla O, et al. Assessment of Arsenic Trioxide and All-trans Retinoic Acid for the Treatment of Pediatric Acute Promyelocytic Leukemia: A Report From the Children's Oncology Group AAML1331 Trial. JAMA Oncol 2022; 8:79.
  129. Creutzig U, Diekamp S, Zimmermann M, Reinhardt D. Longitudinal evaluation of early and late anthracycline cardiotoxicity in children with AML. Pediatr Blood Cancer 2007; 48:651.
  130. Getz KD, Sung L, Ky B, et al. Occurrence of Treatment-Related Cardiotoxicity and Its Impact on Outcomes Among Children Treated in the AAML0531 Clinical Trial: A Report From the Children's Oncology Group. J Clin Oncol 2019; 37:12.
  131. Lipshultz SE, Franco VI, Miller TL, et al. Cardiovascular disease in adult survivors of childhood cancer. Annu Rev Med 2015; 66:161.
  132. Kremer LC, van Dalen EC, Offringa M, et al. Anthracycline-induced clinical heart failure in a cohort of 607 children: long-term follow-up study. J Clin Oncol 2001; 19:191.
  133. Mulrooney DA, Dover DC, Li S, et al. Twenty years of follow-up among survivors of childhood and young adult acute myeloid leukemia: a report from the Childhood Cancer Survivor Study. Cancer 2008; 112:2071.
  134. Meadows AT, Friedman DL, Neglia JP, et al. Second neoplasms in survivors of childhood cancer: findings from the Childhood Cancer Survivor Study cohort. J Clin Oncol 2009; 27:2356.
  135. Park J, Choi EK, Kim JH, et al. Effects of total body irradiation-based conditioning on allogeneic stem cell transplantation for pediatric acute leukemia: a single-institution study. Radiat Oncol J 2014; 32:198.
  136. Chemaitilly W, Sklar CA. Endocrine complications in long-term survivors of childhood cancers. Endocr Relat Cancer 2010; 17:R141.
  137. 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; 24:1422.
  138. Cooper TM, Alonzo TA, Gerbing RB, et al. AAML0523: a report from the Children's Oncology Group on the efficacy of clofarabine in combination with cytarabine in pediatric patients with recurrent acute myeloid leukemia. Cancer 2014; 120:2482.
  139. Fleischhack G, Hasan C, Graf N, et al. IDA-FLAG (idarubicin, fludarabine, cytarabine, G-CSF), an effective remission-induction therapy for poor-prognosis AML of childhood prior to allogeneic or autologous bone marrow transplantation: experiences of a phase II trial. Br J Haematol 1998; 102:647.
  140. Cooper TM, Absalon MJ, Alonzo TA, et al. Phase I/II Study of CPX-351 Followed by Fludarabine, Cytarabine, and Granulocyte-Colony Stimulating Factor for Children With Relapsed Acute Myeloid Leukemia: A Report From the Children's Oncology Group. J Clin Oncol 2020; 38:2170.
  141. Daunorubicin and cytarabine liposome for injection. United States prescribing information. Revised March 2021. US Food and Drug Administration. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/209401s006lbl.pdf (Accessed on April 21, 2021).
  142. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia. J Clin Oncol 2018; 36:2684.
  143. Cooper TM, Absalon M, Alonzo TA, et al. AAML1421, a phase I/II study of CPX-351 followed by fludarabine, cytarabine, and G-CSF (FLAG) for children with relapsed acute myeloid leukemia (AML): A report from the Children’s Oncology Group. J Clin Oncol 2019; 37:Abstract 10003.
  144. Absalon M, O'Brien MM, Phillips CL, et al. A phase I/pilot study of CPX-351 for children, adolescents and young adults with recurrent or refractory hematologic malignancies. J Clin Oncol 2016; 34:Abstract 10541.
  145. Niktoreh N, Lerius B, Zimmermann M, et al. Gemtuzumab ozogamicin in children with relapsed or refractory acute myeloid leukemia: a report by Berlin-Frankfurt-Münster study group. Haematologica 2019; 104:120.
  146. Arceci RJ, Sande J, Lange B, et al. Safety and efficacy of gemtuzumab ozogamicin in pediatric patients with advanced CD33+ acute myeloid leukemia. Blood 2005; 106:1183.
  147. Karol SE, Alexander TB, Budhraja A, et al. Venetoclax in combination with cytarabine with or without idarubicin in children with relapsed or refractory acute myeloid leukaemia: a phase 1, dose-escalation study. Lancet Oncol 2020; 21:551.
  148. Kaspers G. How I treat paediatric relapsed acute myeloid leukaemia. Br J Haematol 2014; 166:636.
  149. Goemans BF, Tamminga RY, Corbijn CM, et al. Outcome for children with relapsed acute myeloid leukemia in the Netherlands following initial treatment between 1980 and 1998: survival after chemotherapy only? Haematologica 2008; 93:1418.
  150. Testi AM, Biondi A, Lo Coco F, et al. GIMEMA-AIEOPAIDA protocol for the treatment of newly diagnosed acute promyelocytic leukemia (APL) in children. Blood 2005; 106:447.
  151. Tasian SK, Pollard JA, Aplenc R. Molecular therapeutic approaches for pediatric acute myeloid leukemia. Front Oncol 2014; 4:55.
  152. Kung Sutherland MS, Walter RB, Jeffrey SC, et al. SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML. Blood 2013; 122:1455.
  153. Krupka C, Kufer P, Kischel R, et al. CD33 target validation and sustained depletion of AML blasts in long-term cultures by the bispecific T-cell-engaging antibody AMG 330. Blood 2014; 123:356.
  154. Chichili GR, Huang L, Li H, et al. A CD3xCD123 bispecific DART for redirecting host T cells to myelogenous leukemia: preclinical activity and safety in nonhuman primates. Sci Transl Med 2015; 7:289ra82.
  155. He SZ, Busfield S, Ritchie DS, et al. A Phase 1 study of the safety, pharmacokinetics and anti-leukemic activity of the anti-CD123 monoclonal antibody CSL360 in relapsed, refractory or high-risk acute myeloid leukemia. Leuk Lymphoma 2015; 56:1406.
  156. Wang QS, Wang Y, Lv HY, et al. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther 2015; 23:184.
  157. Nguyen R, Wu H, Pounds S, et al. A phase II clinical trial of adoptive transfer of haploidentical natural killer cells for consolidation therapy of pediatric acute myeloid leukemia. J Immunother Cancer 2019; 7:81.
  158. Björklund AT, Carlsten M, Sohlberg E, et al. Complete Remission with Reduction of High-Risk Clones following Haploidentical NK-Cell Therapy against MDS and AML. Clin Cancer Res 2018; 24:1834.
Topic 13935 Version 28.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟