Pediatric Myeloid Leukemia, Myelodysplasia, and Myeloproliferative Disease


Acute Myelogenous Leukemia

Acute myelogenous leukemia (AML) comprises a heterogeneous group of disorders characterized by the malignant clonal transformation of a hematopoietic stem or progenitor cell. In recent years, deeper understanding has been gained into the chromosomal changes and specific genetic mutations that precipitate this transformation. AML is a relatively rare disorder in children, with between 500 and 600 newly diagnosed patients in the United States each year. Because of the heterogeneous nature of AML, it has proven difficult to find a treatment strategy that is effective for all patients; thus, overall cure rates for AML have improved only modestly in the past few decades, lagging behind those for acute lymphoblastic leukemia (ALL). Drawing from data from the Medical Research Council (MRC) trials, patients with AML are now risk stratified based on cytogenetic features and emerging data on the prognostic significance of minimal residual disease (MRD) early in therapy. Current ongoing clinical trials are attempting to minimize toxicity for patients with low-risk disease and intensify therapy for patients with high-risk disease. As the molecular genetics behind the biology of AML are better elucidated, there is a pressing need to develop new strategies for targeted therapy to improve outcomes in AML.

Epidemiology and Etiology

According to the National Cancer Institute (NCI) Surveillance Epidemiology and End Results (SEER) data, it was estimated that each year, from 2006 to 2010, 5.1 per 100,000 children younger than 14 years and 4.6 per 100,000 adolescents aged 15 to 19 years were diagnosed with cancer. AML is less common than ALL: it comprises about 16% of childhood leukemias in children younger than 15 years but 36% of leukemias in adolescents 15 to 20 years, for a total of 500 to 600 new pediatric AML patients annually in the United States. AML has a bimodal distribution, occurring with higher incidence in children younger than 2 years and then again in adolescents 15 to 20 years old.

In ALL, boys are more commonly affected than girls; the incidence of AML is similar for all pediatric age groups, however, regardless of gender. Similarly, although the incidence of childhood ALL is higher in white than in black children, the incidence of AML is comparable for both races across all pediatric age groups. With regard to ethnicity, there is a higher incidence of AML in Hispanic versus non-Hispanic children. Of interest, although the incidence of ALL for children younger than 15 years increased from 1977 to 1995, the incidence of AML did not change significantly over this time period.

For a subset of patients with AML, a series of risk factors predisposing to the development of AML has been identified ( Box 51-1 ) These risk factors fall into two general categories: exposures (environmental or toxic) and genetic predisposition. The best understood of the exposures is prior treatment with chemotherapy and radiation. Alkylating agents (e.g., nitrogen mustard, cyclophosphamide, ifosfamide, chlorambucil, and melphalan) are associated with the development of myelodysplastic syndrome (MDS) and secondary AML, with deletions of chromosomes 5 and 7. Treatment with topoisomerase II inhibitors such as the epipodophyllotoxins has been linked to a specific type of AML with chromosome band 11q23 translocations, which produce fusion proteins involving the mixed-lineage leukemia (MLL) gene (MLL) . Treatment with anthracyclines in adults with breast cancer, or in children for leukemias and sarcomas such as osteosarcoma, is associated with the development of secondary leukemias, including ALL, AML, and chronic myelogenous leukemia (CML). Although therapy-related AML generally carries a worse prognosis than de novo AML, there have been reported cases of therapy-related AML harboring the better risk cytogenetic abnormalities: t(8;21), inv(16), t(16;16), and t(15;17), often in conjunction with other cytogenetic abnormalities. In the case of therapy-related AML with inv(16) and t(16;16), a rare core-binding factor β (CBFβ) –myosin heavy chain 11 (MYH11) fusion transcript has been reported, with different breakpoints compared with the usual fusion transcript found in better-risk de novo AML. Similarly, there is a case report of a patient with therapy-related AML with t(8;21), and the breakpoint at 21q22 was outside the AML1 locus.

Box 51-1
Predisposing Factors for the Development of Acute Myelogenous Leukemia or Myelodysplastic Syndromes

Prenatal Exposures

  • Alcohol

  • Pesticides *

    * Conflicting or limited data.

  • Foods naturally high in topoisomerase II inhibitors *

  • Viral infections

Environmental Exposures

  • Ionizing radiation

  • Chemotherapeutic agents

  • Alkylating agents

  • Epipodophyllotoxins

  • Anthracyclines

  • Organic solvents (e.g., benzene)

  • Radon *

  • Pesticides *

  • Viral infections *

Hereditary Conditions

  • Down syndrome

  • Noonan syndrome

  • Neurofibromatosis

  • Fanconi anemia

  • Bloom syndrome

  • Severe congenital neutropenia (Kostmann syndrome)

  • Shwachman-Diamond syndrome

  • Klinefelter syndrome (XXY) *

Germline Predisposition Mutations

  • NF1 (for JMML)

  • RUNX1

  • CEBPA

  • GATA2

  • ANKRD26

Acquired Disorders

  • Aplastic anemia

  • Paroxysmal nocturnal hemoglobinuria

JMML, Juvenile myelomonocytic leukemia.

Radiation exposure may be acquired from the environment or through medical procedures and is well documented to predispose to leukemia. The increased risk for AML and CML in survivors of nuclear blasts in Japan who were exposed to excessive doses of ionizing radiation has been well documented, and these patients have a higher rate of abnormalities of chromosomes 5 and 7. An increased risk for AML and ALL in children exposed to radiation in utero has been described, particularly in the past, when diagnostic radiographs were more common during pregnancy, although a retrospective study of children with leukemia in Sweden did not confirm this finding. Nuclear power plant workers have a higher risk for leukemia, although there does not appear to be a higher risk in residents living near such plants. Air travel is a source of exposure to cosmic radiation, and studies have shown an increase in malignant melanoma and other skin cancers in flight crew members, an increase in breast cancer in female flight crew members, and an increase in AML in male cockpit crew members who logged more than 5000 flight hours annually. Studies designed to measure the potential risk for malignancy from residential or occupational exposure to magnetic fields around high-voltage electrical lines have generated conflicting results, and a causal relationship with AML has not been conclusively established.

There are several other environmental exposures with possible links to AML, some of which have yet to be proven definitively. Benzene exposure, which may occur occupationally or through tobacco smoking, has been shown to increase the risk for AML. Several prenatal exposures have been studied as risk factors for the development of AML, particularly in children younger than 3 years. Maternal alcohol consumption during pregnancy was studied in a Children's Cancer Group (CCG) case-control study and found to have an association with increased rates of childhood AML in a dose-dependent matter, with an odds ratio of 2.64 overall and an odds ratio of 7.62 for classic French-American-British (FAB) classification category M1 (myeloblastic with minimal maturation) and M2 (myeloblastic with maturation) AML; other case-control studies have confirmed this finding. Some early studies had suggested an association between maternal tobacco and marijuana use and childhood AML, but more recent analyses have not confirmed this association. A Children's Oncology Group (COG) case-control study looked at maternal consumption of foods such as soy, green and black tea, cocoa, red wine, and certain fruits and vegetables that are naturally high in DNA topoisomerase II inhibitors and found an odds ratio of 1.9 to 3.2 for the development of MLL -rearranged AML in the setting of high maternal consumption of these foods. Some studies have suggested an increased risk for childhood AML after high exposure to pesticides, either prenatally or postnatally, up to 3 years of age. There is interest in an association between viral infections and AML, but few data. One case-control study has suggested an increase in childhood AML in offspring of mothers who reactivated Epstein-Barr virus during pregnancy. Parvovirus B19 is associated with pure red cell aplasia, and certain human leukocyte antigen (HLA)-DRB1 alleles seem to be associated with symptomatic infection. One study of 16 leukemia patients suggested an association between parvovirus B19 infection and acute leukemia in 4 of the patients, including 1 patient with AML, all of whom carried these particular HLA-DRB1 alleles, although this association needs further investigation. Several retrospective studies have suggested that breastfeeding may be protective against ALL and AML. Risk related to indoor radon exposure is controversial. One French study showed a higher rate of AML in children with high exposure but other studies have showed no association. Rarely used now, chloramphenicol use in children was formerly associated with an increased risk for AML.

An increased incidence of AML has been observed for patients with certain hereditary disorders. Children with congenital disorders of myelopoiesis, such as Kostmann syndrome, Shwachman-Diamond syndrome, and Diamond-Blackfan anemia, are predisposed to AML. Patients with inherited syndromes associated with chromosome fragility and impaired DNA repair mechanisms, such as Fanconi anemia and Bloom syndrome, also have an increased risk for development of AML. Neurofibromatosis type 1, which is caused by mutations in the neurofibromin tumor suppressor gene on chromosome 17, is associated with juvenile myelomonocytic leukemia (JMML). Patients with certain constitutional chromosomal abnormalities carry a higher risk for AML. Down syndrome is one of the most clinically prominent examples in this category. It is estimated that over 10% of infants with Down syndrome exhibit a transient myeloproliferative disease associated with a GATA1 mutation. Down syndrome patients have a 10 to 20 times higher than average risk for acute leukemia. In Down syndrome patients younger than 4 years, AML, usually acute megakaryoblastic (AMKL), is far more common than ALL: an estimated 1 in 500 Down syndrome patients develop AML. In contrast to AMKL in the general pediatric population, which carries an extremely poor prognosis, this disease in Down syndrome patients is extremely sensitive to chemotherapy (see later). Although early studies did not suggest a higher risk for AML in patients with Klinefelter syndrome (XXY), more recent data suggest that these patients may have a higher risk for hematologic malignancy, including AML. More recently, a number of MDS/AML predisposition genes have been identified, including loss of function alterations affecting the key hematopoietic transcription factor genes RUNX1, CEBPA , and most recently GATA2 .

Although the predisposing risk factors and genetic conditions described may provide insight into the causative mechanisms that increase the risk for development of AML, most patients with de novo AML have no known predisposing exposures or conditions. Point mutations and chromosomal deletions and translocations occur at a background rate, even in healthy individuals, during hematopoietic stem and progenitor cell expansion. The extent to which inherited, expressed, single-nucleotide polymorphisms in the general population alter mutational rates during myelopoiesis and increase the risk for AML and other cancers remains an area of intense investigation.

Biology

Clonal Origin of Myeloid Leukemia Cells

Normal myelopoiesis is a complex differentiation program whereby primitive hematopoietic stem cells (HSCs) develop along a multistep pathway into fully differentiated, functionally active circulating blood cells. This exquisitely controlled process is regulated by the intricate interactions among the expression levels of various transcription factors, growth factors and their receptors, cytokines, enzymes, and still unidentified novel molecules. A series of sequential genetic abnormalities perturbs this normal developmental progression and leads to AML. Although the relationships between specific morphologic subtypes of AML and their specific recurring genetic abnormalities have provided some insight into the mechanisms of leukemogenesis, understanding of the process whereby these fusion gene products interact with normal signal transduction pathways to subvert hematopoietic cell development is incomplete.

Several strong lines of evidence support the hypothesis that AML progresses from a single transformed hematopoietic stem or progenitor cell. More than 30 years ago, studies were pioneered by Beutler and colleagues and Fialkow using X inactivation patterns in female patients to establish the clonal origin of human malignancies, including leukemia. By showing the presence of a single glucose-6-phosphate dehydrogenase (G6PD) isoenzyme in the leukemic myeloblasts of heterozygous females, the clonal origin of myeloid leukemia cells was demonstrated. Females who are heterozygous at the G6PD locus express two isoforms of the enzyme. Approximately half of the cells in normal somatic tissue have randomly inactivated one of the X chromosomes, and approximately half of the cells should therefore express each isoform. Unlike normal somatic cells, AML cells of female patients heterozygous for G6PD expressed only one G6PD isoform, indicating cells of clonal origin. In the 1980s, Vogelstein and coworkers developed a strategy using X chromosome–linked DNA restriction length fragment polymorphisms to determine the clonal origin of human tumors. They established that maturing granulocytic cells arise from the malignant clone in patients with AML. Later, X chromosome inactivation to determine clonality in malignancies was carried out using the polymerase chain reaction (PCR) assay to distinguish X-linked polymorphic genes.

Transformation of Stem Cells with Self-Renewal Capacity

These earlier studies, and those performed by Bonnet and Dick and associates in the 1990s, led to the insight that leukemia cell populations form a hierarchy in which leukemia stem cells or leukemia-initiating cells (LICs) are responsible for their own self-renewal and for the generation of the more differentiated progeny within the leukemic clone. This theory was in contrast to the stochastic model, which stated that malignant properties within cancer cell populations follow Gaussian distributions, resulting in the random or stochastic probability that individual cancer cells within the population are able to initiate malignant progression. Whereas AML was the first human cancer in which cancer stem cells were identified, the case has now been made in breast cancer, brain tumors, and colon cancer, among others. In general, AML stem cells are believed to arise through the transformation of HSCs, which then retain the capacity for self-renewal, or through transformation of more differentiated hematopoietic progenitor cells that acquire the ability to self-renew by virtue of the particular transforming mutations, such as the formation of an MLL fusion gene.

AML is a heterogeneous disease in its clinical manifestations, response to therapy, and molecular genetics; thus insight into the cell of origin would have important ramifications regarding diagnosis and treatment. Moreover, refractory disease and relapse are often attributed to the presence of an LIC population that is resistant to chemotherapy; thus the ability to identify and target this subpopulation may be key to improving overall disease outcome. In the 1990s, Dick's group produced a series of convincing papers that strongly implicated the primitive, pluripotent HSC as the LIC in some types of AML. “Stemness,” or the capacity to function as an LIC, includes self-renewal, proliferation, and differentiation and is tested in vitro by replating limiting dilution studies and in vivo by serial transplantation assays with the ability to regenerate a clonal leukemia in secondary and subsequent recipient animals. The HSC, with its inherent gene programs ensuring self-renewal, proliferation, and survival appears to be an ideal target to become sabotaged through a series of genetic events, leading ultimately to a fully transformed AML LIC that is capable of producing a clonal population of leukemia cells. According to this argument, different AML phenotypes often occur as a result of the gene(s) that are altered, leading to differentiation arrest within the progeny of the LIC, but not necessarily reflecting the degree of the commitment of the initially transformed cell. Support for this hypothesis initially came from several clonality studies in leukemic cells from patients with AML, which demonstrated multiple-lineage involvement in a high proportion of cases, and implicated a multipotent HSC as the cell of origin. More supportive evidence has come from studies looking at cytogenetic markers and characteristic cell surface antigen expression patterns. Pluripotent HSCs express CD34 but do not express CD38 or HLA-DR, whereas more committed myeloid progenitor cells are CD34+, CD38+, and HLA-DR+. Using fluorescence-activated cell sorting (FACS) of leukemic cell populations, the same two subpopulations, CD34+/CD38− and CD34+/CD38+, were isolated from the bone marrow of patients with different AML subtypes. The two subpopulations were then evaluated using fluorescence in situ hybridization (FISH) to determine the presence of cytogenetic abnormalities. The studies detected the same characteristic cytogenetic abnormalities in the CD34+/CD38− stem cell fraction as in the original leukemic bone marrow samples, implying origin in a very early HSC compartment.

Further evidence supporting the stem cell origin model of AML was derived from transplantation experiments in which purified human AML cells were transplanted into mice with severe combined immunodeficiency (SCID). These experiments defined a SCID mouse leukemia–initiating cell (SL-IC) in the bone marrow of patients with AML and showed that these SL-ICs are CD34+/CD38− and that their engraftment produces large numbers of colony-forming progenitors. The CD34+/CD38+ and CD34− fractions did not engraft. Similar results were obtained through transplantation studies in a modified SCID mouse, the nonobese diabetic mouse with SCID (NOD/SCID), with cells from patients with AML. These studies identified and analyzed AML stem cells from samples of different FAB subtypes on the basis of their ability to initiate human AML after transplantation in NOD/SCID mice. These SL-ICs were able to proliferate and differentiate after transplantation, producing disease in the mice identical to that in the donor, as well as being able to renew themselves, reestablishing AML in secondary recipients, demonstrating that SL-ICs are capable of self-renewal. Again, the SL-ICs were found to reside in the CD34+/CD38− fraction and not in the CD34+/CD38+ or CD34− fractions. The SL-IC phenotype was consistent regardless of the FAB subtype (e.g., M1, M2, M4, M5). As few as 2 • 10 4 CD34+ cells were able to initiate the leukemic clone in recipient mice, whereas 100 times as many CD34− cells failed to engraft. Cells with the CD34 surface antigen were further fractionated on the basis of CD38 expression. Only the CD34+/CD38− fraction contained SL-ICs.

Whereas the transformation of the HSC provides an attractive mechanism to generate hierarchical myeloid leukemia cell populations, several studies have given new credence to a second mechanism through which committed progenitor cells along the pathway of myeloid differentiation are vulnerable to transforming mutations that confer self-renewal properties to progenitor cells that inherently lack this ability. These transformation events create an LIC capable of generating a population of aberrant cells blocked at the differentiation state at which the initial transforming event occurred. Several translocations involving the MLL gene have been shown to initiate self-renewal in myeloid progenitor cells and to result in an AML phenotype. A head-to-head comparison was conducted by retrovirally transducing the human AML-associated MLL-ENL fusion gene into murine HSC, common myeloid progenitors (CMPs), and granulocytic-monocytic–restricted progenitors (GMPs) and evaluating the relative transforming ability and resultant phenotype in each cell type. AML developed in mice transplanted with any of the three transduced cell populations and, in each case, displayed an identical myelomonocytic phenotype as assessed by morphology and flow cytometry. Similarly, the MOZ-TIF2 fusion gene was able to confer self-renewal properties to committed myeloid progenitor cells and resulted in AML when transplanted into irradiated mice. Although the fusion genes MLL-ENL and MOZ-TIF2 appear to impart leukemogenic capabilities to committed progenitor cells convincingly, other fusion genes such as BCR-ABL can induce leukemic transformation only when expressed in an HSC but not a more mature myeloid cell. Similarly, the potent oncogenic combination of Hoxa9 and Meis1 only resulted in AML when transduced into HSCs but not in more committed myeloid progenitors. In this case, expression of β-catenin was found to be the necessary factor for oncogenesis. Present in HSCs but absent in more committed progenitors, the addition of β-catenin to progenitors transduced with Hoxa9 and Meis1 was sufficient to lead to AML. These findings point to the differing transforming abilities of different oncogenes that are likely cell-type and cell-context specific. Hence both cell-of-origin hypotheses appear to be correct, depending on the specific oncogene or tumor suppressor that provides the initiating genetic lesion. Increased proliferation, survival, and self-renewal are all necessary attributes of a leukemia stem cell. Some genetic abnormalities, such as MLL-ENL and MOZ-TIF2 , may be capable of inducing self-renewal and thus are able to transform a more differentiated myeloid cell into an LIC. By contrast, other oncogene fusions, such as BCR-ABL , or oncogenic transcription factor genes, such as Hoxa9 , do not possess self-renewal capabilities and thus require a cell that inherently has this characteristic, namely, the HSC.

More recently, challenges to the cancer stem cell hypothesis have arisen out of a number of studies, predominantly in solid tumors such as melanoma, where the frequency of cells capable of giving rise to a new tumor in an immunocompromised host is of sufficient frequency to call into question whether the LIC represents a unique subpopulation or the original stochastic model of cancer progression holds true. These studies have again called into question previously accepted tenets. CD34+/CD38+ populations, in addition to the CD34+/CD38−, have also been found to contain LICs when injected into more immune-deficient hosts that lack key components of the innate immune system. These studies and additional syngeneic and cogenic transplant studies in mice as well as zebrafish highlight the critical context of the recipient animal in determining the LIC. The heterogeneity of LICs in AML is further demonstrated by an increasing number of cell surface markers in addition to CD34 and CD38, including CD123, TIM3, and CD47. Additionally, a number of studies have attempted to identify a specific LIC genetic signature, which has been found at least in some cases to independently predict prognosis. In this case, the LIC signature was reminiscent of the normal HSC, hearkening back to the original predictions of Dick's group highlighting the HSC as a putative LIC cell of origin. However, although these findings are provocative, a consistent cadre of LIC genes has not yet been established. Clonality studies demonstrating the progressive acquisition of mutations in a given cell have also been put forward as an alternative to the cancer stem cell hypothesis. Although seemingly at odds, these different concepts can be reconciled into a single integrated framework. Myeloid (and lymphoid) leukemia-initiating events may occur in both HSCs and committed progenitor cells, and each leukemia arises from a combination of multiple genetic abnormalities. Secondary events subsequently are acquired in different subpopulations, generating unique clones with varying malignant potential depending on the genetic lesion and the self-renewal capacity of the cell in which these abnormalities occur. The number of genetic lesions necessary to cause frank AML is being defined based on next-generation sequencing studies recently published by the group at Washington University in St. Louis as well as other centers. The evolution of particular clones appears to determine the eventual phenotype of the AML cells documented at diagnosis ( Fig. 51-1 ).

Figure 51-1, Integration of the leukemic initiating cell (LIC) and clonal evolution models of acute myelogenous leukemia pathogenesis.

These new discoveries have added additional complexity to our understanding of the contribution of LICs to AML pathogenesis by demonstrating that the heterogeneity inherent in this disease extends to the AML stem cells. However, the identification of new LIC surface markers, genetic signatures, and gene-specific mutations hold out promise that LIC-targeted therapies will be realized in the coming years.

Molecular Genetics

Recent AML protocols have incorporated current knowledge regarding the significance of specific genetic abnormalities to direct risk-stratified approaches to treatment. Chromosomal abnormalities and, in particular, translocations identified on cytogenetic examination of the blast cells represent the initial genetic lesions incorporated into disease taxonomy and formed the basis of the World Health Organization (WHO) classification in 2008. Careful analysis of these translocations and other recurring genetic abnormalities in leukemia patients has had a profound impact on our understanding of the molecular genetic basis of leukemia ( Fig. 51-2 ). Although many chromosomal abnormalities from AML blast cells have been identified and studied extensively, no single mutation has been shown to be sufficient to cause acute leukemia. These findings prompted Gilliland and colleagues to propose what is now considered the classic “two-hit model” of AML pathogenesis. In this model, leukemic transformation results from distinct but collaborative sequential mutations in parallel molecular pathways that affect cell survival, proliferation, differentiation, and self-renewal. Class I mutations frequently involve an activated receptor or cytoplasmic-nuclear tyrosine kinase conferring a proliferative and/or survival ability to a specific cell but not affecting differentiation. By contrast, class II mutations specifically result in differentiation arrest and/or self-renewal and often involve key transcription factor oncogenes but alone do not confer a proliferative advantage. Although attractive, this model likely represents an oversimplification of AML pathogenesis. Initially, microarray studies and more recently, deep-sequencing approaches have revealed the presence of novel mutations and absence of tyrosine kinase lesions, particularly in normal karyotype AML, calling into question the general applicability of the “two-hit” model in all cases of AML. However, although the category of genetic lesions may not always subscribe to this classic paradigm, the number of “hits” required may be more universal. Genomic profiling of individual patient leukemias suggest that in some cases only two to three lesions are required for clonal evolution to frank AML, with fewer “hits” necessary in pediatric AML than in its adult counterpart. Regardless, the prevalence of lesions categorized by the “two-hit” model will be outlined here, followed by more recent discoveries of novel genes implicated in AML pathogenesis and the variable frequency of these lesions in pediatric versus adult AML.

Figure 51-2, Approximate frequencies of recurrent genetic lesions in childhood acute myelogenous leukemia.

Class I Mutations

Among the earliest examples of class I mutations, RAS mutations were described in the early 1980s. There are three functional RAS gene family members— NRAS, KRAS, and HRAS —that all act as guanosine nucleotide phosphate (GTP)-binding proteins. Mutations of NRAS are the most prominent RAS mutations in AML and have been found in as many as 30% of adult patients with AML. Mutations, most frequently in codons 12, 13, and 61, result in the retention of the RAS protein in its active GTP-bound state and lead to the constitutive activation of downstream effector proteins, which causes the transcriptional activation of various target genes that direct cellular differentiation, proliferation, and survival ( Fig. 51-3 ). RAS proteins may be alternatively activated by mutations in genes encoding a number of regulatory factors. Activating point mutations in the SHP-2/PTPN11 phosphatase provide a stimulatory signal through guanine nucleotide exchange factors, such as “Son of Sevenless” (SOS), resulting in increased RAS pathway signaling, as do inactivating mutations of neurofibromin-1 (NF1) , which codes for a GTPase-activating protein (GAP) of the same name. NF1, normally functions, at least in part, by increasing the GTPase activity of RAS, thereby inactivating RAS-GTP by converting it into the inactive RAS-GDP form. These three types of mutations function independently to activate RAS signaling and lead to disturbances in hematopoietic cell differentiation and proliferation that may ultimately contribute to the development of MDS, myeloproliferative disease (MPD), and AML (see Fig. 51-3 ). Mutations in NF1 and PTPN11 have been particularly associated with JMML (see later).

Figure 51-3, Constitutive activation of the RAS pathway is central to myeloid leukemogenesis.

There has been significant disagreement regarding the prognostic implications of RAS mutations in MDS and AML. Several studies have demonstrated that RAS mutations in patients with MDS confer a poor prognosis and increase the risk for progression to acute leukemia. Other studies have reported no difference in survival in AML patients who carried RAS mutations compared with patients without RAS mutations, whereas some have suggested improved survival in patients with AML and NRAS mutations. A large study of more than 2500 patients with AML demonstrated no difference in prognosis in patients with or without RAS mutations. This study also demonstrated an association between RAS mutations and a leukemia karyotype containing inv(16), a class II mutation resulting in the CBFβ-MYH11 fusion protein and altered function of the core-binding factor (CBF) transcriptional complex. A recent report from the COG examining 825 pediatric AML samples treated on two recent trials identified NRAS mutations in 10% of patients predominantly in codons 12 and 13, with no mutations in codon 61. Interestingly, most of these mutations were associated with good prognosis NPM1 mutations (see later) and not CBF alterations. However, multivariate analysis confirmed a lack of independent impact of NRAS mutations on prognosis.

Another group of class I mutations involves constitutive activation of receptor or cytoplasmic-nuclear tyrosine kinases. The BCR-ABL translocation that is the sine qua non of CML is an example of a class I mutation causing the activation of the ABL tyrosine kinase. Similarly, receptor tyrosine kinase mutations activate the human protooncogenes KIT and FLT3 in AML. The KIT protein is activated by the binding of its ligand stem cell factor and is critical for the development and growth of mast cells, melanocytes, HSCs, and the interstitial cells of Cajal. Mutations in KIT allow ligand-independent activation of KIT and confer factor-free growth and tumorigenicity in hematopoietic cell lines. Blasts from patients with AML express KIT on their cell surfaces in most cases. Deletional and insertional mutations of KIT have been identified in the blast cells of patients with AML, often in association with the CBF abnormalities inv(16) or t(8;21).

The FLT3 gene encodes a class III fms-like receptor tyrosine kinase expressed in early hematopoietic progenitors. FLT3 mutations occur in two varieties, internal tandem duplications (ITDs) in the juxtamembrane region and activation loop mutations (ALMs), primarily at codons 835 and 836. The FLT3 -ITD is one of the most frequent abnormalities in adult AML, documented in 25% to 30% of patients, and generally confers a poor prognosis. By contrast, ALMs occur in only 10% of adult patients and have not been associated with inferior survival rates. FLT3 -ITDs have been observed in 5% to 16% of pediatric AML cases and ALMs in 2% to 5% of cases. Paralleling the adult studies, FLT3 -ITDs, but not ALMs, were associated with a poor prognosis in children. FLT3 -ITDs result in the constitutive activation of FLT3 and cause interleukin-3–independent growth in Ba/F3 and 32D cells. This activation, in turn, results in the signaling of multiple downstream pathways, including the aforementioned RAS pathway, and the inhibition of caspase-mediated apoptosis through the stimulation of phosphatidylinositol 3′-kinase (PI3K) and AKT (see Fig. 51-3 ). However, mice transplanted with FLT3-transformed hematopoietic cells develop a myeloproliferative disorder characterized by splenomegaly and leukocytosis but they do not develop AML. FLT3 -ITDs often occur in conjunction with other mutations, including NPM1 (see later) and WT1 (see later).

Class II Mutations

In class II mutations, the molecular defect appears to be at the level of transcriptional activation, whereby transcriptional repressors cause a dominant negative inhibition of normal hematopoietic cell differentiation. AML can be subtyped based on the type of class II mutations in the blast cells of patients. The type 1 subset of class II mutations occurs in patients with de novo AML. These are typical chromosomal translocations resulting in chimeric oncoproteins that in some cases may cause the inhibition of differentiation. The type 2 subset of class II mutations usually are found in patients with AML arising after a prodrome of MDS and manifest more commonly in older adults or in patients who have received previous chemotherapy. Cytogenetic studies of the blast cell populations in these patients have complex karyotypes, lacking translocations but harboring deletions such as 5q−, monosomy 7, and 20q−.

Type 1 Mutations

Translocations Involving Core-Binding Factor Complex.

Several of the translocations that have been identified in adult and childhood de novo AML involve the CBF complex. The CBF complex is the most frequent target of chromosomal translocations in the human leukemias. The CBF regulatory complex consists of a DNA-binding subunit, RUNX1 (also called CBFα or AML1) and CBFβ, a subunit that does not bind DNA independently but heterodimerizes with RUNX1 or one of its closely related family members. Chromosomal translocations that modify the CBF complex in AML include the following: t(8;21)(q22;q22), which generates the AML1-ETO fusion protein, more recently referred to as RUNX1-RUNX1T1 ( Fig. 51-4 ); t(3;21)(q26;q22), which gives rise to AML1-EVI1; and inv(16)(p13;q22), which fuses the CBFβ to smooth muscle myosin heavy chain (SMMHC), resulting in the CBFB-MYH11 fusion gene. The AML1 gene was inactivated in the germline of mice and was shown to be essential for definitive hematopoiesis of all cell lineages. Homozygous animals die early in embryogenesis of central nervous system (CNS) hemorrhage, but they have normal morphogenesis and yolk sac hematopoiesis lineages. Inactivation of the CBFβ gene in the mouse has shown a similar phenotype in the homozygous null mice. These experiments have demonstrated that the AML1/CBFα complex is essential for normal hematopoiesis and that chromosomal rearrangements involving this complex may interfere with its regulatory function in ways that lead to disruption of cellular differentiation, and eventually malignant transformation.

Figure 51-4, Consequences of the t(8;21) AML1-ETO (RUNX1-RUNX1T1) translocation.

Evidence primarily from knock-in studies in mice has supported the hypothesis that the AML1-ETO fusion protein functions as a dominant negative inhibitor of wild-type functions. In these experiments, the AML1-ETO fusion gene is “knocked in” to the wild-type AML1 locus. The phenotype of these mice is identical to the AML1 knockout; the mice die early in embryogenesis from CNS hemorrhage and lack any evidence of definitive hematopoiesis, although the rare cells that survive and express AML1-ETO have an increased capacity for self-renewal. Biochemical experiments have shown that the ETO portion of the AML1-ETO chimeric protein recruits nuclear corepressor complexes to the CBF promoters, resulting in transcriptional inhibition of target genes normally activated by the AML1/CBFβ heterodimeric complex (see Fig. 51-4D ). Subsequent studies using inducible murine model systems to bypass the embryonic lethality of AML1-ETO overexpression have demonstrated that overexpression of this transgene at later developmental time points confers enhanced self-renewal capacity to bone marrow progenitors, as demonstrated by serial culture experiments. Importantly, the mice do not develop AML but appear to be predisposed to leukemogenesis. Thus after treatment with N -ethyl- N -nitrosourea (ENU), the mice developed rapid onset of AML compared with similarly exposed wild-type mice. Moreover, as noted, there is an association in myeloid leukemia of AML1-ETO translocations and mutant tyrosine kinases, such as KIT. Interestingly, AML1-ETO transcripts may persist for a long time in patients with AML1-ETO –expressing AML in sustained remission.

Translocations Involving the Retinoic Acid Receptor-α.

Reciprocal chromosomal translocations involving the retinoic acid receptor-α (RARA) gene locus on chromosome 17 are the defining molecular features of acute promyelocytic leukemia (APL). Before the RARA translocations were described, retinoic acid was known to induce myeloid differentiation in vitro and therapy with all- trans -retinoic acid (ATRA) in patients with APL was shown to induce complete remission (CR). The intense investigation of the RARA gene triggered by these observations culminated in identification of the promyelocytic leukemia PML-RARA translocation breakpoint, t(15;17)(q22;q11), reported simultaneously by several groups. Although most APL cases have the associated t(15;17), other reciprocal translocations involving the RARA gene have been identified in which the gene is fused to other gene partners. The most common of these include the promyelocytic leukemia zinc finger (PLZF) gene on chromosome 11 in t(11;17)(p13;q11) and the nucleophosmin gene (NPM) on chromosome 5 in t(5;17)(q31;q11).

Normally RARA functions as a ligand-dependent activator of transcription. It forms a heterodimer with the related protein, retinoid X receptor (RXR), and binds through its zinc finger domain to a specific promoter sequence; in the presence of the retinoic acid ligand, the RARA-RXR heterodimer activates transcription of retinoic acid–responsive genes. This process is accomplished in concert with a coactivator complex that includes proteins such as p300 and pCAF. In the absence of retinoic acid, a corepressor complex is recruited, composed of N-CoR (nuclear receptor corepressor) or SMRT (silencing mediator of retinoid and thyroid receptors), which binds to another protein called Sin3 and then to HDAC1. HDAC1 is a histone deacetylase protein, which epigenetically alters histones to keep DNA in an untranscribable form. The expression of retinoic acid–responsive genes is essential for normal myeloid development.

Much evidence supports the hypothesis that the PML-RARA fusion protein functions as a dominant negative inhibitor of the PML protein and RXR. PML proteins are normally located in macromolecular nuclear organelles, called PML oncogenic domains (PODs). The PML-RARA fusion protein disrupts the PODs, causing normal PML, RXR, and other nuclear proteins to disperse in an abnormal pattern. ATRA and arsenic trioxide (As 2 O 3 ) have shown activity in APL blast cells and in patients with APL, and studies of these drugs have afforded important insights into the mechanisms of leukemogenesis in APL. ATRA and As 2 O 3 appear to act through different biochemical pathways, and promyelocytes resistant to ATRA are often sensitive to treatment with As 2 O 3 . Both drugs induce degradation of the PML-RARA fusion protein but through different mechanisms. Retinoic acid binding to wild-type retinoic acid receptors (RARs) in the cell nucleus causes degradation of the PML-RARA protein through the ubiquitin-proteosome and caspase systems, thus allowing for the terminal differentiation of leukemic promyelocytes. This binding also results in derepression whereby N-CoR is dissociated from the PML-RARA fusion protein and, subsequently, the nuclear coactivator complex is recruited to reverse histone deacetylase-mediated repression. PODs are relocated back to the normal nuclear pattern. By contrast, As 2 O 3 appears to target the PML portion of the PML-RARA fusion protein preferentially and causes its degradation by inducing apoptosis, potentially through downregulation of the antiapoptotic factor BCL2. The activity of these agents has led to the inferences that fusion proteins may interfere with normal myeloid cell development in several ways—through inhibitory effects on assembly of the PODs that contain PML or through dominant inhibitory effects on transcriptional targets of dimeric complexes with normal retinoid receptors, leading to arrest of differentiation at the promyelocyte stage.

Studies using transgenic mice have shown that the PML-RARA fusion product is involved in the development of leukemia. Transgenic mice generated with the PML-RARA fusion protein specifically expressed in the myeloid-promyelocytic lineage develop a myelodysplastic-like disorder during the first year of life. A subset of these mice then develops a form of acute leukemia that closely mimics human APL and responds to ATRA. Given the relatively long latency period and the development of leukemia in only a fraction of the mice, it is likely that the PML-RARA translocation is insufficient by itself to cause APL and that second mutations are necessary for leukemic transformation.

Mixed-Lineage Leukemia Gene Translocations.

Transcriptional coactivators and corepressors have been implicated in leukemogenesis. An example is the myeloid-lymphoid or MLL protein, a large protein containing transcriptional activation and repression domains, which is believed to act epigenetically to modulate gene expression through its effects on chromatin structure and configuration. Wild-type MLL is required for normal hematopoiesis and HSC development. Mice deficient in MLL die on embryonic day 10.5 and have numerous skeletal, neural, and hematologic deficits. Heterozygous mice have defects in embryonic segmentation and yolk sac hematopoiesis, and adults demonstrate a mild anemia and thrombocytopenia. Chromosomal translocations of the MLL gene on chromosome segment 11q23 have been identified in human AML and ALL and confer a poor prognosis. Many gene partners have been identified that are involved in these translocations, including AF4, AF9, ENL, AF6, ELL , and AF10 . In most cases, the fusion proteins incorporate the 5′ end of MLL and the 3′ end of the partner. Despite the number of different fusion partners, leukemias with MLL translocations tend to possess a distinct genetic signature compared with AML and ALL samples lacking an MLL translocation. This genetic signature includes the upregulation of a number of specific HOX family genes, including HOXA9, HOXA7, and MEIS1 . Wild-type MLL is known to play a major role in HOX gene transcriptional regulation. In turn, HOX genes play critical roles in embryonic development and normal hematopoiesis. A number of studies have clearly demonstrated that overexpression of specific major HOX genes in mouse models and human bone marrow samples, whether by involvement in reciprocal translocations or by gene upregulation, is linked to the transformation of malignant HSCs in AML. The mechanism whereby MLL fusion proteins upregulate HOX gene expression may differ, depending on whether the fusion partner encodes a nuclear partner with direct transcriptional activity or a cytoplasmic protein that results in MLL protein dimerization and subsequent transcriptional activation.

Transcriptional control may be mediated through the modification of histone proteins, which function to maintain chromatin structure. Recently, the DOTL1 complex was identified as a critical factor in MLL-induced leukemogenesis. DOT1L is a histone-3-lysine-79 (H3K79) methyltransferase, which was previously believed to play a fairly ubiquitous role in methylating gene targets in concert with transcription. However, Zhang and Armstrong's group definitively demonstrated through a series of elegant in vitro and in vivo experiments a specificity for DOT1L in the epigenetic regulation of MLL-translocated downstream targets. When DOT1L was knocked out or absent from MLL-AF9 –transformed cell lines or transgenic mice, leukemia was prevented. The MLL gene is rearranged in up to 20% of pediatric cases of AML. Translocations of MLL are the single most common genetic alteration in infants with acute leukemia, regardless of phenotype, and account for approximately 70% of all cases of AML and ALL in infants. In pediatric and adult cases of AML, the presence of the 11q23 translocation is generally associated with an unfavorable prognosis.

Although less common than MLL translocations, the MLL gene can be altered by partial tandem duplications (PTDs) of particular exons. These PTDs occur in adult and pediatric AMLs with a normal karyotype, as well as those with trisomy 11. Similar to MLL fusions, the MLL -PTD tends to be associated with a poor prognosis and often with the presence of an FLT3 mutation. The mechanisms underlying MLL-PTD leukemogenesis have not yet been elucidated but appear to be different than those of MLL fusions. Interestingly, MLL -PTD appears to repress wild-type MLL expression, which may be an important feature underlying the leukemogenic potential of this abnormality.

Type 2 Mutations.

The leukemogenic mutations discussed in the previous sections involve balanced translocations, duplications, or substitutions that appear to disrupt regulatory mechanisms controlling hematopoiesis and result in dysregulated cell differentiation. Another group of mutations involve unbalanced chromosome rearrangements, such as chromosome loss (e.g., monosomy 5, monosomy 7) or large chromosomal deletions (e.g., 5q−, 7q−, 20q−). These mutations may lead to leukemic transformation by the loss of a tumor suppressor gene (or genes), which then confers a differentiation block and growth advantage to the mutated cells. Identification of these putative tumor suppressor genes has been problematic because the deleted regions are usually large and include many candidate genes. Moreover, leukemogenesis is triggered by the gene's inactivation rather than by altered structure, making the abnormal gene difficult to identify. However, α-catenin has been identified by our group as a novel tumor suppressor gene on the proximal region of the long arm of chromosome 5. Non–therapy-related AML with these chromosomal losses or deletions occurs most commonly in older patients and is often associated with the prodrome of MDS. In the pediatric population, this class of cytogenetic abnormality is uncommon and is also frequently preceded by MDS. Monosomy 7 is the most frequent chromosomal abnormality documented in children with MDS and is sometimes the sole detectable cytogenetic abnormality. Overall, chromosome loss represents about half of the chromosomal abnormalities identified in MDS.

Therapy-Related AML.

Chromosomal loss and large chromosomal deletions are common in therapy-related AML in the adult and pediatric populations. Although most cases of MDS and AML arise de novo, without evidence of any leukemogenic exposure, in 10% to 20% of patients the disease arises after previous exposure, particularly to topoisomerase II inhibitors, alkylating agents, and ionizing radiation. The 11q23 translocations producing MLL gene fusions are common cytogenetic abnormalities in patients who develop AML after therapy with topoisomerase II inhibitors (e.g., epipodophyllotoxins). AML developing after the use of topoisomerase II inhibitors has a short median latency period of 30 to 34 months, is usually FAB classification M4 or M5, and lacks the antecedent prodrome of MDS. The blast cells of patients with AML or MDS occurring after exposure to alkylating agents often have large chromosomal losses or deletions and have a poor prognosis. The most common chromosomal abnormalities in such cases involve chromosome 7 and chromosome 5. In contrast to the therapy-related AML with MLL fusion genes that develops after treatment with topoisomerase II inhibitors, MDS or AML related to therapy with alkylating agents typically develops after a longer median latency period, 3 to 5 years from the alkylator exposure. Mutations in the TP53 tumor suppressor gene have been previously linked to therapy-related AML and were believed to be infrequent in de novo cases. However, more recently, TP53 mutations have been found to be common in de novo AML with a complex karyotype. In both contexts there is an association with chromosome 5 and 7 copy number alterations.

AML with a Normal Karyotype.

Chromosomal abnormalities have provided some understanding of the mechanisms of malignant transformation in leukemia. However, in many cases of de novo leukemia, no chromosomal abnormality can be detected and the mechanism behind leukemogenesis in these cases is unknown. Because AML involves a block in myeloid differentiation, evidence for mutations in key transcription factors involved in normal myelopoiesis has been sought. Mutations in the early myeloid transcription factors PU.1 and CCAAT/enhancer-binding protein α (C/EBPα) have been identified in a number of AML subtypes and are generally not associated with a known chromosomal translocation. Mouse models have demonstrated that the degree of transcription factor knockdown is critical to the particular phenotype. Mice expressing PU.1 at 20% of normal levels develop AML, whereas those with complete knockdown or 50% of normal levels do not. GATA1, a critical transcription factor for erythrocyte and megakaryocyte development, is mutated in nearly all cases of acute megakaryoblastic leukemia associated with Down syndrome (see later).

Expression profiling using microarrays first used to distinguish prognostic tumor subclasses in breast carcinoma and large B-cell lymphoma has also been applied to leukemia. Using this technology, a set of 50 genes was sufficient to discriminate ALL from AML. More recent validation of this technique in AML has demonstrated the correlation between gene expression profiling studies and the prognostic classification of human AML based on cytogenetic abnormalities. Gene expression profiling has provided valuable new insights into the identification of prognostic subclasses in adult and pediatric AML with a normal karyotype.

Our understanding of the molecular mechanisms underlying de novo AML with a normal karyotype was advanced by the finding of mutations involving the NPM gene in 35% of cases of adult AML and 60% of cases with a normal karyotype. This frequency supersedes the incidence of FLT3 mutations and has established NPM mutations as the most frequent genetic abnormality in adult AML. The frequency of NPM mutations is lower in pediatric AML, occurring in only 6% to 8% of all pediatric patients with AML, but also occurs in a higher number of pediatric patients with a normal karyotype (approximately 27%). The NPM gene on chromosome 5q was previously well known as a partner in a number of oncogenic translocations, including NPM-ALK (anaplastic lymphoma kinase) in anaplastic large cell lymphoma and NPM-RARA in APL. NPM is a molecular chaperone that shuttles between the cytoplasm and nucleus but is found most prominently in nucleoli. It has been shown to function in the prevention of protein aggregation in the nucleolus and the regulation of preribosomal particles through the nuclear membrane. It has also been shown to play a role in the regulation of the alternate reading frame (ARF)-TP53 tumor suppressor pathway. NPM mutations are heterogeneous but occur predominantly in exon 12, uniformly affecting the C-terminal of the protein and causing the formation of a neomorphic nuclear export signal in the resulting NPMc protein. This results in a shift of NPM localization, sequestering it exclusively to the cytoplasm. NPM mutations occur more frequently in children older than 10 years and, as is the case in adults, tend to be associated with a good prognosis. However, there has been a strong association established between patients with NPM mutations and FLT3 -ITDs, which portend a poor prognosis. Patients with both these abnormalities do not fare as well as those with NPM mutations alone.

Similarly, mutations in CEBPA have been associated with a good outcome. CEBPA encodes the CCAAT/enhancer binding protein-alpha (C/EBPα), which functions as a transcription factor that plays a major role in terminal granulocytic differentiation as well as influencing proliferation. C/EBPα is a basic region leucine zipper transcription factor consisting of two amino-terminal transactivation domains, a basic DNA-binding region, and a carboxyl-terminal leucine zipper. Mutations in pediatric AML commonly occur in the amino-terminal domain and bZip domain of the leucine zipper, and many patients harbor both types of mutations.

Mutations in the Wilms tumor 1 (WT1) transcription factor have been identified in approximately 10% of adult and childhood AML, predominantly in patients with a normal karyotype. Mutations are common in exons 7, 8, and 9, comprising three of the four zinc fingers of WT1 . WT1 mutations frequently occur together with FLT3 -ITDs; however, in contrast to the adult studies, they do not independently confer a poor prognosis. Recently, a number of large genomic studies using next-generation sequencing technology revealed novel frequent mutations in DNA methyltransferase 3A (DNMT3A) and IDH1 and IDH2 in adult normal karyotype AML. In adult disease, these mutations confer a poor outcome. By contrast, these mutations are relatively rare in pediatric AML but similarly occur more commonly in patients with a normal karyotype, highlighting the different molecular spectrum of disease in adult and childhood counterparts.

Morphology and Cytochemistry

The initial diagnosis of AML relies on accurate interpretation of the cellular morphology of the bone marrow smears. The diagnosis usually can be made based on the morphologic characteristics of the blasts in a Wright-Giemsa–stained bone marrow smear ( Fig. 51-5 ). In addition to general morphology, the diagnosis can be confirmed in cases exhibiting Auer rods (thin, needle-shaped cytoplasmic deposits that stain pink with Wright-Giemsa and are strongly myeloperoxidase [MPO]-positive). In addition to examination of the morphology of the blasts, the Wright-Giemsa stain is also used to assess for dysplastic features in erythroid, myeloid, and megakaryocytic cell lines.

Figure 51-5, Subtypes of acute myeloid leukemia.

Additional cytochemical stains are important for the accurate diagnosis of AML. MPO and Sudan black B (SBB) are cytochemical stains for granulocyte, eosinophil, and monocyte lineages. Peroxidase is present in the primary granules of myeloid cells, beginning at the promyelocyte stage and throughout subsequent maturation. Typically, leukemic myeloblasts are also strongly peroxidase-positive. MPO staining of Auer rods is particularly robust in leukemic blasts and can demonstrate Auer rods not recognized using the Wright-Giemsa stain. By contrast, the MPO staining of monoblasts shows a fine granular pattern in the monoblast cytoplasm. The staining pattern of SBB is similar to that of peroxidase, particularly in myeloblasts and in the detection of Auer rods. Monoblasts either do not stain or show a weakly positive, diffuse pattern with SBB.

The esterase stains are useful for distinguishing granulocytic cells from cells of the monocytic lineage. Chloroacetate esterase is a specific stain for granulocytes and mast cells. Results of staining of early myeloblasts are often negative, but promyeloblasts and later granulocytic lineage cells stain strongly, as do Auer rods. Monoblasts do not stain with chloroacetate esterase. In contrast, the nonspecific esterases (NSEs) such as α-naphthyl butyrate esterase and α-naphthyl acetate esterase stain monoblasts and monocytes strongly. Granulocytic and lymphoid cells do not stain with the nonspecific esterases. The blast cells of AML-M4 (i.e., myelomonocytic leukemia) should demonstrate positive chloroacetate esterase activity and α-naphthyl butyrate esterase or α-naphthyl acetate activity; this finding is important for making the diagnosis. The periodic acid–Schiff (PAS) reaction is not as informative as the previously described stains, but it can be useful, particularly in the diagnosis of AML-M6 (i.e., erythroleukemia). Leukemic erythroblasts can stain strongly with PAS, whereas normal erythroid precursors generally demonstrate no PAS activity.

Immunophenotype

Since the 1980s, multiparameter flow cytometric analysis has become an important element in the diagnosis and classification of leukemia. More recently, multiparameter flow cytometry has been used as a tool to detect MRD in acute myeloid leukemia and is being incorporated into clinical trials to clarify prognostic groups and help make treatment decisions. Immunophenotyping using a large panel of monoclonal antibodies to myeloid and lymphoid lineage and progenitor cell–associated antigens has been used to discriminate myeloid or lymphoid differentiation correctly in up to 98% of patients. Monoclonal antibodies have been classified based on their reactivity with the lineage or differentiation-associated antigens on the surfaces of normal and malignant cells. Because the monoclonal antibodies are not leukemia cell–specific and most of the hematopoietic antibodies are not strictly lineage specific, it is necessary to use panels of antibodies on blast cell populations and to incorporate the immunophenotypic information with the clinical findings and other morphologic, cytochemical, and cytogenetic results for accurate leukemia cell classification.

Blasts of myeloid origin generally express HLA-DR, CD33, and CD34. Monocytic cells usually express HLA-DR at all stages of maturation, whereas promyelocytes and more mature cells of the granulocyte lineage do not. CD33 and CD13 are expressed on neutrophil and monocyte precursors, but CD33 is absent on mature neutrophils. CD14 is relatively specific for cells of the monocytic lineage. As myeloid differentiation continues, CD15 expression increases, whereas CD34 expression decreases. Megakaryocytic cells and platelets express CD41, CD42, and CD61.

Immunophenotyping has been particularly valuable for lineage assignment in undifferentiated leukemias (i.e., AML-M0), identification of acute megakaryocytic leukemia, and analysis of the lineage of leukemias with MLL gene translocations.

Cytogenetics

Cytogenetic analysis is an essential component in the diagnosis and treatment of AML. The chromosomal aberrations detected by cytogenetic testing also have prognostic value and may be used as tumor markers. In the WHO classification of hematologic malignancies, specific cytogenetic abnormalities have been included in the diagnostic and prognostic criteria for differentiating subclasses of AML.

Cytogenetic analyses of bone marrow samples have revealed chromosomal abnormalities in most patients with AML. In the pediatric population, the incidence of cytogenetic abnormalities is even higher than that in adults. However, the distribution of the specific cytogenetic abnormalities is different in the various age groups. In infants with AML, the most common chromosomal aberrations are translocations involving the MLL gene (band 11q23). The incidence of 11q23 translocations is as high as 40% in AML blasts of children younger than 2 years. After age 2 years, the frequency of 11q23 translocations decreases with age, and the translocation is detectable in AML blasts from less than 10% of older children and adults. One rare chromosomal abnormality, the t(1;22)(p13;q13), is found almost exclusively in non–Down syndrome infants with acute megakaryocytic leukemia. In a published review of 39 patients with this translocation, 95% were younger than 2 years, and it was not identified in adults with AML. This translocation is now known to result in aberrant expression of the OTT-MAL fusion gene.

There are two classification systems for AML (see next section). The FAB classification is based exclusively on morphology. However, there are some morphologic subtypes such as M3 and M4eo that are always associated with particular cytogenetic changes, that is, RARA rearrangements for the former and inv(16) or t(16;16) in the latter. In addition, there are certain cytogenetic changes that are often (if not exclusively) associated with a particular subtype and that carry prognostic significance, such as the good prognosis t(8;21) often associated with the M2 subtype or MLL rearrangements often associated with the M5 subtype. The WHO classification system takes this into account and has a category of “acute myeloid leukemia with recurrent genetic abnormalities,” which includes t(8;21), inv(16) or t(16;16), t(15;17), and 11q23. The fusion proteins thus generated are discussed in more detail earlier (see “Acute Myelogenous Leukemia: Biology”). Some of the more common chromosomal abnormalities and their associated FAB and WHO subtypes are shown in Table 51-1 and Box 51-2 . The prognostic significance of certain cytogenetic abnormalities is discussed later (see “ Prognosis ”).

TABLE 51-1
French-American-British (FAB) Classification of Acute Myelogenous Leukemia with Corresponding Cytogenetic and Immunophenotypic Characteristics
FAB Subtype MPO SBB NSE PAS MYELOID ERYTHROID MEGAKARYOCYTIC T-CELL LINEAGE OTHER Translocations and Rearrangements Genes Involved
CD 11b CD 13 CD 14 CD 15 CD 33 CD 34 CD 65 Glycophorin A CD 41/61 CD 42 CD 36 CD 2 CD 3 CD 4 CD 7 CD 56 HLA-DR CD 117
M0 ++ + + ++ ++ + + + + + + − + − + ++ ++ 11q23
Trisomy 4, 8,13, −5, −7
MLL rearrangements
M1 + + + + + + + + + + ++ ++ t(8;21)(q22;q22)
t(9;22)(q34;q11)
Trisomy 8, −5, −7
AMLI-ETO
BCR-ABL
M2 + + + + + ++ ++ + + + + ++ + t(8;21)(q22;q22)
t(6;9)
inv3(q21;q26), t(3:3) (q21;q26)
Trisomy 8, −5, −7
AMLI-ETO
DEK-CAN
EVI1
M3 + + + + + + + + + + + + t(15;17)(q22;q12)
t(11;17)(q23;q12),
t(5;17) (q32;q12)
PML-RARA
PLZF-RARA
NPM-RARA
M4 + + + ++ ++ ++ + + + ++ + ++ + + +++ + t(6;9)
inv3(q21;q26), t(3:3) (q21;q26)
t(9;11)(p22;q23)
11q23
DEK-CAN
EVII
AF9-MLL
MLL rearrangements
(M4eo) + + + + ++ + + + + + ++ +++ + inv16(p13;q22), t(16:16) MYHII-CBFβ
M5 + ++ ++ ++ ++ + ++ ++ + ++ + + +++ + t(11;17)(q23;q21)
11q23
11q23,t(9;11)(p22;q23) [M5a]
t(8;16), t(10;11)(p12;q23) [M5b]
MLL-AF17
MLL rearrangements
MLL, AF9-MLL
MOZ-CBP, AF10-MLL
M6 + + + + + + ++ + + + Trisomy 8, −5, −7
M7 + + + + + ++ ++ +
+
++ + + + + + t(1;22)(p13;q13) OTT-MAL
M7 (DS) + + + + + ++ ++ ++ ++ + + + + + Trisomy 21 GATA-1
AML, Acute myelogenous leukemia; DS, Down syndrome; MLL, mixed-lineage leukemia; MPO, myeloperoxidase; NSE, nonspecific esterase; PAS, periodic acid–Schiff; SBB, Sudan black B.

Box 51-2
Modified from Vardiman JW, Harris NL, Brunning RD: The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 100:2292–2302, 2002; and Vardiman JW, Thiele J, Arber DA, et al: The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 114:937–951, 2009.
World Health Organization (WHO) Classification of Acute Myeloid Leukemia and Corresponding French-American-British (FAB) Subtype *

* Shown in boldface.

  • Acute Myeloid Leukemia with Recurrent Genetic Abnormalities (FAB)

    • t(8;21)(q22;q22); (AML1/ETO; RUNX1-RUNX1T1) (M2)

    • inv(16)(p13;q22) or t(16;16)(p13;q22); (CBFβ/MYH11) (M4E0)

    • t(15;17)(q22;q12) (PML/RARA) (M3)

    • AML with t(9;11)(p22;q23); MLLT3-MLL

    • AML with t(6;9)(p23;q34); DEK-NUP214

    • AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2); RPN1-EVI1

    • AML (megakaryoblastic) with t(1;22)(p13;q13); RBM15-MKL1

  • Acute Myeloid Leukemia with Myelodysplasia-Related Changes

    • Following myelodysplastic syndrome (MDS) or MDS/myeloproliferative disease (MPD)

  • Therapy-Related Myeloid Neoplasms

  • Acute myeloid leukemia (FAB)

    • Acute myeloid leukemia, minimally differentiated (M0)

    • Acute myeloid leukemia without maturation (M1)

    • Acute myeloid leukemia with maturation (M2)

    • Acute myelomonocytic leukemia (M4 )

    • Acute monoblastic and monocytic leukemia (M5)

    • Acute erythroid leukemia (M6)

    • Acute megakaryoblastic leukemia (M7)

    • Acute basophilic leukemia

    • Acute panmyelosis with myelofibrosis

  • Myeloid Sarcoma

  • Myeloid Proliferations Related To Down Syndrome

  • Blastic Plasmacytoid Dendritic Cell Neoplasm

French-American-British Classification

In 1976, the FAB group proposed a system of classification of AML based on morphologic and cytochemical features (see Table 51-1 ). The system divides AML into seven subtypes, M1 through M7. An M0 subtype was later added to describe undifferentiated leukemia and, since 1976, immunophenotypic data have also been included. In general, AML can be differentiated from ALL based on morphologic features and cytochemical stains. Cells of myeloid origin should stain with myeloperoxidase and SBB. Cells of monocytic derivation usually stain with NSE. AML blasts usually do not have PAS activity, except for erythroblasts in AML-M6 and eosinophils of the M4Eo subtype.

M0: Acute Myeloblastic Leukemia with Minimal Differentiation.

In patients with AML-M0, the bone marrow is usually hypercellular and more than 90% of the cells are blasts. Most blast cells lack cytoplasmic granules, nucleoli, or Auer rods, and results with MPO and SBB stains are negative. Immunophenotypic analysis shows the presence of myeloid or monocytic cell antigens (i.e., CD13, CD14, CD33, or CD34) that are detectable on the cell surface of most AML-M0 blasts, but some AML-M0 blasts express terminal deoxynucleotidyl transferase, an enzyme usually associated with ALL. AML-M0 is associated with a high incidence of cytogenetic abnormalities, most of which are complex and often involve chromosomes 5 and 7, trisomy 8, or MLL rearrangements.

M1: Acute Myeloblastic Leukemia without Maturation.

The bone marrow of patients with AML-M1 is hypercellular and filled with myeloblasts (more than 90% blasts). Most blasts stain with MPO and SBB. The blast cells in this subtype display minimal myeloid differentiation. Morphologically, the blast cells may contain scant gray-blue cytoplasm and few or no azurophilic granules or Auer rods. Prominent nucleoli are usually detectable. Flow cytometric analysis usually shows that the blast cells of AML-M1 express HLA-DR, CD13, CD33, and CD34. Cytogenetic abnormalities for this subclass often include monosomy 5 or 7 or trisomy 8.

M2: Acute Myeloblastic Leukemia with Maturation.

The bone marrow of patients with AML-M2 usually shows evidence of some maturation beyond the myeloblast, with evidence of maturation beyond the promyelocyte stage in more than 10% of nonerythroid cells. Myeloblasts must represent more than 30% of the bone marrow cells but less than 90% of nonerythroid cells. The blasts generally have a few clusters of primary granules and stain with MPO and SBB. Auer rods and prominent nucleoli are common. Immunophenotypic expression of HLA-DR, CD13, CD33, CD11, and CD15 is typical. This subtype may exhibit monosomy 5 or 7, trisomy 8, t(8;21), t(6;9), or abnormalities of chromosome 3.

M3: Acute Promyelocytic Leukemia.

There are two types of AML-M3. The more common type is the hypergranular variant, in which more than 30% of the blasts are promyelocytes and myeloblasts. Most promyelocytes have heavy granulation. Auer rods and Auer rod bundles are common. In the rare microgranular variant, the cells exhibit fine cytoplasmic granules, which are not distinguishable by light microscopy and nuclear morphologic irregularities (i.e., microgranular M3 or M3v). The cells of both subtypes stain strongly with MPO and SBB and also with chloroacetate esterase. The promyelocytes usually do not show PAS and NSE activity. The blasts express CD13, CD33, CD11, and CD15, but they are HLA-DR− and CD14−. This subtype exclusively exhibits RARA rearrangements, the vast majority of which are t(15;17) leading to PML-RARA .

M4: Acute Myelomonocytic Leukemia.

The M4 subtype is defined by the presence of blast cells with both granulocytic and monocytic features. The bone marrow in these patients has more than 30% infiltration by immature myeloid precursors, and there is often extramedullary involvement and a peripheral blood monocytosis. The blasts are usually pleomorphic with regard to size, amount of cytoplasm, granularity, and nuclear morphologic features. Some Auer rods can be seen, and prominent nucleoli are usually present. Staining is variable, with some of the blast cells showing positivity for MPO, SBB, and NSE. M4 AML may be associated with t(6;9), abnormalities of chromosome 3, and MLL gene rearrangements, in particular t(9;11). A small proportion of patients with AML-M4 have a moderate eosinophilia in their bone marrow (M4Eo). The eosinophils are notable for the presence of basophilic and eosinophilic granules and stain with chloroacetate esterase and PAS. Cell surface antigen expression includes CD13, CD15, CD33, CD4, CD11c, CD14, CD64, and HLA-DR. M4eo AML exhibits an inv(16) or t(16;16).

M5: Acute Monocytic Leukemia.

More than 80% of the nonerythroid bone marrow cells in patients with AML-M5 are monocytic. There are two subtypes: M5a (undifferentiated) and M5b (differentiated). In AML-M5a, more than 80% of the cells are monoblasts. Patients with AML-M5a tend to be younger, have higher presenting white blood cell (WBC) counts, and have a poorer prognosis. In AML-M5b, fewer than 80% of the monocytic cells are monoblasts, and most of the cells are recognizable as monocytes or promonocytes. The peripheral blood in patients with AML-M5b exhibits a profound monocytosis. In both subtypes, the blasts demonstrate strong NSE activity. MPO and SBB staining results are usually negative. Cell surface antigen expression includes CD13, CD15, CD33, CD4, CD11c, CD14, CD64, and HLA-DR. M5 AML often exhibits MLL gene rearrangements, t(9;11) or t(11;17) in M5a and t(10;11) in M5b.

M6: Acute Erythrocytic Leukemia.

AML-M6 is an uncommon form of AML overall and is rare in children. This form of leukemia is defined by a more than 50% erythroblast infiltration of the bone marrow. M6 AML may exhibit monosomy 5 or 7 or trisomy 8.

M7: Acute Megakaryoblastic Leukemia.

The bone marrow exhibits an infiltration with pleomorphic megakaryoblasts that often display cytoplasmic budding and may appear in clusters. Aspiration of the bone marrow may be challenging due to associated fibrosis. M7 blasts generally do not stain with MPO and SBB but show activity with PAS. Immunophenotyping is usually required to distinguish AML-M7 from ALL-L2, documenting the presence of the megakaryocytic antigens, glycoprotein Ib, glycoprotein IIb/IIIa, or factor VIII. Non–Down syndrome infants with M7 AML often exhibit a t(1;22). Down syndrome patients have a very high incidence of M7 AML with a particularly good prognosis, which is in contrast to the poor prognosis of M7 AML in non–Down syndrome children and adults.

World Health Organization Classification

The discovery of cytogenetic and molecular genetic abnormalities in malignant disease has had a significant impact on our understanding of malignant transformation. In myeloid malignancies, some genetic abnormalities have prognostic significance whereas others appear to define specific disease subtypes. The WHO has proposed a classification system for neoplastic diseases of the hematopoietic and lymphoid tissues that includes a classification for AML (see Box 51-2 and Fig. 51-5 ). This classification uses the traditional FAB-type morphologic categories of disease and includes additional entities such as immunophenotype, molecular genetic, and clinical characteristics that make the system more clinically relevant for diagnosis, prognosis, and treatment. The WHO classification system divides myeloid diseases into several major subtypes—myeloproliferative neoplasms; myeloid neoplasm associated with eosinophilia and abnormalities of PDGFRA , PDGFRB , or FGFR1 ; myelodysplastic-myeloproliferative neoplasms; MDS, AML and related neoplasms; and acute leukemias of ambiguous lineage. The 2008 revision of the WHO classification has expanded the category of AML to include AML with recurrent cytogenetic translocations, AML with myelodysplasia-related changes, therapy-related myeloid neoplasms, AML not otherwise specified, myeloid sarcoma, myeloid proliferations related to Down syndrome, and blastic plasmacytoid dendritic cell neoplasms.

Within the subtype containing recurrent cytogenetic translocations, seven additional categories have been described:

  • AML with t(8;21)(q22;q22); AML1-ETO ; RUNX1-RUNX1T1

  • AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11

  • APL with t(15;17)(q22;q12); PML-RARA

  • AML with t(9;11)(p22;q23); MLLT3-MLL

  • AML with t(6;9)(p23;q34); DEK-NUP214

  • AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2); RPN1-EVI1

  • AML (megakaryoblastic) with t(1;22)(p13;q13); RBM15-MKL1

AML with t(6;9)(p23;q34) and AML with inv(3)(q21q26.2) is relatively uncommon in children. Myeloid sarcomas, formerly called chloromas, may precede hematopoietic evidence of disease and so are now classified as a separate entity. The myeloid diseases of Down syndrome were incorporated into the revised WHO classification in 2008 and are described later in this chapter. Plasmacytoid dendritic cell neoplasms were formerly referred to as blastic NK-cell lymphomas, but the nomenclature has changed to reflect the origin of this myeloid malignancy in this subset of dendritic cells. This rare disease can manifest in children with skin lesions, adenopathy, and ultimately bone marrow involvement. Blast cells characteristically express CD4, CD43, CD56, and CD123 without expression of CD34 or CD117.

The FAB standard used to define AML had been 30% replacement of the bone marrow by the blast cells, but later studies have shown that patients with 20% to 30% blasts (previously classified as refractory anemia with excess blasts in transformation [RAEB-T]) have a prognosis similar to that of patients with more than 30% blasts. The blast count for the diagnosis of AML in the WHO classification was therefore changed to 20%, and the category of RAEB-T was eliminated.

Certain specific and recurrent cytogenetic abnormalities are common in MDS, in alkylating agent–related AML, and in de novo AML with a poor prognosis. These abnormalities include 3q−, −5, 5q−, −7, 7q−, +8, +9, 11q−, 12p−, −8, −19, 20q−, +21, t(1;7), t(2;11), and complex karyotypes. In the classification scheme, these cytogenetic abnormalities were considered to indicate a poor prognosis. Prior therapy with topoisomerase II inhibitors (i.e., epipodophyllotoxins and doxorubicin) was also associated with a poor prognosis. Typically, patients in whom AML develops after exposure to these agents have translocations involving 11q23 ( MLL ). The WHO classification includes these patients in the poor-prognosis category but distinguishes them from having alkylating agent–related secondary leukemia.

Patients may have MDS with unilineage or multilineage dysplasia, and these are reflected as distinct diseases in the most recent iteration of the WHO classification. Dysplasia is defined as being observed in greater than or equal to 10% of the cells of a given myeloid lineage. Children with 2% to 19% peripheral blasts or 5% to 19% bone marrow blasts can be classified according to the same criteria as adult MDS, but a new category of refractory cytopenia of childhood has been introduced to define children with persistent cytopenias and dysplasia of two or more lineages but less than 2% peripheral blasts or less than 5% bone marrow blasts.

Clinical Presentation

The presenting signs and symptoms of AML result from leukemic blast cell infiltration of the bone marrow. The leukemia cells overwhelm the processes of normal hematopoiesis, resulting in anemia, thrombocytopenia, and neutropenia. It is rare for AML to be diagnosed incidentally on a routine medical evaluation, but there is considerable variability in the range of presenting signs and symptoms in patients with de novo AML. Usually, patients seek medical attention for fever, fatigue, pallor, skin or mucosal bleeding, bone pain, or infections not responding to appropriate antibiotic therapy. Bone pain is a common symptom, and patients may present with a limp, rib pain, or back pain.

The WBC count of patients with newly diagnosed AML can range from less than 1,000/µL to more than 500,000/µL. The leukocyte count is more than 100,000/µL in approximately 25% of pediatric patients with AML, and an elevated count is more common in those with the M4 and M5 subtypes and in those with FLT3 -ITD mutations. The number of circulating granulocytes is often critically decreased, regardless of the total leukocyte count, and, because their function is usually impaired, the risk for overwhelming bacterial infection in patients with newly diagnosed AML is markedly increased. Patients with AML who have a fever require immediate treatment with broad-spectrum antibiotics after appropriate culture samples have been obtained. The hemoglobin level is occasionally normal but is usually less than 9 g/dL, and levels as low as 3 g/dL at diagnosis are not uncommon. About half of children with new-onset AML have platelet counts of 50,000/µL or less, which increases the risk for life-threatening bleeding.

Disseminated intravascular coagulation (DIC) has been observed in patients with all FAB subtypes of AML but is most common in acute promyelocytic leukemia (APL; see later). Before the development of ATRA, treatment with low-dose heparin during induction was initiated for patients with AML-M3 to prevent DIC. The later addition of ATRA to induction therapy rapidly resolved DIC in patients with AML-M3 and markedly reduced early mortality.

Patients with hyperleukocytosis have an increased risk for mortality from leukostasis, particularly in the brain or lung, and urgent leukapheresis or treatment with leukocyte-reducing agents such as hydroxyurea may be necessary (see “Complications”). Extramedullary leukemia occurs in 20% to 25% of children with AML and may include chloromas (tumor nodules), skin infiltration, CNS disease, gingival infiltration, hepatosplenomegaly, or testicular involvement. Chloromas (e.g., myeloblastomas and granulocytic sarcomas) are solid tumors of myeloblasts that may occur in any area of the body but are most common in the orbit and epidural area. Skin infiltration, or leukemia cutis, usually presents as slightly purple lesions (i.e., “blueberry muffin” spots), is more common in infants with monocytic leukemia, and may be the initial sign of disease. CNS involvement at diagnosis is more common in AML than in ALL. Up to 15% of patients with AML have myeloblasts in the cerebrospinal fluid at diagnosis. Gingival infiltration may present as hyperplasia, often accompanied by bleeding. Severe extramedullary disease with massive hepatosplenomegaly is also a more common finding in infants. Extramedullary involvement of the testes can also be documented but is less common than in ALL. Extramedullary infiltration, particularly gingival involvement, is more common with the M4 and M5 (acute myelomonocytic and acute monoblastic and monocytic) leukemias. Chloromas and CNS disease are more common in AMLs with t(8;21). Extramedullary infiltration in pediatric patients is not clearly associated with worse prognosis unless accompanied by a high presenting WBC count.

Treatment

As is true for any pediatric cancer, the goal of therapy for children with AML is to completely eradicate the disease while limiting treatment-induced toxicity. In general, therapy for AML includes immediate supportive care measures followed by intensive remission induction and consolidation chemotherapy. At any given time, several large, multicenter phase III clinical trials are ongoing, each aimed to improve overall survival (OS) through randomized comparisons of treatment options while minimizing short- and long-term toxicities.

Remission Induction

During the past 40 years, survival rates for patients with AML have risen from less than 10% to more than 50% of patients because of intensification of chemotherapy, use of hematopoietic stem cell transplantation (HSCT), and improved supportive care. The first phase of chemotherapy is termed induction and consists of two to four cycles of intensive chemotherapy with the goal of inducing remission. Morphologic remission is defined as the presence of fewer than 5% blasts visible on bone marrow aspirate obtained after recovery of counts. Patients may also achieve cytogenetic and FISH-negative remission, as defined by the absence of a previously detected leukemic clone–associated cytogenetic abnormality in the bone marrow cells. Remission is also now being defined by the absence of MRD, assessed by multiparameter flow cytometry that is designed to detect as few as 0.1% leukemic blasts in the marrow. The inclusion of MRD evaluation has not only identified children at risk for relapse who appeared to be in morphologic remission but has also revealed false-positive recovering marrows that had been deemed refractory disease.

Induction chemotherapy should be initiated as soon as the diagnosis of AML is confirmed, preferably by morphologic, immunophenotypic, and cytogenetic studies of the bone marrow. In some cases, particularly in patients presenting with hyperleukocytosis, the patient's condition may be too unstable for bone marrow evaluation, in which case the diagnosis can often be confirmed on studies of the peripheral blood. Induction chemotherapy regimens consist of high-dose myelosuppressive cytotoxic agents that result in prolonged pancytopenia. The period of bone marrow hypoplasia generally lasts from 21 to 30 days from the initiation of therapy but may last considerably longer. During this period, patients have a high risk for life-threatening infection and bleeding. Aggressive supportive care is necessary (see later).

Historical North American Approaches to Treating AML.

The basis of modern induction therapy for AML is a combination of cytarabine and an anthracycline. The classic combination therapy was the so-called 7 + 3 regimen, which consists of a continuous 7-day intravenous infusion of cytarabine at 100 to 200 mg/m 2 /day and 3 days of bolus infusions of daunorubicin at 45 to 60 mg/m 2 /day. Remission is achieved in 60% to 70% of patients with newly diagnosed AML using this regimen. In children, several strategies have been used to intensify this regimen to achieve higher rates of CR. CCG trial 213 added etoposide, thioguanine, and dexamethasone to the traditional 7 + 3 regimen to create the five-drug Denver regimen. The Denver and 7 + 3 regimens were compared in a randomized fashion, and there was no significant difference in CR rate, with 79% of children achieving CR with 7 + 3 and 76% achieving CR with Denver regimen.

In CCG trial 2891, the same drugs were used but the timing was intensified in an attempt to improve CR. The hypothesis was that leukemia cells may be recruited synchronously into the cell cycle after chemotherapy, and therefore reexposure at an earlier time point may affect cells while more of them are at a sensitive phase in the cell cycle. Thus the Denver regimen was modified into the five-drug DCTER regimen, again consisting of d examethasone, c ytarabine, t hioguanine, e toposide, and R ubidomycin (daunorubicin). Induction consisted of a total of four cycles of chemotherapy, and patients were randomized to standard or intensive timing. For standard timing, each cycle of induction began with count recovery from the previous cycle. With intensive timing, cycles 2 and 4 began at day 10 after the start of cycles 1 or 3, respectively, regardless of hematologic status. Not surprisingly, the intensive timing resulted in far greater toxicity, with 11% of patients in this arm of the trial dying of toxicity compared with only 4% of patients dying of toxicity in the standard timing trial arm. Because of this, better supportive care measures were instituted for the intensive timing trial arm, including more aggressive empiric antibiotic and antifungal coverage and the use of granulocyte colony-stimulating factor (G-CSF). Interestingly, the CR rates were similar for the two arms in the trial—75% for intensive timing and 70% for standard timing—but the reason for failure to achieve remission was different, with 11% toxic death and only 14% resistant disease in the intensive timing arm versus only 4% toxic death but 26% resistant disease in the standard timing arm. The most dramatic results, however, came later, when it was shown that for patients who achieved a CR there was a 3-year disease-free survival (DFS) of 55% for those who had been treated in the intensive timing arm, versus a DFS of only 37% for patients who had received standard timing. For this reason, the standard timing arm of the trial was closed early and an intensive timing DCTER/DCTER schedule became the new standard combination therapy for newly diagnosed AML patients.

In the meantime, a comprehensive review of five randomized trials comparing idarubicin with daunorubicin as induction therapy for AML had shown improved remission induction rates and OS for patients receiving idarubicin. For this reason, the CCG pilot trial 2941 tried to intensify therapy further by using idarubicin in place of daunorubicin in each cycle of the intensively timed regimen. Because of unacceptably high toxicity, with a toxic death rate of 14%, this was modified to idarubicin only in cycles 1 and 3, and more strict supportive care guidelines were put into place, including mandatory hospitalization during neutropenia, and stricter infection prophylaxis (see later, “ Supportive Therapy at Diagnosis and during Therapy ”). The addition of idarubicin showed no advantage in remission induction or event-free survival (EFS) but did show a decrease in marrow blasts at day 14 of induction. CCG trial 2961 continued to use the hybrid idaDCTER/DCTER intensive timing induction regimen resulting in induction remission rates of 89%, which were similar to historic controls. Most of the improvement was believed to be related to better supportive care rather than to the change in chemotherapy.

Medical Research Council Trials (MRC10, MRC12, and MRC15).

During this period, data emerged from the British MRC trials that seemed to show better survival and less toxicity. MRC10 induction chemotherapy randomized DAT (daunorubicin, cytarabine, thioguanine) to ADE (cytarabine, daunorubicin, etoposide) in a 10-day cycle of chemotherapy followed by an 8-day cycle of the same drugs on WBC count recovery. The CR rate for children in this study was 93%, with a 4% rate of toxic death and a 3% rate of resistant disease. Early data suggested a benefit for pediatric patients using ADE versus DAT, but the later data showed no difference in 10-year OS (DAT, 57%; ADE, 51%; P = .3), DFS (DAT, 53%; ADE, 48%; P = .3) or EFS (DAT, 48%; ADE, 45%; P = .5). Because of the early results possibly favoring ADE, MRC12 compared ADE with MAE (mitoxantrone, cytarabine, etoposide) in a 10-day cycle followed by an 8-day cycle on count recovery. The CR rates were similar for the two groups (ADE, 92%; MAE, 90%; P = .3) with a slight increase in toxic deaths for the MAE group (ADE, 3%; MAE, 6%) but a lower relapse rate (ADE, 39%; MAE, 32%) and better DFS rate for the MAE group (ADE, 55%; MAE, 63%), although the 10-year EFS and OS rates were not significantly different (54% and 63%, respectively). In MRC15 there were three randomized induction backbones—ADE; cytarabine and daunorubicin without etoposide, or idarubicin, fludarabine, cytarabine, and G-CSF (Ida-FLAG)—and three randomized consolidation backbones—amsacrine, cytarabine, and etoposide; cytarabine 1.5 g/m 2 ; or cytarabine 3 g/m 2 with and without the addition of gemtuzumab ozogamicin (GMTZ, GO, Mylotarg). GMTZ, a recombinant humanized anti-CD33 monoclonal agent linked to the tumor antibiotic calicheamicin, was incorporated as a targeted therapy in these studies. Because CD33 is expressed on most AML cells but not on pluripotent HSCs or other tissues, this drug can target leukemia cells more specifically. GMTZ was well tolerated overall, but no overall improvements in survival or relapse rate were observed regardless of the chemotherapy backbone in either induction or consolidation.

Recent U.S. Trials.

The next series of U.S. trials built on the data from these recent MRC studies. The COG has undertaken three therapeutic trials to date based on an MRC backbone. The COG AAML03P1 pilot and AAML0531 phase III groupwide study backbone included cytarabine/daunorubicin/etoposide (ADE 10+3+5) followed by ADE 8+3+5, cytarabine/etoposide (AE), mitoxantrone/cytarabine (MA), and, finally, high-dose cytarabine (Capizzi II) for children who did not go on to receive HSCT. The COG pilot trial AAML03P1 assessed the safety of adding a single dose of GMTZ to ADE 10+3+5 and MA cycles. Toxicity on this pilot study was acceptable, so GMTZ was added to these same two cycles on AAML0531 in a 1 : 1 randomization. Subsequent outcome data from the AAML03P1 trial demonstrated a 3-year EFS of 53% and OS of 66% with no increased treatment-related mortality over backbone chemotherapy. Outcome data have not yet been released for AAML0531. However, after data were released from the Southwest Oncology Group (SWOG) Study 106, the U.S. Food and Drug Administration (FDA) removed GMTZ from the U.S. market because this adult-based study showed no benefit in outcome to the addition of GMTZ but an increase in induction deaths. By contrast, subgroup analysis on the MRC 15 trial found that the addition of GMTZ to induction chemotherapy was of benefit to CBF AML and to intermediate-risk patients and did not contribute to increased toxicity. Data from the COG AAML0531 trial demonstrated mixed results. GMTZ improved 3 year EFS and relapse risk for all patients but not OS. Interestingly, positive trends were seen for GMTZ in subgroup analysis, including OS in high risk patients and relapse risk in low risk patients, although this was also associated with increased TRM in the low risk subgroup. Given this controversy regarding both toxicity and efficacy and the absence of data from the AAML0531 at the time of study initiation, GMTZ was not included on the current COG AAML1031 trial. However, other co-operative groups are planning future trials to examine the optimal dosing of GMTZ in pediatric AML (A. Gamis, personal communication, July 2014). The current COG trial risk stratifies patients into high- and low-risk AML based on cytogenetics and MRD and randomizes the addition of the proteosome inhibitor bortezomib as a potential AML stem cell targeted therapy. In addition, patients harboring the FLT3 -ITD with sufficient allelic ratio have the option of being nonrandomly assigned to an MRC-based backbone arm with the addition of the FLT3 inhibitor sorafenib. Given recent MRC data that four cycles appear as effective as five cycles of therapy, only four cycles are included on this protocol (ADE 10+3+5; ADE 8+3+5; cytarabine and etoposide; and mitoxantrone and cytarabine) for good responders. Patients with positive MRD after cycle 1, defined as more than 0.1% by multiparameter flow cytometry (MPFC) receive mitoxantrone and cytarabine instead of an additional ADE cycle, and high-risk patients receive high-dose cytarabine for cycle 4.

The St. Jude Children's Research Hospital (SJCRH) trial AML02 randomized patients to standard low- or high-dose cytarabine during the first cycle of induction together with etoposide and daunorubicin (ADE), although this was not found to impact remission rate, EFS, or OS. Toxicity was generally similar in both arms of the trial, although patients treated on the high-dose arm had a greater incidence of serious fungal infections. The SJCRH trial incorporated MRD measurements during induction to guide therapy. MRD by MPFC was determined on a bone marrow sample obtained on day 22 of induction. Patients with MRD greater than 1% on day 22 received induction II before WBC count recovery. Patients with MRD greater than 1% at the end of the first month of induction I received GMTZ in addition to the second cycle of ADE. Patients with CNS-positive disease received triple intrathecal therapy. Patients who were MRD positive after induction I had a significantly worse outcome than patients who were MRD negative (3-year EFS, 43% vs. 74%). However, subgroup analysis revealed that this difference was only found in patients with high-risk AML at diagnosis ( FLT3 -ITD positive, monosomy 7, t(6;9), megakaryoblastic leukemia, therapy-related AML, or AML after MDS) or those with MRD levels greater than 1%. Low- and standard-risk AML patients or those with what was referred to in this study as low-level MRD (0.1% to 1%) did not demonstrate any outcome differences versus those with negative MRD. However, 20 patients who were MRD negative ultimately experienced relapse. In the current SJCRH study, AML08, patients are randomly assigned to receive high-dose cytarabine, etoposide, and daunorubicin versus clofarabine and cytarabine in induction I. Low-risk patients (those with CBF leukemia with negative MRD after induction I) receive 4 courses of chemotherapy, whereas those at high risk for relapse— FLT3 -ITD positive, monosomy 7, t(6;9), t(8;16), t(16;21), megakaryoblastic leukemia without t(1;22), treatment-related AML, AML after MDS, or MRD greater than 0.1% after induction II—receive two to three courses of chemotherapy followed by allogeneic HSCT. Standard-risk patients receive four courses of chemotherapy and are then eligible to receive one course of natural killer cell therapy if they have a killer cell immunoglobulin-like receptor (KIR)-mismatched haploidentical family member. Patients with FLT3 -ITD receive sorafenib after each course of chemotherapy (J. Rubnitz, personal communication).

The BFM (Berlin-Frankfurt-Münster) trial for de novo AML, BFM-2010, is for all comers, including patients with APL. Patients are being randomized to ADE ± GMTZ in induction I with MRD measurements on day 8, 15 and 28. Induction II includes cytarabine and idarubicin (AI) for patients with inv(16) and APL (who also receive ATRA). Other favorable-risk patients, including those with t(8;21), NPM1 mutations, and CEBPA , receive HAM (high-dose cytarabine 3 g/m 2 and mitoxantrone), and high-risk patients receive HAM plus targeted therapy, such as sorafenib if FLT3 -ITD positive, or dasatinib if KIT mutant positive. The third course is hAM (high-dose cytarabine at 1 g/m 2 ) for those who received AI and AI for low-risk patients who received HAM, with AI plus cladribine for the high-risk patients. Course 4 is hAM, except for the inv(16) and APL patients who receive cytarabine or cytarabine and etoposide as a final cycle. For all other patients, course 5 is high-dose cytarabine and etoposide. Liposomal daunorubicin, available in Europe, will be used in this trial. Prior studies have shown it to have equal efficacy in induction to idarubicin with less toxicity (G. Kaspars, D. Reinhardt, and D. Johnston, personal communication, January 2013).

Postremission Therapy

Several randomized studies have demonstrated that without postremission therapy almost all patients with newly diagnosed AML suffer a relapse within 2 years. The strategies for postremission therapy include autologous HSCT, allogeneic HSCT, or continued courses of chemotherapy. The CCG trials have shown no benefit to autologous HSCT over continued cycles of chemotherapy so, although that strategy continues to be used in some European trials, autologous HSCT is not included in current U.S. trials. Most studies have shown a lower relapse rate for AML patients treated with matched related donor HSCT versus chemotherapy, but the OS is often no better because of the significant morbidity and mortality of HSCT. For this reason, until the most recent trials, unrelated donor HSCT was believed to be too morbid for patients in first remission and thus trials used a genetic randomization, wherein all patients with a matched related donor proceeded to HSCT and all others proceeded with further cycles of chemotherapy.

Based on the success of the MRC trials with chemotherapy alone using a combination of cytogenetic characteristics and response to therapy, the COG and SJCRH adopted risk-based criteria for HSCT. HSCT, even from a matched related donor, is not recommended for patients with low-risk disease. This was previously defined by COG as the presence of t(8;21) or inv(16) in the leukemic clone at diagnosis, but more recently the presence of a CEBPA or NPM1 mutation has been added to this definition, based on the data revealing the better outcome for these patients. The COG has classified high-risk disease as monosomy 7, −5/5q−, FLT3 -ITD with high allelic ratio, MRD positive after induction chemotherapy, or more than 15% blasts in the marrow after one course of induction chemotherapy. * The incorporation of MRD after induction has enabled the COG to relegate normal karyotype patients to high- and low-risk groups, thereby dichotomizing patients and eliminating the intermediate-risk category. Therefore on the current AAML1031 protocol, high-risk patients will receive best available donor HSCT for consolidation, whereas good-risk patients will not go to transplant even if a sibling donor match is available. With recent advances in supportive care and improved therapy to prevent and treat graft-versus-host disease (GVHD), outcomes have improved for matched related and matched unrelated donor HSCT. Therefore for patients with high-risk disease, both trials recommend HSCT in first CR with an unrelated donor if no matched family donor is identified. The optimal number of cycles of chemotherapy before HSCT to balance minimizing toxicity while minimizing MRD is variable, with the MRC10 trial using four cycles, the current SJCRH trial including two to three cycles, and the COG trial using three cycles before HSCT.

* References .

For patients who do not proceed to HSCT there is controversy as to how many courses of chemotherapy are needed and what chemotherapy should be included. Several trials have shown that cycles of high-dose cytarabine after remission are associated with improved long-term survival. MRC12 randomized patients to receive four versus five total cycles of chemotherapy, with early results suggesting that five cycles are superior; thus that strategy was subsequently used in the current U.S. trials. However, more recent data from the MRC have shown that the fifth cycle added toxicity with no additional therapeutic benefit. These findings are reflected in the current COG trial in which only four cycles of chemotherapy are used for good-risk patients with elimination of the high-dose cytarabine and asparaginase cycle (Capizzi II) that previously comprised the fifth cycle on AAML0531 and AAML03P1. High-risk patients for whom a donor is not identified also only receive four cycles of chemotherapy, but they do receive Capizzi II as their last consolidation cycle. The SJCRH trial has tried to make up for the lack of amsacrine by tailoring the first postinduction cycle based on the patient's AML subtype and cytogenetics. On AML02, patients with t(9;11) or inv(16) AML receive cytarabine and cladribine, patients with M4/M5 AML receive cytarabine and etoposide, and patients with t(8;21) and all others receive high-dose cytarabine and mitoxantrone followed by two courses consisting of cytarabine and asparaginase followed by cytarabine and mitoxantrone. However, based on small subgroup numbers, AML08 uses a single backbone for all patients with the addition of sorafenib for patients with FLT3 -ITD (J. Rubnitz, personal communication, January 2013). CNS leukemia in patients with AML is highly sensitive to chemotherapy, and the addition of cranial irradiation does not decrease CNS relapse compared with systemic and intrathecal chemotherapy. Therefore, cranial irradiation is no longer part of the therapy for patients with AML. However, CNS-positive patients defined on the current COG AML study as having even one blast in the cerebrospinal fluid are treated with additional intrathecal cytarabine until the fluid is clear of blasts and for two additional intrathecal treatments beyond. Furthermore, in pediatrics, the addition of prolonged low-intensity maintenance therapy is associated with lower OS because of more resistant disease at relapse, so maintenance therapy is not used in the current U.S. trials.

References .

Supportive Therapy at Diagnosis and during Therapy

The diagnosis and treatment of childhood AML require a multidisciplinary approach involving pediatric oncologists, hematopathologists, pediatric surgeons, infectious disease specialists, pediatric intensivists, and pediatric nurses. While studies and procedures are initiated to make the diagnosis, special care must be taken to prevent and treat the life-threatening consequences of leukemia and accompanying pancytopenia. The most serious complications of leukemia at diagnosis are infection and bleeding with pancytopenia, tumor lysis syndrome, and leukostasis. The most common causes of death once remission is achieved are relapse, infection, hemorrhage, and cardiac events. The improvement in OS of patients with AML over the past several decades is attributable as much to better supportive care efforts as to more effective treatment regimens.

Infectious Complications.

At the time of initial presentation, up to 40% of patients with leukemia exhibit fever, and most of these patients are neutropenic or have impaired neutrophil function. During remission induction and subsequent cycles of chemotherapy, all AML patients experience profound prolonged neutropenia and are at high risk for life-threatening bacterial infections, particularly viridans group streptococcal infections. Almost all AML patients experience at least one febrile neutropenic episode during therapy, and approximately 40% of these patients will have a documented infection; most commonly these are bacterial infections, followed by viral infections and then fungal infections. Infections account for nearly two thirds of deaths in patients in remission. Therefore supportive care during induction and postremission therapy must pay particular attention to prophylactic, empiric, and treatment coverage for infectious complications. Several strategies are used to prevent infection or decrease mortality from infection. Some centers isolate patients receiving AML therapy in laminar airflow rooms during episodes of neutropenia. The supportive care guidelines for the CCG 2961 study included mandatory hospitalization for all patients, whether febrile or not, until the absolute phagocyte count had risen, and this rule has been strongly recommended in current COG trials. On CCG 2961, almost 50% of patients developed gram-positive infections and approximately 25% developed gram-negative infections across all phases of therapy. Infection-related mortality was 11%, including death from fungus. Bacterial agents included coagulase-negative staphylococci and α-hemolytic streptococci (including viridans streptococci) as well as Klebsiella and Pseudomonas species. To decrease the rate of bacterial infections, some programs routinely prescribe the use of gut decontamination, mouthwashes, or prophylactic antibiotics. A study by SJCRH demonstrated that initiation of antibiotics at the onset of neutropenia prevented sepsis, with the optimal regimen including both vancomycin and ciprofloxacin. This remains an area of controversy because, in some studies, although prophylaxis may decrease the rate of bacterial infection it may also be associated with an increase in drug-resistant organisms. Further investigation is indicated in a prospective randomized fashion before it is widely adopted. For example, in the SJCRH study, an earlier regimen of vancomycin and cefepime resulted in two cases of life-threatening resistant gram-negative bacteria and one of Bacillus cereus , leading to the change to ciprofloxacin for gram-negative coverage. The COG is currently conducting a study of a related fluoroquinolone, levofloxacin, for bacterial prophylaxis in AML and patients with relapsed ALL. Patients will be randomized to receive levofloxacin versus no bacterial prophylaxis from the initiation of chemotherapy for two consecutive cycles. In contrast to the controversy regarding bacterial prophylaxis, there is broad agreement that patients who are febrile at initial presentation and patients who experience febrile neutropenia during therapy should be immediately started empirically on broad-spectrum antibiotics, including vancomycin, once cultures have been obtained. In many centers, vancomycin is discontinued after 48 to 72 hours if there is no evidence of gram-positive bacteremia. Typhlitis, or generalized bacterial infection of the cecum, is more common in children than in adults and is more common in patients treated with doxorubicin instead of daunorubicin. Although this is a potentially life-threatening complication, conservative management with broad-spectrum antibiotics and nothing by mouth is generally preferable to surgical treatment in the setting of profound neutropenia.

AML patients are at high risk for invasive fungal infection, which carries a high risk for mortality. The routine use of fungal prophylaxis is becoming more routine given the intensity of AML protocols. Data from the CCG 2961 study demonstrated Candida and Aspergillus infections in approximately 13% of patients across all phases of therapy and a strong association with infection-related mortality. Some early studies have shown a decrease in some Candida species with fluconazole prophylaxis, but an increase in Candida krusei infections was documented, with no decrease in infection by invasive molds. Two meta-analyses have concluded that there is a reduction in morbidity because of invasive fungal infections when prophylaxis is used for those with prolonged neutropenia and post-HSCT patients. A randomized controlled trial of posaconazole versus fluconazole or itraconazole as fungal prophylaxis in neutropenic patients with AML or MDS has shown a lower rate of invasive fungal infections and improved OS in the posaconazole group. However, posaconazole has not been studied in children younger than age 13 years and, owing to exclusively oral administration, may have limitations during administration of emetogenic AML chemotherapy. Voriconazole has been studied as a prophylactic agent that has activity against both yeast and aspergillus, but the incidence of invasive fungal infections was no different compared with fluconazole or itraconazole. Voriconazole, similar to posaconazole, is metabolized through hepatic CYP450, leading to numerous interactions with other drugs and contributing further to a lack of enthusiasm in using either of these agents prophylactically. Thus fluconazole remains the most frequently used agent for fungal prophylaxis but lacks activity against Aspergillus . Currently the COG is conducting a study randomizing patients with AML to caspofungin versus fluconazole for fungal prophylaxis. Caspofungin is from the echinocandin family of antifungal agents that functions by targeting fungal cell wall biosynthesis of β-1,3-glucan. It is effective against both Candida and Aspergillus species and has safety data in children as young as 3 months of age. Caspofungin is given intravenously and is generally well tolerated but has minimal CNS penetration.

A test to detect galactomannan-containing Aspergillus antigens in the sera of immunocompromised patients has become commercially available. At this point, the galactomannan assay has been used most advantageously to follow the status of invasive aspergillosis and its response to therapy. However, some centers do not use fungal prophylaxis during AML therapy but rather screen twice weekly with the galactomannan assay and use a low threshold for computed tomography (CT) to detect early invasive disease. Whether or not fungal prophylaxis is used, empirical fungal therapy should be initiated for neutropenic patients who remain febrile for longer than 5 days, although some centers use a threshold of 72 hours. Patients who need empiric fungal therapy should be routinely scanned during neutropenia and after the recovery of circulating neutrophils be screened for evidence of fungal disease, and the diagnosis of fungal infection should be made by biopsy whenever feasible. Amphotericin has long been the antifungal agent of choice for empiric therapy, and the liposomal forms appear to be equivalent in efficacy and less toxic to renal function. One prospective randomized trial has shown that caspofungin as empiric fungal therapy is as effective as liposomal amphotericin B and is generally better tolerated. Whereas caspofungin is well tolerated with few drug interactions, as mentioned earlier, it does not cross the blood-brain barrier and thus is ineffective at treating suspected or proven CNS fungal infections. Voriconazole as empiric fungal therapy was compared with amphotericin in a prospective randomized trial and found to provide comparable coverage with less renal toxicity but with more visual changes and hallucinations. Voriconazole is seldom used empirically unless Aspergillus is highly suspected, such as in the case of prolonged febrile neutropenia with pulmonary lesions on imaging.

All patients undergoing AML therapy should receive prophylaxis against Pneumocystis carinii (now called P. jirovecii ) pneumonia (PCP). The first-line agent in PCP prophylaxis is trimethoprim-sulfamethoxazole, but in some patients this is associated with significant allergic reaction or prolonged bone marrow suppression. Dapsone is also effective as PCP prophylaxis in pediatric oncology patients but is associated with significant hemolysis and methemoglobinemia in some patients. Atovaquone has been shown to be highly effective as PCP prophylaxis for pediatric leukemia patients, although it may be associated with gastrointestinal side effects, including pancreatitis. Aerosolized pentamidine has also been shown to be effective as PCP prophylaxis in children with leukemia, although it may be associated with significant bronchospasm and can only be administered to children old enough to comply with the inhalational administration (usually >5 years).

Viral infections can be a significant cause of morbidity and mortality in AML patients as well. CCG 2961 and COG AAML0531 advocate intravenous immune serum globulin (IVIG) administration for low IgG levels, and some centers also recommend respiratory syncytial virus (RSV) prophylaxis for infants. All blood products transfused should be leukoreduced and cytomegalovirus (CMV) negative whenever possible. Acyclovir may be required for patients with recurrent zoster or mucocutaneous herpes simplex.

Growth factors, such as G-CSF and granulocyte-macrophage colony-stimulating factor (GM-CSF), have been used in the past, both as priming agents to synchronize the cell cycle before chemotherapy and as supportive care to decrease the duration of neutropenia after chemotherapy. Trials have shown that although G-CSF does shorten the duration of neutropenia its use does not lead to fewer episodes of febrile neutropenia, fewer microbiologically documented infections, less infection-related mortality, or improved OS. Based on these data, most centers no longer advocate the routine use of growth factors as supportive care during AML therapy.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here