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

Acute myeloid leukemia: Pathogenesis

Acute myeloid leukemia: Pathogenesis
Literature review current through: Jan 2024.
This topic last updated: Mar 08, 2022.

INTRODUCTION — Acute myeloid leukemia (AML) develops as the consequence of a series of genetic changes in a hematopoietic precursor cell. These changes alter normal hematopoietic growth and differentiation, resulting in an accumulation of large numbers of abnormal, immature myeloid cells in the bone marrow and peripheral blood. These cells are capable of dividing and proliferating, but cannot differentiate into mature hematopoietic cells (ie, neutrophils).

This topic will review the cell of origin and the multistep and multicausal pathogenesis of AML. More detailed descriptions of the molecular basis of AML and the genetic abnormalities seen in AML are presented separately, as is a discussion of familial acute leukemia and myelodysplastic syndromes. (See "Acute myeloid leukemia: Molecular genetics" and "Acute myeloid leukemia: Cytogenetic abnormalities" and "Acute myeloid leukemia: Risk factors and prognosis" and "Familial disorders of acute leukemia and myelodysplastic syndromes".)

CELL OF ORIGIN

Normal counterpart — Leukemia is a heterogeneous group of diseases characterized by clonal cells that exhibit maturation defects that correspond to stages in hematopoietic differentiation. Hematopoietic stem cells are multipotent and have the capacity to differentiate into the cells of all 10 blood lineages — erythrocytes, platelets, neutrophils, eosinophils, basophils, monocytes, T and B lymphocytes, natural killer cells, and dendritic cells. In order to sustain hematopoiesis, stem cells are part of a developmental hierarchy capable of three basic functions:

Maintenance in a non-cycling state (ie, not actively progressing through the cell cycle)

Self-renewal, allowing production of additional stem cells

Production of committed progenitor cells

These progenitor cells commit to subsets of myeloid and lymphoid lineages, and ultimately to single developmental pathways, resulting in the expression of the terminally differentiated stage of each cell type (figure 1) [1,2]. (See "Overview of hematopoietic stem cells".)

Normal hematopoiesis is a dynamic, highly regulated process controlled by the combined effects of growth factors that permit cellular proliferation, and nuclear transcription factors that activate specific genetic programs, resulting in commitment to a specific lineage and in terminal differentiation (figure 2). Many of the regulatory growth factors and a number of specific transcription factors have been identified that play critical roles in lineage commitment, and in the subsequent development of the mature lymphoid and myeloid (erythroid, granulocytic/monocytic, and megakaryocytic) lineages [3,4].

A number of genes encoding these transcription factors are involved in recurring chromosomal translocations seen in AML, suggesting that the AML variants arise because the translocations result in significant alterations in regulatory processes controlling growth and differentiation programs [5]. The novel fusion genes created by these translocations will be reviewed separately. (See "Acute myeloid leukemia: Molecular genetics".)

Clonality — AML is a clonal process that develops from a single transformed hematopoietic progenitor cell. Virtually all cases of AML are thought to be preceded by a premalignant proliferative disorder characterized by clonal hematopoiesis without other evidence of a malignancy [6-8]. Such clonal hematopoiesis of indeterminate potential (CHIP) increases in association with age and most commonly involves a mutation in DNMT3A, TET2, or ASXL1 (mutations that are associated with myeloid malignancies). While clonal hematopoiesis was associated with an increased risk of hematologic cancer, the absolute risk of progression is small. (See "Clonal hematopoiesis of indeterminate potential (CHIP) and related disorders of clonal hematopoiesis".)

Evidence for clonal hematopoiesis has also been demonstrated using biochemical assays of X-linked enzyme isoenzymes (eg, glucose-6-phosphate dehydrogenase [G6PD]), standard cytogenetics, recombinant DNA probes, and fluorescence in situ hybridization (FISH) [9-14].

Leukemic stem cells — AML is a heterogeneous disease with leukemic cells of different subtypes resembling normal cells at various stages of maturation. However, there is growing information to support that all leukemias, including AML, appear to be maintained by a pool of self-renewing malignant cells. Based on their ability to serially transfer the disease upon xenotransplantation into immunodeficient mice, it has been hypothesized that limited numbers of cells within the bulk population of leukemic cells have the capacity to function as stem cells that maintain the potential for unlimited self-renewal [15]. These leukemic stem cells (LSC, also leukemia-initiating cells) may be more immature than the majority of circulating leukemic cells and are thought to have originated from cells with existing self-renewal capacity or from progenitors that have re-acquired this stem cell-like property.

According to this hypothesis, the majority of leukemia cells do not have unlimited self-renewal and exhibit some features of partial differentiation depending on the genetic aberration present. However, xenotransplantation of human leukemia cells might not be the best experimental system to examine leukemia stem cell capacity. When lymphomas and leukemias of mouse origin are transplanted into histocompatible mice, a very high frequency (at least 1 in 10) of these cells can transfer the disease to recipient mice suggesting that the low frequency of leukemia-initiating cells observed in xenotransplantation studies may reflect the limited ability of human tumor cells to adapt to growth in a foreign (mouse) milieu [16].

Stage of leukemic transformation

Two models have been proposed to explain the heterogeneity of AML observed at the molecular, cytogenetic, phenotypic, and clinical level.

Transformation to leukemia occurring at one of several developmental stages

Transformation to leukemia occurring within primitive multipotent cells

There is no universal agreement as to which of these two models best reflects the truth.

Transformation at one of several developmental stages — This model proposes that any cell type within the stem cell/progenitor cell hierarchy, from primitive multipotent stem cell to lineage-committed progenitor cell, is susceptible to leukemic transformation, resulting in the expansion of abnormal cells that exhibit different stages of differentiation. For AML, this model predicts that the phenotype of the leukemic stem cells restricted to the granulocytic-monocytic series differs from that of cells in the erythroid or megakaryocytic lineages (figure 1).

The correlation between specific cytogenetic and molecular genetic aberrations and the morphologic appearance of leukemic cells might suggest that the transforming event occurs at different stages of myeloid differentiation. This hypothesis is underscored by the classification for AML, which distinguishes some subtypes of AML based upon the stage of apparent differentiation (eg, AML with minimal differentiation). Support for this model includes flow cytometric/molecular analyses of the leukemic cell in acute promyelocytic leukemia (APL) that suggest that the leukemic cell arises in a committed lineage-restricted, CD34+/CD38+ progenitor cell [17].

Transformation within primitive multipotent cells — A second model proposes that mutations responsible for leukemic transformation and progression occur only in primitive multipotent stem cells, with disease heterogeneity resulting from a variable ability of these primitive stem cells to differentiate and acquire specific phenotypic lineage markers [18,19].

Hematopoietic stem cells express a characteristic cell surface antigen (CD34), and can be further subdivided by the expression of additional cell surface antigens, including CD38 and HLA-DR [20,21]:

CD34+/CD38-/HLA-DR- cells are multipotential hematopoietic stem cells, give rise to mixed-lineage granulocytic-erythroid-megakaryocytic colonies in culture, can repopulate immune deficient mice with normal hematopoietic cells in vivo, and demonstrate self-renewal capacity, as assessed by their ability to be serially transplanted into recipient mice. There are data to suggest that, in some cases of AML, the leukemic stem cell may be quite similar to normal hematopoietic stem cells [22].

CD34+/CD38+/HLA-DR+ cells define a committed population of myeloid progenitor cells [23]. (See "Overview of hematopoietic stem cells".)

Cytogenetic and FISH studies of sorted stem cell compartments from patients with AML evolving from a prior myelodysplastic syndrome and patients with de novo AML have shown that the characteristic cytogenetic abnormality from both groups was present in the CD34+/CD38- multipotential stem cell compartment [24-26]. Similar findings were noted in patients with the 5q- syndrome [27] and monosomy 7 [28], myelodysplastic disorders with differing risks of leukemic transformation. (See "Clinical manifestations, diagnosis, and classification of myelodysplastic syndromes (MDS)".)

More compelling evidence comes from studies in which purified stem cell subpopulations from normal subjects and those with AML were transplanted into mice with severe combined immunodeficiency disease (SCID) [29]. These experiments have detected approximately one SCID mouse leukemia-initiating cell (SL-IC) in 105 AML cells, which can repopulate immune deficient mice with leukemic cells phenotypically identical to those of the AML patient from which they were derived [26,29,30].

Using a non-obese diabetic (NOD)/SCID mouse [31], SL-ICs were found to reside only in the CD34+/CD38- fraction [15]. This was consistent regardless of the AML subtype, lineage markers, or percentage of leukemic blast cells expressing the CD34 antigen. The SL-ICs also demonstrated self-renewal capacity, a requirement for maintenance of the leukemic clone. The uniformity of the leukemic stem cell phenotype strongly suggests that the leukemia initiating transformation and progression-associated genetic events occur in primitive cells and not in committed progenitors. Similar conclusions about the site of the leukemia initiating transformation have been made in acute lymphoblastic leukemia [32].

Additional evidence for the second model derives from the use of a retroviral gene transfer system to express AML1/ETO, a fusion gene linked to the pathogenesis of AML, in normal human hematopoietic stem and progenitor cells [33]. When this fusion gene was expressed in more mature progenitor cells, the result was growth arrest and abrogated colony formation in primary clonogenic assays. On the other hand, AML1/ETO expression in stem cells resulted in their preferential expansion and/or self-renewal [34]. (See "Acute myeloid leukemia: Molecular genetics", section on 'Cooperating mutations in AML'.)

Further supporting evidence comes from studies of clonal hematopoiesis in AML, which indicate that, although the majority of cells derived from the leukemic clone can undergo differentiation to cells of the granulocytic-monocytic lineage, they also may differentiate into cells of the erythroid and/or megakaryocytic pathways [12-14,35]. Such multi-lineage involvement has been noted more frequently in older patients, in secondary AML that arose from myelodysplastic syndrome, or following treatment for another malignancy [14,35].

Protection by stromal cells — AML cells develop in the bone marrow where interaction with the microenvironment may foster chemoresistance [36-39]. As an example, two studies in mouse models of APL demonstrated that dislodgement of leukemic cells from their stromal environment via interruption of CXCR4 signaling resulted in an increased sensitivity of the leukemic cells to chemotherapy [40,41]. Other studies have suggested that this abnormal microenvironment is at least partially due to leukemic cell growth that disrupts normal hematopoietic progenitor cell niches in the bone marrow [37,42].

Suppression of normal hematopoiesis — Pancytopenia is common among patients with AML and many patients initially present with symptoms related to complications of leukopenia, anemia, and thrombocytopenia. Historically, pancytopenia in AML has been attributed to the leukemic cells either displacing or killing normal hematopoietic stem cells in the bone marrow. However, subsequent studies suggest that, in patients with AML, the number of normal hematopoietic stem cells in the bone marrow is normal or increased [43]. Instead, the leukemic cells appear to inhibit the ability of the hematopoietic stem cells to produce more mature hematopoietic cells. When the stem cells were removed from the leukemic environment the ability to produce mature hematopoietic cells was restored.

Aberrant cellular metabolism — Altered cellular metabolism is a hallmark of cancer and may contribute to AML initiation and maintenance [44-46].

As an example, up to 15 to 20 percent of patients with AML have mutations in IDH1 or IDH2 that result in neomorphic enzymatic activity and production of the onco-metabolite, 2-hydroxyglutarate (2HG) from alpha-ketoglutarate [44]. 2HG inhibits multiple alpha-ketoglutarate-dependent dioxygenase reactions, leads to aberrant DNA hypermethylation and a differentiation block in myeloid precursors, and promotes leukemogenesis. Allosteric inhibitors of the mutant IDH isoforms, ivosidenib (IDH1) and enasidenib (IDH2) can overcome the differentiation block and have been approved for treatment.

TWO-HIT HYPOTHESIS OF LEUKEMOGENESIS — Progression to acute leukemia may require a series of genetic events beginning with clonal expansion of a transformed leukemic stem cell [47-49]. The specific mutational event(s) required for this progression are not currently well defined. Single nucleotide polymorphism (SNP) studies have been performed on tumor samples from adults and children with AML as a genomic screen to locate previously unidentified genes that may play a role in the pathobiology of AML [50,51].

The "two-hit hypothesis" of leukemogenesis implies that AML is the consequence of at least two mutations, one conferring a proliferative advantage (class I mutations) and another impairing hematopoietic differentiation (class II mutations) [52]. Type I mutations include those of FLT3-ITD, K-RAS mutations, and KIT mutations, while mutations in CEBPA are type II abnormalities [53]. (See "Acute myeloid leukemia: Risk factors and prognosis".)

Important insights have been obtained from human leukemias:

In chronic phase chronic myeloid leukemia (CML), all leukemic cells contain t(9;22), resulting in formation of the BCR/ABL fusion gene [54], whose product is of critical significance in the pathogenesis of CML [55]. As the disease progresses, additional cytogenetic abnormalities are acquired [56], which are often accompanied by loss of important tumor suppressor genes such as p53 [57]. (See "Cellular and molecular biology of chronic myeloid leukemia", section on 'The BCR-ABL1 fusion protein' and "Cellular and molecular biology of chronic myeloid leukemia", section on 'Progression to acute phase CML'.)

A variety of clonality studies have shown that patients with AML in clinical remission may still have clonal, rather than polyclonal, hematopoiesis [12,58-60]. Such clonal remission may represent the presence of a "preleukemic stem cell" that has undergone an initial transforming event but has not acquired the additional mutation(s) essential to progression to overt leukemia. In these cases, it is presumed that the transformed, overtly leukemic cell probably represented a subclone of the original "preleukemic stem cell" which secondarily acquired the additional genetic mutations required for the definitive block in differentiation and manifestation of the leukemic phenotype. Although some studies suggest that "clonal" remissions may be the result of skewed Lyonization (preferential inactivation of one X chromosome over another) [61,62], more carefully controlled studies suggest that clonal remissions do occur following treatment for AML [63,64].

On average, AML clones have 8 to 13 mutations found within the coding regions of the genome. The accumulation of these lesions in a step-wise process within a hematopoietic stem cell was demonstrated in a study that analyzed gene mutations found in primary tumor-relapse pairs of de novo AML and patient-matched skin samples [65]. Identification of individual cells containing subsets of these mutations allowed for the identification of mutations that occurred early and late in the process. This finding suggests not only that several hits are required for the development of AML, but also that relapsed disease can represent the further replication of the original dominant clone, the emergence of a minor clone present at diagnosis [66,67], or further evolution of a preleukemic clone after treatment (figure 3).

Whole genome or whole exome sequencing of 200 cases of de novo AML reported an average of 13 gene mutations per tumor [68]. Of these, each tumor had an average of five genes known to be one of a group of 23 genes recurrently mutated in AML that can be broadly grouped into nine categories of genes thought to be involved in leukemogenesis. Mutations were found in genes associated with transcription-factor fusions (18 percent), nucleophosmin (27 percent), tumor suppression (16 percent), DNA-methylation (44 percent), signaling (59 percent), chromatin-modification (30 percent), myeloid transcription factor (22 percent), the cohesin-complex (13 percent), and the spliceosome complex (14 percent). Some mutation pairs occurred more commonly than expected (eg, NPM1 and FLT3), suggesting synergy, while others were mutually exclusive, suggesting duplicative pathways. (See "Acute myeloid leukemia: Molecular genetics", section on 'Gene mutations'.)

In one study of seven patients with AML that had evolved from myelodysplastic syndrome, approximately 85 percent of bone marrow cells were clonal at the time of myelodysplastic syndrome diagnosis [69]. Whole genome sequencing of paired skin and bone marrow samples identified 11 recurrently mutated genes. Genotyping of bone marrow samples from the same patients collected at the time of AML diagnosis identified those mutations that were present at the time of myelodysplastic syndrome diagnosis (ie, NPM1, RUNX1, SMC3, STAG2, TP53, U2AF1, UMODL1, and ZSWIM4) and those that developed subsequently (ie, CDH23, PTPN11, WT1).

A retrospective study that compared mutational analyses of paired diagnostic and relapse leukemia samples from 69 children with AML reported that 38 percent of patients had a change in their mutation status between diagnosis and relapse. Patients whose AML demonstrated a type I/II mutation at relapse had a shorter time to relapse. Consistent with the two-hit theory, expression of a chimeric protein represents only one of the genetic modifications necessary for the development of cancer and leukemia, and that the affected cell requires additional mutational events in order to express the transformed phenotype. (See "Acute myeloid leukemia: Cytogenetic abnormalities", section on 't(8;21); RUNX1::RUNX1T1'.)

As an example, a number of examples indicate that the presence of cells with the RUNX1/RUNX1T1 fusion transcript may not be sufficient, in itself, to result in AML:

Transgenic mice expressing RUNX1/RUNX1T1 were healthy throughout their lifespan, developing AML only after exposure to an alkylating mutagen [70], or in cooperation with Wilms tumor gene (WT1) overexpression [71].

Remission bone marrow samples from patients with de novo AML (FAB -M2) with t(8;21)(q22;q22) and the RUNX1/RUNX1T1 fusion transcript have been found to harbor the aberrant fusion transcript for as many as eight years following cessation of all chemotherapy [72].

The RUNX1/RUNX1T1 fusion transcript has been detected in bone marrow samples from patients in remission following allogeneic bone marrow transplantation for AML [73].

In five children who developed AML with t(8;21) at 3 to 12 years of age, in whom archived blood samples for metabolic studies (Guthrie cards) were available, RUNX1/RUNX1T1 sequences were detected at birth [74]. Of interest, similar observations were made in three children developing acute lymphoblastic leukemia at three to five years of age with t(12;21) [75].

As noted above, RUNX1/RUNX1T1 is not immediately leukemogenic in either animals or man, and may require a "second hit" for the development of AML [76,77]. However, the presence of an alternatively spliced isoform AML1/ETO9a has been shown to be present in the majority of patients with t(8;21) [78]. Expression of this alternative isoform leads to the rapid development of leukemia in a mouse model, and coexpression of RUNX1/RUNX1T1 and AML1/ETO9a results in the substantially earlier onset of AML and blocks myeloid cell maturation at a more immature stage. These early results suggest that fusion proteins from alternatively spliced isoforms resulting from a chromosomal translocation may work together to induce this malignancy.

MECHANISMS OF GENETIC DAMAGE — Genetic changes associated with leukemogenesis can occur following chemotherapy, ionizing radiation, chemical exposure, and infection with retroviruses. In addition, certain familial disorders are associated with an increased incidence of AML.

While environmental and hereditary conditions serve as excellent models for obtaining insights into the molecular pathogenesis of AML, it must be emphasized that the vast majority of patients with de novo AML show no evidence of any of these risk factors, and the etiologic factors contributing to the development of AML remain unknown. Interestingly, in a series of 127 patients with a previous primary malignancy and secondary AML, 30 percent did not receive any chemotherapy or radiation treatment prior to the development of AML [79].

Chemotherapy-induced AML — The development of myelodysplastic syndromes (MDS) and AML following chemotherapy for a variety of malignancies (eg, breast cancer, Hodgkin lymphoma) is an unfortunate complication of curative treatment strategies [80], such as dose-intensive therapy with or without hematopoietic cell transplantation and growth factor support [80-84]. This identification of an increasing incidence of therapy-related AML (t-AML) in an attempt to improve cure rates emphasizes the critical importance of understanding the underlying pathogenetic mechanisms for development of t-AML [85,86]. (See "Secondary cancers after hematopoietic cell transplantation" and "Second malignancies after treatment of classic Hodgkin lymphoma", section on 'Acute leukemia' and "Overview of long-term complications of therapy in breast cancer survivors and patterns of relapse", section on 'Risks associated with chemotherapy'.)

t-AML typically develops following alkylating agent-induced damage, at a median of three to five years following therapy for the primary malignancy and is usually associated with an antecedent myelodysplastic disorder [87]. This latency period suggests that multiple mutational events are involved in the development of the malignant phenotype [47].

Clonal chromosomal abnormalities have been reported in the majority of cases of t-AML (see "Acute myeloid leukemia: Cytogenetic abnormalities", section on 'Therapy-related myeloid neoplasms'). The most frequently reported abnormalities involve complete loss or interstitial deletions of the long arm of chromosomes 7 and/or 5.

Other therapy-related leukemias are associated with rearrangements of the MLL gene in chromosome band 11q23 [88]. AML associated with 11q23 often develops after treatment with drugs that target DNA-topoisomerase II (eg, epipodophyllotoxins, anthracyclines) with a very short latency of 12 to 18 months following treatment, and are not typically associated with an antecedent MDS [89-91]. (See "Acute myeloid leukemia: Molecular genetics", section on 'Involvement of the KMT2A (MLL) locus' and "Acute myeloid leukemia: Cytogenetic abnormalities", section on 't(9;11); KMT2A::MLLT3'.)

Accelerated telomere loss may precede the development of t-MDS/AML after autologous hematopoietic cell transplantation resulting in genetic instability and thereby contributing to the leukemic transformation [92,93].

Genetic polymorphisms of a number of drug-metabolizing enzymes may alter the risk of t-AML [86,94,95]. As an example, polymorphisms in genes that encode glutathione S-transferases (GST), which detoxify potentially mutagenic chemotherapeutic agents, may increase susceptibility to t-AML as well as MDS [94,96,97]. In one study, relative to de novo AML, the GSTP1 codon 105 Val allele occurred more often among patients with t-AML with prior exposure to chemotherapy, particularly those with exposure to known GSTP1 substrates (odds ratio 4.3; 95% CI 1.4-13), and not among those t-AML patients with prior exposure to radiotherapy alone.

Ionizing radiation — Ionizing radiation shares with alkylating agents the ability to damage DNA, usually by inducing double strand breaks that may cause the mutations, deletions, or translocations required for hematopoietic stem cell transformation [80,98]. As examples, an increased incidence of AML, which may have been directly proportional to the radiation exposure [99], has been noted in atomic bomb survivors [100] as well as in radiologists and radiologic technologists chronically exposed to high levels of radiation in the period before 1950 [101].

Ionizing radiation used in the treatment of malignancies (eg, Hodgkin lymphoma, breast cancer, uterine cancer, lung cancer) has also been linked to the development of AML [102]. This risk appears to be quite low when radiation alone is used as treatment, and is associated with age of the patient, and doses of more than 20 Gy [103,104]. Whether irradiation adds to the risk of t-AML associated with chemotherapy remains controversial. Although some studies suggest that the risk of development of AML is significantly increased when the two modalities are combined, other studies demonstrated that high doses of radiotherapy confined to small volumes in combination with chemotherapy did not significantly increase leukemogenic risk [105-108].

It is unknown whether ionizing radiation used for medical examination, such as computed tomography (CT) scans, results in an increased risk of leukemia in adults. However, there is epidemiologic evidence that exposure to x-rays and gamma rays during childhood is associated with a small absolute increase in the incidence of leukemia. In a cohort study of individuals who had CT scans when they were younger than 22 years of age, compared with those who received a cumulative radiation dose <5 mGy, the risk of subsequent leukemia was tripled for those who received a cumulative radiation dose ≥30 mGy, which is equivalent to approximately 5 to 10 head CT scans [109]. Radiation risk in children is compounded with their longer lifespan following exposure so that there is a longer time over which radiation-induced cancers can occur.

Chemical exposure — Exposure to high levels of benzene has been associated with a higher risk of developing AML [110,111]. Relatively low-level exposure to benzene by petroleum distribution workers has been associated with an increased risk of developing MDS, but not AML [112,113]. The risk of developing a myeloid malignancy after benzene exposure appears to be dose-related and it is unknown whether there is any safe threshold for benzene exposure [114-116]. A potential association between formaldehyde exposure and AML has been controversial, with conflicting data from meta-analyses of epidemiologic studies [117-120]. Except for special groups exposed to high levels of benzene or radiation, the reported risks associated with occupation and chemicals have generally been less than twofold [121].

The presence of RAS mutations in patients with AML has been associated with specific occupational exposure to chemicals, suggesting that these exposures may induce genetic damage culminating in acute leukemia [122]. In a case-control study, cigarette smoking was associated with only a modest increase in leukemic risk; however, a twofold increase in risk for AML was noted in study subjects over the age of 60 [123].

Polymorphisms resulting in inactivation of NAD(P)H:quinone oxidoreductase 1 (NQO1, originally called DT-diaphorase), an enzyme which detoxifies quinones and reduces oxidative stress, have been associated with an increased risk of de novo [124] and therapy-related acute leukemia [125], as well as a greater risk of benzene-induced hematotoxicity and leukemia [126]. For de novo AML, the most significant effect of low or null NQO1 activity was observed among patients with chromosomal translocations and inversions (odds ratio: 2.4), and was especially high for those with inv(16) (odds ratio: 8.1) [124].

Genetic polymorphisms in the microsomal epoxide hydrolase (HYL1) gene, an enzyme involved in benzene metabolism, have also been associated with an increased incidence of AML. Data from one study suggest that smoking and/or exposure to a carcinogen that is activated by HYL1, such as benzene, may be important in subsets of patients with AML, such as males with t(8;21) or -7/del(7q) [127,128].

Infections — Patients with AML often have infections at the time of diagnosis, some of which are severe. However, there are limited data regarding whether infections play a role in the pathogenesis of AML. A large population-based retrospective registry study from Sweden suggested that a personal history of infection was associated with an increased risk of developing AML or myelodysplastic syndrome [129]. While interesting, further study is needed to confirm these results and to study potential mechanisms. Studies have also suggested that chronic acetaminophen use might increase the risk of developing AML [130,131].

In a number of animal models, retroviruses have been demonstrated to play an important role in leukemogenesis, and the human T-lymphotropic virus type I (HTLV-I) is associated with adult T cell leukemia-lymphoma [132]. In AML, however, despite extensive investigation, there has been no clear association of a retrovirus with leukemogenesis [133]. (See "Acute myeloid leukemia: Molecular genetics", section on 'Animal models of AML' and "Clinical manifestations, pathologic features, and diagnosis of adult T cell leukemia-lymphoma".)

Familial acute leukemia and myelodysplasia syndromes — Most patients diagnosed with AML do not have a clear familial predisposition towards the development of MDS or leukemia [134]. However, rare pedigrees with multiple cases of AML have been described and termed familial leukemia. Familial leukemia can occur in the context of a medical syndrome in which AML is one component of the overall disease, or it can occur as an isolated leukemia not specifically associated with co-morbid conditions [135]. Inherited disorders associated with defective DNA repair have also been associated with a high incidence of hematologic malignancies, including AML. The adoption of next-generation sequencing technologies has facilitated the rapid discovery of an increasing number of mutations associated with familial leukemia predisposition syndrome [136]. Familial acute leukemia and myelodysplastic syndromes are discussed in more detail separately. (See "Familial disorders of acute leukemia and myelodysplastic syndromes".)

ROLE OF HEMATOPOIETIC GROWTH FACTORS — Using in vitro clonogenic assays, leukemic cells have been shown to proliferate in response to many of the endogenous hematopoietic growth factors critical for normal hematopoiesis, including granulocyte, granulocyte-monocyte, macrophage, and stem cell colony-stimulating factors (G-CSF, GM-CSF, M-CSF, SCF), interleukin 3, and Flt3 ligand (flt3-L) [137-142], with combinations of these factors producing a synergistic growth response.

Mutations in the G-CSF receptor gene — Mutations in the granulocyte colony-stimulating factor (G-CSF) receptor gene have been described in patients with severe congenital neutropenia [143]. A number of patients with severe congenital neutropenia with documented nonsense mutations in the G-CSF receptor have developed AML, supporting the notion that defective signaling function by the aberrant receptor increased the susceptibility to AML [144]. It is also possible that mutations in the G-CSF receptor predispose to the myelodysplastic syndrome (MDS) [145], or to AML via a resistance to apoptosis [146], allowing more time for a "second hit" mutation to occur. (See "Congenital neutropenia".)

In a substantial number of patients with AML, autonomous growth has been reported to occur as a result of autocrine or paracrine stimulation by a number of factors, including G-CSF, GM-CSF, IL-1b, and IL-6 [147,148]. Several investigators have noted that the presence of autonomous growth characteristics by AML cells grown in vitro correlates with lower remission rates, and poor survival [149,150]. In a multivariate analysis, expression by leukemic blasts of c-mpl, the receptor for thrombopoietin, correlated with a significantly decreased remission duration in patients with AML [151].

Several explanations for these observations have been proposed. One possibility is that the acquisition of autonomous growth capability allows AML cells to become more aggressive, by making them independent of stromal cell production of essential growth factors [150]. Others have suggested that autonomous production of growth factors, such as GM-CSF, may reduce the cytotoxicity of chemotherapy agents, by altering intracellular drug metabolism [152]. Some data suggest that exogenously administered, as well as endogenously produced, hematopoietic growth factors not only stimulate in vitro growth and proliferation, but also inhibit apoptosis of AML cells [153,154].

As a result of in vitro data demonstrating the growth-promoting effects of a variety of cytokines on AML cells, one of the controversies in the treatment of AML has centered on the use of hematopoietic growth factors during or following induction chemotherapy. The desired goal of reducing the toxicity of treatment and the duration of cytopenia with exogenous administration of G-CSF or GM-CSF has been tempered by concerns about their leukemogenic potential. A number of large, randomized clinical trials in AML demonstrate variable clinical efficacy of these growth factors with respect to significant decreases in morbidity and mortality [155]. While different conclusions have been reached regarding clinical efficacy, there is consensus about the safety and lack of increased leukemogenic potential of these growth factors when they are administered following induction or consolidation chemotherapy. Although similar theoretical arguments have been made against the use of growth factors in MDS, they appear to be safe and have a role in the treatment of selected patients. (See "Myelodysplastic syndromes/neoplasms (MDS): Management of hematologic complications in lower-risk MDS", section on 'Thrombocytopenia'.)

Exogenous growth factors — The therapeutic use of granulocyte colony-stimulating factor (G-CSF) may result in an increased risk of AML or MDS, although the magnitude of the risk appears to be small and may be dose related. Initial suggestions of this association came from retrospective analyses and prospective trials mostly involving its use in breast cancer [156,157]. A systemic review which included data from 12,104 patients enrolled in randomized controlled clinical trials of cancer chemotherapy with G-CSF reported the following at a median follow-up time of 54 months [158]:

The estimated relative risk for AML/MDS among patients assigned to receive G-CSF was 1.9 with an absolute increase in risk of 0.4 percent.

The estimated relative risk for mortality among patients assigned to receive G-CSF was 0.9 with an estimated absolute decrease of 3.4 percent.

Despite a suggested increased risk of AML/MDS among patients receiving G-CSF, patients who received G-CSF demonstrated superior survival. By its nature, this analysis of randomized trials avoids many of the potential biases that confound the earlier retrospective studies; however, this study design cannot differentiate between an increase in rates of AML/MDS due to the use of G-CSF versus an increase due to the use of dose-intensified systemic chemotherapy. Further studies are required to address this issue. (See "Acute myeloid leukemia: Cytogenetic abnormalities", section on 'Therapy-related myeloid neoplasms'.)

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

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

Beyond the Basics topics (see "Patient education: Acute myeloid leukemia (AML) treatment in adults (Beyond the Basics)")

SUMMARY

Acute myeloid leukemia (AML) develops as the consequence of a series of genetic changes in a hematopoietic precursor cell. These changes alter normal hematopoietic growth and differentiation, resulting in an accumulation of large numbers of abnormal, immature myeloid cells in the bone marrow and peripheral blood. These cells are capable of dividing and proliferating, but cannot differentiate into mature hematopoietic cells (ie, neutrophils).

AML is a heterogeneous group of diseases characterized by clonal cells that exhibit maturation defects that correspond to stages in hematopoietic differentiation. (See 'Cell of origin' above.)

All leukemias, including AML, appear to be maintained by a pool of self-renewing malignant cells. These leukemic stem cells (also leukemia-initiating cells) may be more immature than the majority of circulating leukemic cells and are thought to have originated from cells with existing self-renewal capacity or from progenitors that have reacquired this stem cell-like property. (See 'Leukemic stem cells' above.)

The "two-hit hypothesis" of leukemogenesis implies that AML is the consequence of at least two mutations, one conferring a proliferative advantage (class I mutations) and another impairing hematopoietic differentiation (class II mutations). (See 'Two-hit hypothesis of leukemogenesis' above.)

Leukemogenic mutations can occur following chemotherapy, ionizing radiation, chemical exposure, and infection with retroviruses. In addition, certain familial disorders are associated with an increased incidence of AML. (See 'Mechanisms of genetic damage' above.)

  1. Godin IE, Garcia-Porrero JA, Coutinho A, et al. Para-aortic splanchnopleura from early mouse embryos contains B1a cell progenitors. Nature 1993; 364:67.
  2. Botnick LE, Hannon EC, Hellman S. Nature of the hemopoietic stem cell compartment and its proliferative potential. Blood Cells 1979; 5:195.
  3. Orkin SH. Transcription factors and hematopoietic development. J Biol Chem 1995; 270:4955.
  4. Tenen DG, Hromas R, Licht JD, Zhang DE. Transcription factors, normal myeloid development, and leukemia. Blood 1997; 90:489.
  5. Rabbitts TH. Chromosomal translocations in human cancer. Nature 1994; 372:143.
  6. Genovese G, Kähler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014; 371:2477.
  7. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014; 371:2488.
  8. Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014; 20:1472.
  9. Fialkow PJ, Singer JW, Adamson JW, et al. Acute nonlymphocytic leukemia: expression in cells restricted to granulocytic and monocytic differentiation. N Engl J Med 1979; 301:1.
  10. Fialkow PJ, Singer JW, Adamson JW, et al. Acute nonlymphocytic leukemia: heterogeneity of stem cell origin. Blood 1981; 57:1068.
  11. Ferraris AM, Canepa L, Mareni C, et al. Reexpression of normal stem cells in erythroleukemia during remission. Blood 1983; 62:177.
  12. Fearon ER, Burke PJ, Schiffer CA, et al. Differentiation of leukemia cells to polymorphonuclear leukocytes in patients with acute nonlymphocytic leukemia. N Engl J Med 1986; 315:15.
  13. Keinänen M, Griffin JD, Bloomfield CD, et al. Clonal chromosomal abnormalities showing multiple-cell-lineage involvement in acute myeloid leukemia. N Engl J Med 1988; 318:1153.
  14. van Lom K, Hagemeijer A, Smit EM, Löwenberg B. In situ hybridization on May-Grünwald Giemsa-stained bone marrow and blood smears of patients with hematologic disorders allows detection of cell-lineage-specific cytogenetic abnormalities. Blood 1993; 82:884.
  15. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997; 3:730.
  16. Kelly PN, Dakic A, Adams JM, et al. Tumor growth need not be driven by rare cancer stem cells. Science 2007; 317:337.
  17. Turhan AG, Lemoine FM, Debert C, et al. Highly purified primitive hematopoietic stem cells are PML-RARA negative and generate nonclonal progenitors in acute promyelocytic leukemia. Blood 1995; 85:2154.
  18. McCulloch EA. Stem cells in normal and leukemic hemopoiesis (Henry Stratton Lecture, 1982). Blood 1983; 62:1.
  19. Bonnet D. Normal and leukaemic stem cells. Br J Haematol 2005; 130:469.
  20. Civin CI, Strauss LC, Brovall C, et al. Antigenic analysis of hematopoiesis. III. A hematopoietic progenitor cell surface antigen defined by a monoclonal antibody raised against KG-1a cells. J Immunol 1984; 133:157.
  21. Terstappen LW, Huang S, Safford M, et al. Sequential generations of hematopoietic colonies derived from single nonlineage-committed CD34+CD38- progenitor cells. Blood 1991; 77:1218.
  22. Stubbs MC, Armstrong SA. Therapeutic implications of leukemia stem cell development. Clin Cancer Res 2007; 13:3439.
  23. Huang S, Terstappen LW. Formation of haematopoietic microenvironment and haematopoietic stem cells from single human bone marrow stem cells. Nature 1992; 360:745.
  24. Mehrotra B, George TI, Kavanau K, et al. Cytogenetically aberrant cells in the stem cell compartment (CD34+lin-) in acute myeloid leukemia. Blood 1995; 86:1139.
  25. Haase D, Feuring-Buske M, Könemann S, et al. Evidence for malignant transformation in acute myeloid leukemia at the level of early hematopoietic stem cells by cytogenetic analysis of CD34+ subpopulations. Blood 1995; 86:2906.
  26. Levis M, Murphy KM, Pham R, et al. Internal tandem duplications of the FLT3 gene are present in leukemia stem cells. Blood 2005; 106:673.
  27. Nilsson L, Astrand-Grundström I, Arvidsson I, et al. Isolation and characterization of hematopoietic progenitor/stem cells in 5q-deleted myelodysplastic syndromes: evidence for involvement at the hematopoietic stem cell level. Blood 2000; 96:2012.
  28. Miura I, Kobayashi Y, Takahashi N, et al. Involvement of natural killer cells in patients with myelodysplastic syndrome carrying monosomy 7 revealed by the application of fluorescence in situ hybridization to cells collected by means of fluorescence-activated cell sorting. Br J Haematol 2000; 110:876.
  29. Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994; 367:645.
  30. Sutherland H, Blair A, Vercauteren S, Zapf R. Detection and clinical significance of human acute myeloid leukaemia progenitors capable of long-term proliferation in vitro. Br J Haematol 2001; 114:296.
  31. Bhatia M, Wang JC, Kapp U, et al. Purification of primitive human hematopoietic cells capable of repopulating immune-deficient mice. Proc Natl Acad Sci U S A 1997; 94:5320.
  32. Cox CV, Evely RS, Oakhill A, et al. Characterization of acute lymphoblastic leukemia progenitor cells. Blood 2004; 104:2919.
  33. Mulloy JC, Cammenga J, MacKenzie KL, et al. The AML1-ETO fusion protein promotes the expansion of human hematopoietic stem cells. Blood 2002; 99:15.
  34. Tonks A, Pearn L, Tonks AJ, et al. The AML1-ETO fusion gene promotes extensive self-renewal of human primary erythroid cells. Blood 2003; 101:624.
  35. Fialkow PJ, Singer JW, Raskind WH, et al. Clonal development, stem-cell differentiation, and clinical remissions in acute nonlymphocytic leukemia. N Engl J Med 1987; 317:468.
  36. Konopleva M, Konoplev S, Hu W, et al. Stromal cells prevent apoptosis of AML cells by up-regulation of anti-apoptotic proteins. Leukemia 2002; 16:1713.
  37. Colmone A, Amorim M, Pontier AL, et al. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science 2008; 322:1861.
  38. Lane SW, Scadden DT, Gilliland DG. The leukemic stem cell niche: current concepts and therapeutic opportunities. Blood 2009; 114:1150.
  39. Ustun C, Miller JS, Munn DH, et al. Regulatory T cells in acute myelogenous leukemia: is it time for immunomodulation? Blood 2011; 118:5084.
  40. Zeng Z, Shi YX, Samudio IJ, et al. Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML. Blood 2009; 113:6215.
  41. Nervi B, Ramirez P, Rettig MP, et al. Chemosensitization of acute myeloid leukemia (AML) following mobilization by the CXCR4 antagonist AMD3100. Blood 2009; 113:6206.
  42. Cheng H, Hao S, Liu Y, et al. Leukemic marrow infiltration reveals a novel role for Egr3 as a potent inhibitor of normal hematopoietic stem cell proliferation. Blood 2015; 126:1302.
  43. Miraki-Moud F, Anjos-Afonso F, Hodby KA, et al. Acute myeloid leukemia does not deplete normal hematopoietic stem cells but induces cytopenias by impeding their differentiation. Proc Natl Acad Sci U S A 2013; 110:13576.
  44. Cairns RA, Mak TW. Oncogenic isocitrate dehydrogenase mutations: mechanisms, models, and clinical opportunities. Cancer Discov 2013; 3:730.
  45. Abdel-Wahab O, Levine RL. Metabolism and the leukemic stem cell. J Exp Med 2010; 207:677.
  46. Dembitz V, Gallipoli P. The Role of Metabolism in the Development of Personalized Therapies in Acute Myeloid Leukemia. Front Oncol 2021; 11:665291.
  47. Jacobs A. Leukaemia Research Fund annual guest lecture 1990. Genetics lesions in preleukaemia. Leukemia 1991; 5:277.
  48. Ley TJ, Mardis ER, Ding L, et al. DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome. Nature 2008; 456:66.
  49. Mardis ER, Ding L, Dooling DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med 2009; 361:1058.
  50. Radtke I, Mullighan CG, Ishii M, et al. Genomic analysis reveals few genetic alterations in pediatric acute myeloid leukemia. Proc Natl Acad Sci U S A 2009; 106:12944.
  51. Walter MJ, Payton JE, Ries RE, et al. Acquired copy number alterations in adult acute myeloid leukemia genomes. Proc Natl Acad Sci U S A 2009; 106:12950.
  52. Reilly JT. Pathogenesis of acute myeloid leukaemia and inv(16)(p13;q22): a paradigm for understanding leukaemogenesis? Br J Haematol 2005; 128:18.
  53. Bachas C, Schuurhuis GJ, Hollink IH, et al. High-frequency type I/II mutational shifts between diagnosis and relapse are associated with outcome in pediatric AML: implications for personalized medicine. Blood 2010; 116:2752.
  54. Kurzrock R, Gutterman JU, Talpaz M. The molecular genetics of Philadelphia chromosome-positive leukemias. N Engl J Med 1988; 319:990.
  55. Daley GQ, Van Etten RA, Baltimore D. Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science 1990; 247:824.
  56. Rubin CM, Larson RA, Anastasi J, et al. t(3;21)(q26;q22): a recurring chromosomal abnormality in therapy-related myelodysplastic syndrome and acute myeloid leukemia. Blood 1990; 76:2594.
  57. Feinstein E, Cimino G, Gale RP, et al. p53 in chronic myelogenous leukemia in acute phase. Proc Natl Acad Sci U S A 1991; 88:6293.
  58. Fialkow PJ, Janssen JW, Bartram CR. Clonal remissions in acute nonlymphocytic leukemia: evidence for a multistep pathogenesis of the malignancy. Blood 1991; 77:1415.
  59. Bartram CR, Ludwig WD, Hiddemann W, et al. Acute myeloid leukemia: analysis of ras gene mutations and clonality defined by polymorphic X-linked loci. Leukemia 1989; 3:247.
  60. Busque L, Gilliland DG. Clonal evolution in acute myeloid leukemia. Blood 1993; 82:337.
  61. Gale RE, Mein CA, Linch DC. Quantification of X-chromosome inactivation patterns in haematological samples using the DNA PCR-based HUMARA assay. Leukemia 1996; 10:362.
  62. Busque L, Mio R, Mattioli J, et al. Nonrandom X-inactivation patterns in normal females: lyonization ratios vary with age. Blood 1996; 88:59.
  63. Jowitt SN, Liu Yin JA, Saunders MJ, Lucas GS. Clonal remissions in acute myeloid leukaemia are commonly associated with features of trilineage myelodysplasia during remission. Br J Haematol 1993; 85:698.
  64. Jinnai I, Nagai K, Yoshida S, et al. Incidence and characteristics of clonal hematopoiesis in remission of acute myeloid leukemia in relation to morphological dysplasia. Leukemia 1995; 9:1756.
  65. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 2012; 481:506.
  66. Parkin B, Ouillette P, Li Y, et al. Clonal evolution and devolution after chemotherapy in adult acute myelogenous leukemia. Blood 2013; 121:369.
  67. Bochtler T, Stölzel F, Heilig CE, et al. Clonal heterogeneity as detected by metaphase karyotyping is an indicator of poor prognosis in acute myeloid leukemia. J Clin Oncol 2013; 31:3898.
  68. Cancer Genome Atlas Research Network, Ley TJ, Miller C, et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013; 368:2059.
  69. Walter MJ, Shen D, Ding L, et al. Clonal architecture of secondary acute myeloid leukemia. N Engl J Med 2012; 366:1090.
  70. Yuan Y, Zhou L, Miyamoto T, et al. AML1-ETO expression is directly involved in the development of acute myeloid leukemia in the presence of additional mutations. Proc Natl Acad Sci U S A 2001; 98:10398.
  71. Nishida S, Hosen N, Shirakata T, et al. AML1-ETO rapidly induces acute myeloblastic leukemia in cooperation with the Wilms tumor gene, WT1. Blood 2006; 107:3303.
  72. Nucifora G, Larson RA, Rowley JD. Persistence of the 8;21 translocation in patients with acute myeloid leukemia type M2 in long-term remission. Blood 1993; 82:712.
  73. Jurlander J, Caligiuri MA, Ruutu T, et al. Persistence of the AML1/ETO fusion transcript in patients treated with allogeneic bone marrow transplantation for t(8;21) leukemia. Blood 1996; 88:2183.
  74. Wiemels JL, Xiao Z, Buffler PA, et al. In utero origin of t(8;21) AML1-ETO translocations in childhood acute myeloid leukemia. Blood 2002; 99:3801.
  75. Hjalgrim LL, Madsen HO, Melbye M, et al. Presence of clone-specific markers at birth in children with acute lymphoblastic leukaemia. Br J Cancer 2002; 87:994.
  76. Kuchenbauer F, Schnittger S, Look T, et al. Identification of additional cytogenetic and molecular genetic abnormalities in acute myeloid leukaemia with t(8;21)/AML1-ETO. Br J Haematol 2006; 134:616.
  77. Krejci O, Wunderlich M, Geiger H, et al. p53 signaling in response to increased DNA damage sensitizes AML1-ETO cells to stress-induced death. Blood 2008; 111:2190.
  78. Yan M, Kanbe E, Peterson LF, et al. A previously unidentified alternatively spliced isoform of t(8;21) transcript promotes leukemogenesis. Nat Med 2006; 12:945.
  79. Pagana L, Pulsoni A, Tosti ME, et al. Clinical and biological features of acute myeloid leukaemia occurring as second malignancy: GIMEMA archive of adult acute leukaemia. Br J Haematol 2001; 112:109.
  80. Levine EG, Bloomfield CD. Leukemias and myelodysplastic syndromes secondary to drug, radiation, and environmental exposure. Semin Oncol 1992; 19:47.
  81. DeCillis A, Anderson S, Wickerham DL, et al. Acute myeloid leukemia in NSABP-25. Proc ASCO 1995; 14:98.
  82. Miller JS, Arthur DC, Litz CE, et al. Myelodysplastic syndrome after autologous bone marrow transplantation: an additional late complication of curative cancer therapy. Blood 1994; 83:3780.
  83. Stone RM, Neuberg D, Soiffer R, et al. Myelodysplastic syndrome as a late complication following autologous bone marrow transplantation for non-Hodgkin's lymphoma. J Clin Oncol 1994; 12:2535.
  84. Darrington DL, Vose JM, Anderson JR, et al. Incidence and characterization of secondary myelodysplastic syndrome and acute myelogenous leukemia following high-dose chemoradiotherapy and autologous stem-cell transplantation for lymphoid malignancies. J Clin Oncol 1994; 12:2527.
  85. Seedhouse C, Russell N. Advances in the understanding of susceptibility to treatment-related acute myeloid leukaemia. Br J Haematol 2007; 137:513.
  86. Guillem V, Tormo M. Influence of DNA damage and repair upon the risk of treatment related leukemia. Leuk Lymphoma 2008; 49:204.
  87. Le Beau MM, Albain KS, Larson RA, et al. Clinical and cytogenetic correlations in 63 patients with therapy-related myelodysplastic syndromes and acute nonlymphocytic leukemia: further evidence for characteristic abnormalities of chromosomes no. 5 and 7. J Clin Oncol 1986; 4:325.
  88. Thirman MJ, Gill HJ, Burnett RC, et al. Rearrangement of the MLL gene in acute lymphoblastic and acute myeloid leukemias with 11q23 chromosomal translocations. N Engl J Med 1993; 329:909.
  89. Albain KS, Le Beau MM, Ullirsch R, Schumacher H. Implication of prior treatment with drug combinations including inhibitors of topoisomerase II in therapy-related monocytic leukemia with a 9;11 translocation. Genes Chromosomes Cancer 1990; 2:53.
  90. Pedersen-Bjergaard J, Philip P. Two different classes of therapy-related and de-novo acute myeloid leukemia? Cancer Genet Cytogenet 1991; 55:119.
  91. Super HJ, McCabe NR, Thirman MJ, et al. Rearrangements of the MLL gene in therapy-related acute myeloid leukemia in patients previously treated with agents targeting DNA-topoisomerase II. Blood 1993; 82:3705.
  92. Chakraborty S, Sun CL, Francisco L, et al. Accelerated telomere shortening precedes development of therapy-related myelodysplasia or acute myelogenous leukemia after autologous transplantation for lymphoma. J Clin Oncol 2009; 27:791.
  93. Calado RT, Regal JA, Hills M, et al. Constitutional hypomorphic telomerase mutations in patients with acute myeloid leukemia. Proc Natl Acad Sci U S A 2009; 106:1187.
  94. Allan JM, Wild CP, Rollinson S, et al. Polymorphism in glutathione S-transferase P1 is associated with susceptibility to chemotherapy-induced leukemia. Proc Natl Acad Sci U S A 2001; 98:11592.
  95. Bolufer P, Collado M, Barragan E, et al. Profile of polymorphisms of drug-metabolising enzymes and the risk of therapy-related leukaemia. Br J Haematol 2007; 136:590.
  96. Chen H, Sandler DP, Taylor JA, et al. Increased risk for myelodysplastic syndromes in individuals with glutathione transferase theta 1 (GSTT1) gene defect. Lancet 1996; 347:295.
  97. Dahabreh IJ, Giannouli S, Gota V, Voulgarelis M. GSTT1 and GSTM1 polymorphisms and myelodysplastic syndrome risk: a systematic review and meta-analysis. Int J Cancer 2010; 126:1716.
  98. Little JB. Cellular, molecular, and carcinogenic effects of radiation. Hematol Oncol Clin North Am 1993; 7:337.
  99. Ishimaru T, Otake M, Ischimaru M. Dose-response relationship of neutrons and gamma rays to leukemia incidence among atomic bomb survivors in Hiroshima and Nagasaki by type of leukemia, 1950--1971. Radiat Res 1979; 77:377.
  100. Bizzozero OJ Jr, Johnson KG, Ciocco A. Radiation-related leukemia in Hiroshima and Nagasaki, 1946-1964. I. Distribution, incidence and appearance time. N Engl J Med 1966; 274:1095.
  101. Yoshinaga S, Mabuchi K, Sigurdson AJ, et al. Cancer risks among radiologists and radiologic technologists: review of epidemiologic studies. Radiology 2004; 233:313.
  102. Shuryak I, Sachs RK, Hlatky L, et al. Radiation-induced leukemia at doses relevant to radiation therapy: modeling mechanisms and estimating risks. J Natl Cancer Inst 2006; 98:1794.
  103. Kaldor JM, Day NE, Band P, et al. Second malignancies following testicular cancer, ovarian cancer and Hodgkin's disease: an international collaborative study among cancer registries. Int J Cancer 1987; 39:571.
  104. Thirman MJ, Larson RA. Therapy-related myeloid leukemia. Hematol Oncol Clin North Am 1996; 10:293.
  105. Valagussa P, Santoro A, Fossati-Bellani F, et al. Second acute leukemia and other malignancies following treatment for Hodgkin's disease. J Clin Oncol 1986; 4:830.
  106. Blayney DW, Longo DL, Young RC, et al. Decreasing risk of leukemia with prolonged follow-up after chemotherapy and radiotherapy for Hodgkin's disease. N Engl J Med 1987; 316:710.
  107. van Leeuwen FE, Somers R, Taal BG, et al. Increased risk of lung cancer, non-Hodgkin's lymphoma, and leukemia following Hodgkin's disease. J Clin Oncol 1989; 7:1046.
  108. Andrieu JM, Ifrah N, Payen C, et al. Increased risk of secondary acute nonlymphocytic leukemia after extended-field radiation therapy combined with MOPP chemotherapy for Hodgkin's disease. J Clin Oncol 1990; 8:1148.
  109. Pearce MS, Salotti JA, Little MP, et al. Radiation exposure from CT scans in childhood and subsequent risk of leukaemia and brain tumours: a retrospective cohort study. Lancet 2012; 380:499.
  110. Brandt L, Nilsson PG, Mitelman F. Occupational exposure to petroleum products in men with acute non-lymphocytic leukaemia. Br Med J 1978; 1:553.
  111. Austin H, Delzell E, Cole P. Benzene and leukemia. A review of the literature and a risk assessment. Am J Epidemiol 1988; 127:419.
  112. Schnatter AR, Glass DC, Tang G, et al. Myelodysplastic syndrome and benzene exposure among petroleum workers: an international pooled analysis. J Natl Cancer Inst 2012; 104:1724.
  113. Rushton L, Schnatter AR, Tang G, Glass DC. Acute myeloid and chronic lymphoid leukaemias and exposure to low-level benzene among petroleum workers. Br J Cancer 2014; 110:783.
  114. Irons RD, Gross SA, Le A, et al. Integrating WHO 2001-2008 criteria for the diagnosis of Myelodysplastic Syndrome (MDS): a case-case analysis of benzene exposure. Chem Biol Interact 2010; 184:30.
  115. Carlos-Wallace FM, Zhang L, Smith MT, et al. Parental, In Utero, and Early-Life Exposure to Benzene and the Risk of Childhood Leukemia: A Meta-Analysis. Am J Epidemiol 2016; 183:1.
  116. Houot J, Marquant F, Goujon S, et al. Residential Proximity to Heavy-Traffic Roads, Benzene Exposure, and Childhood Leukemia-The GEOCAP Study, 2002-2007. Am J Epidemiol 2015; 182:685.
  117. Hauptmann M, Stewart PA, Lubin JH, et al. Mortality from lymphohematopoietic malignancies and brain cancer among embalmers exposed to formaldehyde. J Natl Cancer Inst 2009; 101:1696.
  118. Beane Freeman LE, Blair A, Lubin JH, et al. Mortality from lymphohematopoietic malignancies among workers in formaldehyde industries: the National Cancer Institute Cohort. J Natl Cancer Inst 2009; 101:751.
  119. Bachand AM, Mundt KA, Mundt DJ, Montgomery RR. Epidemiological studies of formaldehyde exposure and risk of leukemia and nasopharyngeal cancer: a meta-analysis. Crit Rev Toxicol 2010; 40:85.
  120. Coggon D, Ntani G, Harris EC, Palmer KT. Upper airway cancer, myeloid leukemia, and other cancers in a cohort of British chemical workers exposed to formaldehyde. Am J Epidemiol 2014; 179:1301.
  121. Linet MS. The Leukemias: Epidemiologic Aspects, Oxford University Press, New York 1985.
  122. Taylor JA, Sandler DP, Bloomfield CD, et al. ras oncogene activation and occupational exposures in acute myeloid leukemia. J Natl Cancer Inst 1992; 84:1626.
  123. Sandler DP, Shore DL, Anderson JR, et al. Cigarette smoking and risk of acute leukemia: associations with morphology and cytogenetic abnormalities in bone marrow. J Natl Cancer Inst 1993; 85:1994.
  124. Smith MT, Wang Y, Kane E, et al. Low NAD(P)H:quinone oxidoreductase 1 activity is associated with increased risk of acute leukemia in adults. Blood 2001; 97:1422.
  125. Larson RA, Wang Y, Banerjee M, et al. Prevalence of the inactivating 609C-->T polymorphism in the NAD(P)H:quinone oxidoreductase (NQO1) gene in patients with primary and therapy-related myeloid leukemia. Blood 1999; 94:803.
  126. Rothman N, Smith MT, Hayes RB, et al. Benzene poisoning, a risk factor for hematological malignancy, is associated with the NQO1 609C-->T mutation and rapid fractional excretion of chlorzoxazone. Cancer Res 1997; 57:2839.
  127. Lebailly P, Willett EV, Moorman AV, et al. Genetic polymorphisms in microsomal epoxide hydrolase and susceptibility to adult acute myeloid leukaemia with defined cytogenetic abnormalities. Br J Haematol 2002; 116:587.
  128. Moorman AV, Roman E, Cartwright RA, Morgan GJ. Smoking and the risk of acute myeloid leukaemia in cytogenetic subgroups. Br J Cancer 2002; 86:60.
  129. Kristinsson SY, Björkholm M, Hultcrantz M, et al. Chronic immune stimulation might act as a trigger for the development of acute myeloid leukemia or myelodysplastic syndromes. J Clin Oncol 2011; 29:2897.
  130. Ross JA, Blair CK, Cerhan JR, et al. Nonsteroidal anti-inflammatory drug and acetaminophen use and risk of adult myeloid leukemia. Cancer Epidemiol Biomarkers Prev 2011; 20:1741.
  131. Walter RB, Milano F, Brasky TM, White E. Long-term use of acetaminophen, aspirin, and other nonsteroidal anti-inflammatory drugs and risk of hematologic malignancies: results from the prospective Vitamins and Lifestyle (VITAL) study. J Clin Oncol 2011; 29:2424.
  132. Poiesz BJ, Ruscetti FW, Gazdar AF, et al. Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma. Proc Natl Acad Sci U S A 1980; 77:7415.
  133. Gallo R, Ruscetti F, Collins S, et al. Human myeloid leukemia cells: Studies on oncornaviral related information and in vitro growth and differentiation. In: Hematopoietic Cell Differentiation, Golde D, Cline M, Metcalf D, et al. (Eds), Academic Press, Orlando 1979. Vol 10, p.335.
  134. Goldin LR, Kristinsson SY, Liang XS, et al. Familial aggregation of acute myeloid leukemia and myelodysplastic syndromes. J Clin Oncol 2012; 30:179.
  135. Klco JM, Mullighan CG. Advances in germline predisposition to acute leukaemias and myeloid neoplasms. Nat Rev Cancer 2021; 21:122.
  136. University of Chicago Hematopoietic Malignancies Cancer Risk Team. How I diagnose and manage individuals at risk for inherited myeloid malignancies. Blood 2016; 128:1800.
  137. Miyauchi J, Kelleher CA, Yang YC, et al. The effects of three recombinant growth factors, IL-3, GM-CSF, and G-CSF, on the blast cells of acute myeloblastic leukemia maintained in short-term suspension culture. Blood 1987; 70:657.
  138. Delwel R, Salem M, Pellens C, et al. Growth regulation of human acute myeloid leukemia: effects of five recombinant hematopoietic factors in a serum-free culture system. Blood 1988; 72:1944.
  139. Vellenga E, Ostapovicz D, O'Rourke B, Griffin JD. Effects of recombinant IL-3, GM-CSF, and G-CSF on proliferation of leukemic clonogenic cells in short-term and long-term cultures. Leukemia 1987; 1:584.
  140. Ikeda H, Kanakura Y, Tamaki T, et al. Expression and functional role of the proto-oncogene c-kit in acute myeloblastic leukemia cells. Blood 1991; 78:2962.
  141. Piacibello W, Fubini L, Sanavio F, et al. Effects of human FLT3 ligand on myeloid leukemia cell growth: heterogeneity in response and synergy with other hematopoietic growth factors. Blood 1995; 86:4105.
  142. Meshinchi S, Appelbaum FR. Structural and functional alterations of FLT3 in acute myeloid leukemia. Clin Cancer Res 2009; 15:4263.
  143. Dong F, Brynes RK, Tidow N, et al. Mutations in the gene for the granulocyte colony-stimulating-factor receptor in patients with acute myeloid leukemia preceded by severe congenital neutropenia. N Engl J Med 1995; 333:487.
  144. Dong F, Dale DC, Bonilla MA, et al. Mutations in the granulocyte colony-stimulating factor receptor gene in patients with severe congenital neutropenia. Leukemia 1997; 11:120.
  145. Wölfler A, Erkeland SJ, Bodner C, et al. A functional single-nucleotide polymorphism of the G-CSF receptor gene predisposes individuals to high-risk myelodysplastic syndrome. Blood 2005; 105:3731.
  146. Hunter MG, Avalos BR. Granulocyte colony-stimulating factor receptor mutations in severe congenital neutropenia transforming to acute myelogenous leukemia confer resistance to apoptosis and enhance cell survival. Blood 2000; 95:2132.
  147. Young DC, Griffin JD. Autocrine secretion of GM-CSF in acute myeloblastic leukemia. Blood 1986; 68:1178.
  148. Cozzolino F, Rubartelli A, Aldinucci D, et al. Interleukin 1 as an autocrine growth factor for acute myeloid leukemia cells. Proc Natl Acad Sci U S A 1989; 86:2369.
  149. Löwenberg B, van Putten WL, Touw IP, et al. Autonomous proliferation of leukemic cells in vitro as a determinant of prognosis in adult acute myeloid leukemia. N Engl J Med 1993; 328:614.
  150. Hunter AE, Rogers SY, Roberts IA, et al. Autonomous growth of blast cells is associated with reduced survival in acute myeloblastic leukemia. Blood 1993; 82:899.
  151. Wetzler M, Baer MR, Bernstein SH, et al. Expression of c-mpl mRNA, the receptor for thrombopoietin, in acute myeloid leukemia blasts identifies a group of patients with poor response to intensive chemotherapy. J Clin Oncol 1997; 15:2262.
  152. Koistinen P, Wang C, Curtis JE, McCulloch EA. Granulocyte-macrophage colony-stimulating factor and interleukin-3 protect leukemic blast cells from ara-C toxicity. Leukemia 1991; 5:789.
  153. Lotem J, Sachs L. Hematopoietic cytokines inhibit apoptosis induced by transforming growth factor beta 1 and cancer chemotherapy compounds in myeloid leukemic cells. Blood 1992; 80:1750.
  154. Lisovsky M, Estrov Z, Zhang X, et al. Flt3 ligand stimulates proliferation and inhibits apoptosis of acute myeloid leukemia cells: regulation of Bcl-2 and Bax. Blood 1996; 88:3987.
  155. Geller RB. Use of cytokines in the treatment of acute myelocytic leukemia: a critical review. J Clin Oncol 1996; 14:1371.
  156. Hershman D, Neugut AI, Jacobson JS, et al. Acute myeloid leukemia or myelodysplastic syndrome following use of granulocyte colony-stimulating factors during breast cancer adjuvant chemotherapy. J Natl Cancer Inst 2007; 99:196.
  157. Relling MV, Boyett JM, Blanco JG, et al. Granulocyte colony-stimulating factor and the risk of secondary myeloid malignancy after etoposide treatment. Blood 2003; 101:3862.
  158. Lyman GH, Dale DC, Wolff DA, et al. Acute myeloid leukemia or myelodysplastic syndrome in randomized controlled clinical trials of cancer chemotherapy with granulocyte colony-stimulating factor: a systematic review. J Clin Oncol 2010; 28:2914.
Topic 4493 Version 43.0

References

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