Fact-checked by Grok 2 weeks ago

Acute myeloid leukemia

Acute myeloid leukemia (AML) is a rapidly progressing form of cancer that originates in the and affects the , characterized by the uncontrolled proliferation of immature known as myeloblasts, which crowd out healthy cells and impair their production. This clonal expansion of abnormal myeloid cells leads to bone marrow failure, disrupting the formation of cells, functional , and platelets. AML is the most common type of in adults, typically diagnosed in individuals over age 65, though it can occur at any age. The disease manifests through symptoms stemming from the deficiency of normal blood components, including persistent fatigue and weakness due to , frequent infections from low functional , easy bruising or from reduced platelets, fever, bone or joint pain, , and paleness. In advanced cases, complications such as or organ infiltration may occur, exacerbating the clinical presentation. Without prompt intervention, AML advances quickly, often within weeks or months, leading to life-threatening conditions. The underlying cause of AML involves acquired genetic mutations in the DNA of myeloid stem cells, resulting in their abnormal differentiation and proliferation, though the precise triggers remain unclear in most cases. Key risk factors include advanced age, with a median diagnosis age of 68 years; exposure to ionizing radiation or chemicals like benzene; smoking; prior treatment with chemotherapy or radiation for other cancers; and certain genetic conditions such as Down syndrome or myelodysplastic syndromes. AML is classified using systems such as the WHO and International Consensus Classification (ICC), which incorporate genetic, immunophenotypic, and morphologic features to define subtypes. Epidemiologically, AML has an annual incidence of approximately 4.3 cases per 100,000 people in the United States, affecting approximately 22,000 individuals yearly as of 2025, with a higher prevalence among non-Hispanic White males. Diagnosis typically begins with a revealing cytopenias and circulating blasts, followed by bone marrow aspiration and to confirm ≥20% myeloblasts (or defining genetic abnormalities in some cases), along with cytogenetic, immunophenotypic, and molecular testing to classify subtypes and guide . AML is categorized into groups—favorable, intermediate, or adverse—based on genetic abnormalities, influencing treatment decisions. Treatment for AML generally involves intensive multi-phase , starting with (often the "7+3" regimen of cytarabine and an ) to achieve remission by reducing blasts to less than 5%, followed by consolidation with high-dose or allogeneic , which can offer curative potential, particularly in higher-risk cases. Targeted therapies, such as FLT3 inhibitors or IDH1/2 inhibitors, are increasingly used for specific mutations, while supportive care addresses complications like infections and . Overall survival varies by risk group and age, with five-year rates ranging from 10-70% depending on factors like .

Introduction

Definition and overview

Acute myeloid leukemia (AML) is a hematologic characterized by the rapid proliferation of abnormal myeloid precursor cells, known as myeloblasts, in the and peripheral blood, which disrupts normal hematopoiesis and leads to the accumulation of immature cells. These abnormal cells fail to mature into functional blood components, resulting in deficiencies of blood cells, platelets, and mature . According to the (WHO) classification, AML is typically diagnosed when there is an accumulation of 20% or more blasts in the or peripheral blood, though certain genetically defined subtypes, such as those with recurrent genetic abnormalities, may be diagnosed with lower blast percentages. Subtypes are further delineated based on the degree of cellular maturation and specific features, including (APL), a distinct variant involving promyelocytes with the t(15;17) translocation. AML originates from the myeloid lineage, distinguishing it from (ALL), which arises from lymphoid precursors and primarily affects lymphocytes rather than myeloid cells like , monocytes, erythrocytes, or megakaryocytes. In contrast to chronic myeloid leukemia (CML), an indolent driven by the BCR-ABL1 fusion and characterized by a chronic phase with mature overproduction, AML presents with an acute onset and blast crisis without the preceding chronic features typical of CML. This rapid progression in AML often manifests with symptoms such as fatigue, easy bruising, and recurrent infections due to impaired production. As one of the principal acute leukemias, AML falls within the broader category of blood cancers and accounts for the majority of cases in adults. It predominantly affects older individuals, with a age at of 69 years, though it can occur across all age groups, including rare pediatric cases. In the United States, approximately 22,000 new cases are diagnosed annually, underscoring its significance in .

Classification systems

The French-American-British (FAB) classification system, introduced in 1976, was the first widely adopted framework for categorizing acute myeloid leukemia (AML) based primarily on morphological and cytochemical features of leukemic blasts observed in or peripheral smears. It divided AML into eight subtypes (M0 through M7), with M0 representing minimally differentiated AML lacking clear myeloid markers, M1 and M2 as myeloblastic variants with varying maturation, M3 as (APL) characterized by promyelocytes and the t(15;17) translocation, M4 as myelomonocytic, M5 as monocytic, M6 as erythroid, and M7 as megakaryoblastic. This system emphasized blast percentage thresholds (≥30% for most subtypes) and cytochemical stains like to distinguish myeloid lineage, providing a standardized approach for diagnosis and initial prognostic assessment despite its limitations in incorporating genetic data. The (WHO) 5th edition classification, published in 2022, marked a significant by integrating genetic, immunophenotypic, and clinical features alongside , aiming to better reflect and guide therapy. It eliminated the 20% blast threshold for AML with defining genetic abnormalities (except for BCR::ABL1-positive cases and certain mutations requiring ≥20% blasts), introducing subtypes such as AML with mutated , AML with in-frame biallelic or bZIP mutations, and core-binding factor (CBF) AML (e.g., t(8;21)/RUNX1::RUNX1T1 or inv(16)/CBFB::MYH11). Additional categories include AML with myelodysplasia-related changes (AML-MR), therapy-related myeloid neoplasms, and those with mutated TP53, emphasizing molecular drivers like FLT3-ITD (noted briefly for its role in ). Updates integrated into clinical practice by 2025 further refined these through harmonized guidelines, incorporating emerging data on variant allele frequencies and for precise subtyping. In parallel, the International Consensus Classification (ICC) of 2022 offered an alternative framework, prioritizing prognostic relevance and requiring ≥10% blasts for most genetically defined AML (with exceptions like ≥20% for BCR::ABL1), while distinguishing MDS/AML for cases with 10-19% blasts. It defines similar genetic subtypes to WHO, such as NPM1-mutated AML and CBF AML, but restricts CEBPA mutations to in-frame bZIP types and separately categorizes AML with mutated TP53 (requiring multi-hit mutations with >10% variant allele fraction and ≥20% blasts) and therapy-related AML without myelodysplasia history. Key differences from WHO include stricter blast criteria for some entities and the introduction of MDS/AML as a bridge category, reflecting debates on low-blast myeloid neoplasms. The European LeukemiaNet (ELN) 2025 recommendations harmonize elements of WHO and for risk stratification, classifying AML into favorable, intermediate, and adverse groups based on and to inform decisions. Favorable risk includes CBF AML and -mutated cases without FLT3-ITD; intermediate encompasses wild-type with FLT3-ITD or certain abnormalities like t(9;11); and adverse features complex karyotypes, TP53 mutations, or mutations in ASXL1/RUNX1. These systems collectively impact clinical practice by determining eligibility for targeted therapies, such as all-trans (ATRA) plus for PML-RARA-positive (formerly FAB M3), enabling personalized approaches beyond alone.

Clinical presentation

Symptoms

Acute myeloid leukemia (AML) primarily manifests through symptoms resulting from the replacement of normal bone marrow cells by leukemic blasts, leading to deficiencies in red blood cells, white blood cells, and platelets. Patients commonly report fatigue and weakness due to anemia caused by reduced red blood cell production, which impairs oxygen delivery to tissues. Shortness of breath and chest tightness may also occur as a consequence of this anemia. Frequent infections, often presenting as fever, sore throat, or pneumonia, arise from neutropenia, where dysfunctional white blood cells fail to combat pathogens effectively. Thrombocytopenia contributes to bleeding tendencies, including easy bruising, bleeding gums, and prolonged bleeding from minor injuries, as low platelet counts impair normal clotting. or pain results from the expansion of leukemic cells within the , causing pressure on surrounding tissues. Constitutional symptoms such as unintentional , , and loss of appetite can accompany these hematologic effects, reflecting the systemic burden of the disease. In specific subtypes like (APL), a variant of AML, patients may experience exacerbated symptoms due to (DIC), including severe such as , petechiae, oral mucosal hemorrhages, and bleeding from intravenous sites. These symptoms develop rapidly, often over days to weeks, underscoring the aggressive nature of AML.

Signs

Patients with acute myeloid leukemia (AML) often present with pallor on , resulting from caused by bone marrow infiltration and ineffective . leads to observable bleeding manifestations, including petechiae, , and ecchymoses, which appear as small red or purple spots or larger bruises on the skin due to impaired platelet production. In some cases, or may be detected on abdominal , attributed to or leukemic infiltration of these organs. is uncommon in AML, occurring less frequently than in lymphoid leukemias, though enlargement of lymph nodes may be noted if there is extramedullary involvement. In monocytic subtypes such as FAB M4 (acute myelomonocytic leukemia) and M5 (), gum hypertrophy due to leukemic infiltration of the gingival tissue is a characteristic finding, often presenting as diffuse swelling that can lead to oral discomfort. Skin infiltration, known as leukemia cutis, may also occur in these subtypes, manifesting as nodules, plaques, or erythematous lesions from myeloid blast accumulation in the . Neutropenia predisposes patients to infections, with clinical signs including oral ulcers from mucosal breakdown and respiratory distress due to pulmonary infections such as .

Risk factors

Genetic predispositions

Inherited genetic predispositions to acute myeloid leukemia (AML) involve that confer susceptibility to the disease, distinct from alterations acquired during leukemogenesis. These predispositions account for approximately 5-10% of AML cases overall, with affected individuals often presenting at younger ages compared to sporadic AML, though the exact varies by and testing methodology. Such disrupt critical cellular processes like , transcription regulation, or maintenance, leading to clonal hematopoietic evolution and increased AML risk. Familial syndromes represent key examples of germline predispositions. In Li-Fraumeni syndrome, caused by germline TP53 mutations, individuals face an elevated lifetime cancer risk of 70-90%, with hematologic malignancies, including AML, comprising 4-10% of cases, often presenting in childhood or early adulthood. , resulting from trisomy 21, dramatically increases AML susceptibility, with children under 5 years old exhibiting a 150-fold higher incidence compared to the general population, frequently manifesting as megakaryoblastic AML. Bone marrow failure disorders also predispose to AML through inherited defects in or telomere biology. Fanconi anemia, characterized by germline mutations in FANC genes, confers a more than 500-fold increased risk of AML, with cumulative incidence reaching 10-37% by age 50, driven by chromosomal instability and in hematopoietic stem cells. Dyskeratosis congenita, involving mutations in telomere maintenance genes such as DKC1 or TERT, heightens AML risk alongside failure, with myeloid neoplasms emerging as a significant complication in up to 20-30% of cases, often in adolescence or early adulthood. Rare germline mutations in transcription factors like and RUNX1 define specific familial AML predisposition syndromes. Biallelic CEBPA mutations exhibit near-complete (>90% lifetime risk), typically leading to AML onset in the second or third decade of life, with favorable upon standard . Germline RUNX1 variants cause familial platelet disorder with associated myeloid , carrying a 35-44% lifetime risk of AML or , often presenting in adulthood with preceding . Given the implications for family members and treatment decisions, screening for predispositions is recommended for all AML patients, particularly those with young onset, family history of hematologic malignancies, or features suggestive of syndromes like bone marrow failure. and testing using non-hematopoietic tissues (e.g., skin fibroblasts) facilitate early identification and cascade screening in relatives.

Environmental exposures

Environmental exposures to certain chemicals, radiation, and prior chemotherapeutic agents have been established as modifiable risk factors for acute myeloid leukemia (AML), primarily through mechanisms involving DNA damage and disruption of hematopoiesis. These agents can induce chromosomal abnormalities and mutations in hematopoietic stem cells, leading to clonal expansion and leukemogenesis. Among chemical exposures, benzene stands out as a well-documented carcinogen associated with AML, particularly in occupational settings such as the petroleum, rubber, and chemical manufacturing industries where it is used as a solvent. Long-term exposure to benzene increases AML risk in a dose-dependent manner, with cohort and case-control studies reporting odds ratios ranging from 2 to 5 for high-exposure groups compared to unexposed individuals. Tobacco smoke, which contains benzene and other aromatic hydrocarbons, also elevates AML risk, with current smokers facing a 20-40% increased odds compared to never-smokers, based on meta-analyses of epidemiological data. Ionizing radiation exposure, such as from therapeutic treatments or high-dose environmental incidents, is another established for AML. Studies of atomic bomb survivors in and demonstrated a dose-dependent excess of , with the elevated incidence appearing as an early delayed effect of , peaking 5-10 years post-exposure before declining. This latency pattern and risk profile have been corroborated in cohorts exposed to medical , underscoring the role of in inducing leukemogenic mutations. Prior chemotherapy, particularly with alkylating agents like used in treatments for solid tumors or lymphomas, can lead to therapy-related AML (t-AML), accounting for 1-5% of cases in exposed patient populations. t-AML following alkylating agents typically manifests 4-7 years after exposure, often preceded by a myelodysplastic phase, and is characterized by adverse cytogenetic features such as deletions in chromosomes 5 or 7. Other environmental agents, including certain pesticides and organic solvents, have been linked to AML in occupational studies, with evidence suggesting elevated risks for agricultural workers and those in or . For instance, meta-analyses of data indicate odds ratios of 1.5-2.0 for , though associations vary by specific compound and exposure duration. Overall, these exposures exhibit dose-response relationships, where higher cumulative doses correlate with greater AML risk, and latency periods generally range from 2-10 years depending on the agent, allowing time for accumulation of genetic damage.

Prior medical conditions

Secondary acute myeloid leukemia (AML) arises in the context of prior medical conditions and accounts for 10-20% of all AML cases, often carrying an adverse prognosis with a 5-year overall survival rate below 30%. Among hematologic disorders, myelodysplastic syndromes (MDS) represent a major precursor, with approximately 20-30% of patients progressing to AML, typically within 5-10 years of diagnosis. This transformation is more common in higher-risk MDS subtypes, where ineffective hematopoiesis evolves into overt leukemia. Myeloproliferative neoplasms (MPN), such as polycythemia vera and essential thrombocythemia, also predispose to AML, often involving JAK2 mutations that drive clonal expansion and eventual leukemic transformation; the 10-year risk is estimated at 2.3-8.7% for polycythemia vera. Aplastic anemia and paroxysmal nocturnal hemoglobinuria (PNH), particularly following immunosuppressive or supportive treatments, confer a smaller but notable risk of AML development, with malignant progression observed in up to 13% of cases over time. These bone marrow failure syndromes can lead to secondary AML through clonal evolution in surviving hematopoietic cells. Non-hematologic conditions, including prior solid tumors treated with or , are associated with therapy-related AML, which comprises 5-10% of secondary cases and arises due to genotoxic damage to hematopoietic cells. Common examples include or ovarian cancers exposed to alkylating agents or inhibitors, highlighting the long-term risks of cytotoxic therapies.

Pathophysiology

Hematopoietic stem cell origin

Hematopoiesis is the process by which (HSCs), residing primarily in the , give rise to all lineages through a tightly regulated . HSCs are multipotent, quiescent cells capable of self-renewal and ; they first generate multipotent progenitors (MPPs), which then branch into common myeloid progenitors (CMPs) and common lymphoid progenitors (CLPs). CMPs further differentiate into granulocyte-macrophage progenitors (GMPs) and megakaryocyte-erythroid progenitors (MEPs), ultimately producing mature myeloid cells such as granulocytes, monocytes, erythrocytes, and megakaryocytes. This ensures balanced production of s to maintain . Acute myeloid leukemia (AML) originates from genetic alterations in HSCs or early myeloid s, such as MPPs or CMPs, disrupting normal hematopoiesis at its foundational level. These mutations confer a proliferative advantage and block , preventing progenitors from maturing into functional myeloid cells and instead leading to the accumulation of immature blasts. Unlike normal HSCs, which balance self-renewal with , leukemic stem cells (LSCs)—the transformed counterparts—exhibit enhanced self-renewal and resistance to , enabling their persistence and propagation. Experimental evidence from mouse models, such as those transducing MLL-AF9 into HSCs versus progenitors, demonstrates that HSC-derived AML often results in more invasive disease with prolonged latency, while progenitor origins yield rapid leukemogenesis, highlighting the critical role of cellular in disease initiation. The clonal expansion of LSCs drives AML progression, as these cells asymmetrically divide to produce both LSCs and differentiating blasts, outcompeting normal hematopoiesis and leading to failure. LSCs typically reside in the +- fraction of leukemic cells, though heterogeneity exists across AML subtypes, with some LSCs appearing in - populations in NPM1-mutated cases. By hijacking the niche—the supportive microenvironment provided by stromal cells, endothelial cells, and —LSCs alter niche signaling, such as through upregulated CXCL12-CXCR4 interactions, to promote their quiescence and survival while suppressing normal HSC function. This niche remodeling fosters immune evasion and therapy resistance, contributing to . Seminal studies in immunodeficient mice first identified LSCs' self-renewal capacity, confirming their HSC-like properties. In distinction from lymphoid leukemias, which arise from lymphoid-committed progenitors like CLPs, AML is confined to the myeloid lineage, with LSCs emerging after the myeloid-lymphoid divergence point in the hematopoietic . This myeloid-specific commitment underlies AML's characteristic blast morphology and immunophenotype, such as expression of and CD13, and precludes involvement of lymphoid pathways.

Genetic mutations and leukemogenesis

Acute myeloid leukemia (AML) arises from the accumulation of genetic alterations in hematopoietic cells (HSCs), leading to uncontrolled and blocked of myeloid progenitors. A foundational in AML leukemogenesis is the two-hit model, which posits that leukemic transformation typically requires cooperative mutations: I mutations that confer proliferative and survival advantages, and II mutations that impair hematopoietic . Class I mutations, such as internal tandem duplications in FLT3 (FLT3-ITD, occurring in 20-35% of cases), activate downstream signaling pathways like /MAPK and PI3K/AKT, promoting cell growth and survival. Similarly, activating mutations in enhance in subsets of AML with core-binding factor abnormalities. Class II mutations, exemplified by the t(8;21) translocation resulting in the RUNX1::RUNX1T1 (5-10% frequency), disrupt transcription factors essential for normal myeloid . Another key class II example is the inv(16) abnormality producing the CBFB::MYH11 (5-8% frequency), which interferes with core-binding factor activity and blocks granulocytic maturation. Among the most prevalent somatic mutations in AML are those in (nucleophosmin 1), found in 25-35% of adult cases, primarily involving exon 12 insertions that cause aberrant cytoplasmic relocation of the NPM1 protein, disrupting nucleolar functions and stabilizing key oncoproteins. Mutations in (DNA methyltransferase 3A), occurring in approximately 20-25% of cases, predominantly at the R882 hotspot, lead to reduced DNA methylation activity, resulting in global hypomethylation and altered patterns that favor leukemic transformation. IDH1 and IDH2 mutations, present in 10-20% of AML patients, encode mutant isocitrate dehydrogenases that produce the oncometabolite 2-hydroxyglutarate (2-HG), which inhibits TET2-mediated and TET enzymes, thereby promoting epigenetic dysregulation and blocking . These mutations often cooperate; for instance, alterations frequently co-occur with FLT3-ITD, driving enhanced clonal expansion. Chromosomal abnormalities further contribute to AML , with balanced translocations like t(8;21) and inv() generating proteins that dominantly repress normal transcription, as seen in core-binding factor AML subtypes. Aneuploidies, such as (+8, in 5-10% of cases), promote genomic instability and may amplify oncogenes or disrupt tumor suppressors, facilitating additional mutational hits. In the (APL) subtype of AML, the t(15;17) translocation creates the PML::RARA in nearly 95% of cases, which heterodimerizes with retinoid receptors to block myeloid at the promyelocytic stage through aberrant transcriptional repression. Leukemogenesis in AML unfolds through sequential steps beginning with initiating mutations in long-term HSCs, which confer self-renewal advantages and establish pre-leukemic clones. These founding events, such as DNMT3A or NPM1 mutations, persist across disease evolution and provide a platform for secondary hits like FLT3-ITD, leading to clonal expansion and overt leukemia. Clonal evolution then drives intratumor heterogeneity, with branching patterns of subclones acquiring further alterations that enhance fitness, such as additional epigenetic or signaling mutations, ultimately contributing to disease progression and potential resistance mechanisms via heterogeneous subpopulations. Inherited predispositions can amplify the risk of acquiring these somatic changes, though the core leukemogenic process remains driven by acquired mutations.

Diagnosis

Clinical and laboratory evaluation

The clinical evaluation of acute myeloid leukemia (AML) begins with a thorough , focusing on the rapid onset of symptoms such as , recurrent infections, easy bruising, or , which typically develop over days to weeks. Patients may report a of prior , , or exposure to environmental toxins, which are known risk factors prompting suspicion for secondary AML. The also assesses for associated conditions like disseminated intravascular coagulation (DIC), evidenced by excessive or purpura. Physical examination reveals signs of cytopenias, including pallor from , petechiae or ecchymoses from , and occasional due to . Bleeding manifestations, such as oral mucosal hemorrhages or gingival bleeding, may be prominent, particularly in cases with . is uncommon in AML compared to lymphoid leukemias, though can occur in up to 20-30% of cases. Laboratory evaluation starts with a (CBC) with , which typically shows (hemoglobin <10 g/dL in most patients), thrombocytopenia (platelet count <100,000/μL), and variable white blood cell counts ranging from leukopenia (<4,000/μL) to marked leukocytosis (>100,000/μL), often with circulating blasts comprising 20% or more of leukocytes. A peripheral is essential for identifying morphologic abnormalities, such as large immature blasts with high nuclear-to-cytoplasmic ratios and —crystalline cytoplasmic inclusions for myeloid lineage—in up to 20-30% of AML cases. Schistocytes may appear in the setting of . Initial biochemical tests include serum (LDH), which is elevated in over 80% of patients due to high cell turnover, and uric acid levels, which are often raised, signaling risk for even before therapy initiation. Coagulation studies, such as and fibrinogen, are performed to detect , while assesses for renal or imbalances secondary to cytopenias. Given the severity of cytopenias and risk of life-threatening complications like or hemorrhage, patients with suspected AML require immediate hospitalization for supportive care, including transfusions and prophylaxis.

Bone marrow examination

Bone marrow examination is a critical diagnostic for confirming acute myeloid leukemia (AML) and identifying its subtype, typically performed after initial blood tests suggest abnormalities such as elevated counts or circulating blasts. The involves aspiration and , usually obtained from the posterior under , where a needle is inserted to extract liquid marrow for aspiration and a small core of bone and marrow for . Morphological analysis of the aspirate and biopsy samples examines the cellular composition, with AML diagnosed when blasts comprise at least 20% of nucleated cells in the or peripheral ; however, this threshold does not apply to cases with certain recurrent genetic abnormalities (e.g., t(8;21)/RUNX1::RUNX1T1, inv(16)/CBFB::MYH11, t(15;17)/PML::RARA, mutations, or biallelic mutations), which are classified as AML regardless of blast count, per WHO 5th edition and ICC guidelines. Blasts in AML often appear as large cells with high nucleus-to- ratios, prominent nucleoli, and scant , aiding in distinguishing AML from other myeloid disorders. Flow cytometry on the marrow sample provides immunophenotyping to characterize blasts, which typically express myeloid-associated antigens such as CD13 and , often alongside , CD117, and , helping to confirm the myeloid lineage and exclude lymphoid leukemias. This multiparametric analysis allows for rapid identification of aberrant antigen expression patterns unique to AML subtypes. Cytogenetic evaluation through karyotyping of marrow cells detects chromosomal abnormalities in approximately 50% of AML cases, including recurrent translocations and aneuploidies; for instance, a complex karyotype—defined as three or more unrelated abnormalities—occurs in 10-15% of patients and is identified via standard G-banding techniques. Molecular studies, including polymerase chain reaction (PCR) or next-generation sequencing on marrow DNA or RNA, detect gene fusions and mutations essential for subtype classification, such as the RUNX1-RUNX1T1 fusion associated with t(8;21), which disrupts normal hematopoiesis. Although generally safe, the procedure carries rare risks including bleeding, which may require intervention in patients with , and infection at the puncture site, occurring in less than 1% of cases.

Classification and risk stratification

The and risk stratification of acute myeloid leukemia (AML) primarily relies on the European LeukemiaNet (ELN) 2022 guidelines, which categorize patients into favorable, intermediate, and adverse risk groups based on cytogenetic and molecular abnormalities identified at . Favorable-risk features include core-binding factor leukemias such as t(8;21)(q22;q22.1)/RUNX1::RUNX1T1 and inv(16)(p13.1q22)/CBFB::MYH11, as well as mutated without FLT3-ITD or with low allelic ratio FLT3-ITD, and biallelic mutated . Intermediate-risk encompasses mutated with high allelic ratio FLT3-ITD, wild-type with FLT3-ITD (low allelic ratio and no adverse features), t(9;11)(p21.3;q23.3)/MLLT3::KMT2A, and abnormalities not classified as favorable or adverse. Adverse-risk is defined by karyotype (≥3 abnormalities), monosomal karyotype, TP53 mutations, inv(3)(q21.3q26.2)/,MECOM or other MECOM rearrangements, and mutations in ASXL1, BCOR, , RUNX1, SF3B1, SRSF2, STAG2, U2AF1, or ZRSR2. For patients receiving less-intensive therapies, the ELN 2024 recommendations refine stratification, with favorable risk including mutated or IDH2 without FLT3-ITD, NRAS, , or TP53 mutations, and mutated DDX41; intermediate risk covers cases with FLT3-ITD, NRAS, or mutations but wild-type TP53; and adverse risk is primarily mutated TP53. These genetic classifications integrate with the (WHO) 5th edition and International Consensus Classification (ICC) frameworks, which emphasize myeloid entities based on genetic drivers rather than blast counts alone. The ICC introduces a MDS/AML category for cases with 10–19% blasts and defining genetic abnormalities, bridging myelodysplastic syndromes and AML. Measurable residual disease (MRD) assessment, performed post-induction via multiparameter (sensitivity ≥0.1%) or quantitative (e.g., for or CBF targets), further refines risk by identifying persistent disease that may upgrade favorable cases to intermediate. Prognostic scoring systems incorporate clinical variables alongside , such as the European Scoring System for patients ≥70 years, which weights age, secondary AML, count ≥100 × 10^9/L, and to predict long-term survival. The 2025 National Comprehensive Cancer Network (NCCN) guidelines and ELN 2022 recommendations (with 2024 refinements for less-intensive therapies) stress comprehensive next-generation sequencing (NGS) panels at to detect actionable mutations (e.g., FLT3, IDH1/2) and support precise risk assignment, with panels covering at least 50-100 genes for broad coverage. In clinical trials, these stratification tools guide patient allocation to novel agent arms, ensuring balanced evaluation of therapies like or menin inhibitors across risk groups.

Treatment

Initial induction therapy

The initial induction therapy for acute myeloid leukemia (AML) aims to rapidly reduce leukemic blasts and achieve complete remission in newly diagnosed patients who are medically fit for intensive treatment. For such patients, the standard regimen is the "7+3" protocol, which combines continuous intravenous cytarabine at a dose of 100-200 mg/m² on days 1 through 7 with at 60-90 mg/m² on days 1 through 3. This approach has been established as the backbone of induction for fit adults based on clinical trials demonstrating its efficacy in blast clearance. In cases of high-risk AML, alternative intensive regimens may be employed to improve response rates. One such option is FLAG-IDA, consisting of , high-dose cytarabine, (G-CSF), and idarubicin, which has shown superior outcomes compared to standard 7+3 in select high-risk populations. The selection of these regimens is guided by risk stratification to tailor intensity to disease biology and patient fitness. For (APL), a distinct subtype of AML characterized by t(15;17) translocation, induction therapy differs markedly and focuses on differentiation induction rather than cytotoxicity alone. The preferred regimen combines all-trans retinoic acid (ATRA) with arsenic trioxide (ATO), yielding complete remission rates greater than 90% while minimizing chemotherapy-related toxicity. Achievement of complete remission (CR) following induction is a key endpoint, defined by the International Working Group criteria as blasts less than 5%, greater than 1,000/μL, platelet count greater than 100,000/μL, independence from transfusions, and absence of extramedullary disease. Response is typically assessed via evaluation around day 14-21, with persistent blasts prompting regimen adjustment. Induction therapy carries risks of acute complications, notably due to rapid cell destruction, which manifests as , , and renal impairment. Prophylaxis involves aggressive intravenous to maintain output above 100 mL/hour and administration of to inhibit and reduce production. In high-burden disease, may be added for established .

Consolidation and maintenance

Consolidation therapy in acute myeloid leukemia (AML) follows the achievement of complete remission () after initial induction therapy and is designed to eliminate (MRD) and reduce the risk of relapse. This phase typically lasts 3 to 6 months post-induction, involving multiple cycles of tailored to the patient's risk group based on cytogenetic and molecular features. For favorable-risk patients, such as those with core-binding factor abnormalities (e.g., t(8;21) or inv(16)), high-dose cytarabine (HiDAC) is the standard regimen, administered as 3 g/m² intravenously every 12 hours on days 1, 3, and 5 for 3 to 4 cycles. This approach has been established through risk-stratified protocols that prioritize intensive cytarabine to consolidate remission in lower-risk cases. In intermediate-risk AML, consolidation often mirrors HiDAC regimens for favorable risk but may incorporate therapy to further prolong remission, particularly for patients not proceeding to transplantation. Oral , a hypomethylating agent, is recommended as following consolidation, given as 300 mg orally for 14 days every 28-day cycle for up to 24 months or until disease progression. This strategy, supported by the phase 3 QUAZAR AML-001 trial, demonstrated improved overall survival (median 24.7 months) compared to in patients in first after intensive chemotherapy. MRD assessment, often using next-generation sequencing (NGS), guides escalation during consolidation; persistent MRD positivity may prompt intensified cycles or alternative approaches within guidelines. For patients deemed unfit for intensive due to age or comorbidities, low-intensity consolidation regimens are preferred to balance efficacy and tolerability. Hypomethylating agents (HMAs) such as , combined with , provide an option, with dosed at 75 mg/m² subcutaneously or intravenously for 7 days every 4 to 5 weeks. These regimens aim to maintain while minimizing toxicity, with durations adjusted based on response and MRD status, typically extending several months. Overall, and maintenance strategies are individualized to optimize long-term disease control without overlapping into transplant or experimental modalities.

Hematopoietic stem cell transplantation

Allogeneic (HSCT) serves as a potentially curative for acute myeloid leukemia (AML) by replacing the patient's diseased with healthy donor cells, leveraging the graft-versus-leukemia (GVL) effect to eradicate residual leukemic cells. This immunologic mechanism, primarily mediated by donor T cells recognizing tumor-specific antigens on AML blasts, contributes significantly to control and is a key distinction from non-transplant therapies. Indications for allogeneic HSCT are primarily guided by risk stratification, with the procedure recommended in first complete remission (CR1) for patients with adverse-risk AML, such as those with complex karyotypes or TP53 mutations, and for the majority of intermediate-risk cases as defined by the 2025 European LeukemiaNet (ELN) guidelines. It is also indicated for relapsed AML or in second complete remission (CR2), particularly for fit patients who achieve remission after salvage . For older patients (typically ≥60 years) with high- or intermediate-risk , reduced-intensity conditioning () regimens enable HSCT while minimizing toxicity, expanding eligibility beyond younger individuals. Donor selection prioritizes (HLA)-matched siblings as the preferred source due to lower risks of complications, followed by HLA-matched unrelated donors from registries. Advances in post-transplant (PTCy) prophylaxis have broadened access through haploidentical donors (half-matched family members), as emphasized in the 2025 ELN and Society for Blood and Marrow Transplantation (EBMT) recommendations, allowing transplantation for nearly all eligible patients regardless of donor availability. The procedure involves high-dose to ablate the recipient's marrow, followed by infusion of donor hematopoietic stem cells from , peripheral blood, or blood. Myeloablative (MAC), such as the busulfan- regimen, is standard for younger, fit patients to maximize leukemia eradication, while RIC (e.g., lower-dose busulfan with ) is used for older or comorbid patients to reduce non-relapse mortality, though it may slightly increase relapse risk offset by the GVL effect. Major complications include acute and chronic (GVHD), affecting 30-50% of recipients and managed with immunosuppressive agents like cyclosporine and , though severe cases can be life-threatening. Infections, particularly during the early post-transplant period of , occur in up to 40% of patients due to impaired immunity, necessitating prophylactic antimicrobials. remains a significant issue, with rates of 20-40% depending on disease risk and MRD status pre-transplant, often mitigated by donor lymphocyte infusions to enhance GVL. In eligible patients, long-term survival reaches 40-60% at 3-5 years, with better outcomes in those achieving MRD negativity before HSCT and receiving MAC in CR1.

Targeted and novel therapies

Targeted therapies in acute myeloid leukemia (AML) represent a shift toward precision medicine by addressing specific molecular aberrations that drive leukemogenesis, such as in FLT3, IDH1/IDH2, and , allowing for more tailored in addition to standard backbone. These agents, including inhibitors and small-molecule disruptors, have demonstrated improved outcomes in subsets of patients with actionable , particularly those unfit for intensive . As of 2025, integration of these therapies into frontline regimens has become standard for mutation-positive AML, guided by comprehensive genomic profiling at diagnosis. FLT3 inhibitors target mutations in the FMS-like 3 gene, present in approximately 30% of AML cases, which confer a poor . Midostaurin, a multi-kinase inhibitor, added to standard "7+3" induction (cytarabine plus ) significantly improved overall survival (OS) in newly diagnosed FLT3-mutated AML patients aged 18-59 in the phase 3 RATIFY trial, with a of 0.78 for OS and median OS not reached versus 25.6 months with . Long-term follow-up at 10 years confirmed a durable event-free survival benefit, though the OS advantage attenuated over time due to subsequent therapies and patient aging. , a selective FLT3 inhibitor, is approved for relapsed or FLT3-mutated AML following the ADMIRAL trial, where it extended median OS to 9.3 months compared to 5.6 months with salvage . In frontline settings, is increasingly combined with or hypomethylating agents (HMAs) for FLT3-mutated disease, per 2025 guidelines. IDH1 and IDH2 inhibitors block mutant enzymes that produce the oncometabolite 2-hydroxyglutarate, inhibiting in AML blasts; these mutations occur in 15-20% of cases. , an IDH1 inhibitor, achieved a complete remission () rate of 30.4% as monotherapy in relapsed/refractory IDH1-mutated AML unfit for intensive , with OS of 9.3 months. Enasidenib, targeting IDH2, yielded a CR rate of 28.9% in similar patients, also improving OS to 9.3 months. In unfit newly diagnosed patients, combinations with enhance responses, with ivosidenib plus azacitidine achieving CR in 61% and enasidenib plus azacitidine in 53%, establishing these as preferred options for mutation-positive unfit AML per 2025 NCCN guidelines. The inhibitor , combined with HMAs, has transformed treatment for older or unfit AML patients by promoting in cells dependent on anti-apoptotic pathways. In the phase 3 VIALE-A trial, plus in patients aged ≥75 or unfit for intensive therapy resulted in a CR/CR with incomplete hematologic recovery rate of 66.4%, with median OS of 14.7 months versus 9.6 months with alone. Real-world data and 2025 NCCN guidelines confirm CR rates of 60-80% in this population, positioning venetoclax-HMA as a frontline standard regardless of status, though responses are shorter in secondary AML. Emerging therapies include menin inhibitors, which disrupt the menin-MLL interaction critical for leukemogenesis in NPM1-mutated (6% of AML) or KMT2A-rearranged (5-10%) cases. Revumenib, the first-in-class menin inhibitor, received FDA approval in 2025 for relapsed/ NPM1-mutated AML, achieving MRD-negative CR rates of 30% in phase 2 trials. Ziftomenib, another menin inhibitor, showed a composite CR/CRh rate of 22% and an overall response rate of 35% in phase 2 NPM1-mutated trials and received FDA approval on November 13, 2025, for relapsed or NPM1-mutated AML in patients aged 1 year and older. Bispecific antibodies, such as flotetuzumab (CD123 x CD3), induce T-cell mediated against CD123-expressing blasts; phase 1/2 trials in AML reported CR rates of 30-40% with a step-up dosing to mitigate . Immunotherapies like CAR-T cells targeting or CD123 remain investigational as of 2025, with phase 1 trials showing 50-60% response rates but challenges in heterogeneity and persistence limiting approvals.

Management in special populations

Management of acute myeloid leukemia (AML) in pregnant patients requires a multidisciplinary approach involving hematologists, obstetricians, and neonatologists to balance maternal and fetal risks. is generally withheld during the first due to the high risk of congenital malformations from teratogenic agents like and cytarabine; instead, supportive care such as transfusions and hydroxyurea may be used if necessary to control disease until the second . In patients with (APL), all-trans retinoic acid (ATRA) combined with (ATO) is preferred, as this regimen has demonstrated safety and efficacy in without significant fetal harm when initiated after the first . For those requiring (HSCT), delivery is typically planned around 32-34 weeks gestation to allow maternal recovery and proceed with transplant post-partum, minimizing preterm complications while addressing leukemia progression. In elderly or unfit patients, defined as those over 75 years or with frailty, low-intensity therapies are favored over intensive regimens due to better tolerability and comparable or superior outcomes in this population. The combination of hypomethylating agents (HMA) such as or with achieves complete remission (CR) rates of approximately 66%, surpassing historical CR rates of 40-50% with intensive "7+3" cytarabine-anthracycline , which carries early mortality risks of 20-30% in older adults. This approach reduces treatment-related toxicity while extending overall survival, particularly in those with adverse or comorbidities. Patients with significant comorbidities undergo comprehensive fitness assessments to guide therapy selection, with tools like the Hematopoietic Cell Transplantation-Comorbidity Index (HCT-CI) evaluating organ-specific risks such as cardiac, pulmonary, hepatic, and renal dysfunction to predict treatment tolerance. A high HCT-CI score (≥3) correlates with increased early mortality and poor , often leading to best supportive care focused on symptom palliation, transfusions, and antimicrobial prophylaxis rather than disease-modifying treatments. Fitness evaluation integrates data, , and geriatric assessments to tailor interventions and avoid aggressive therapies that exacerbate underlying conditions. Pediatric AML management follows intensive multi-agent chemotherapy protocols similar to those in adults, including cytarabine and , but achieves higher cure rates of 65-70% five-year overall survival due to favorable biology and tolerance of dose-intensive regimens. For certain subtypes, such as those with lymphoid features or specific , ALL-like protocols incorporating , corticosteroids, and have been adapted to improve outcomes beyond standard AML approaches. As of 2025, evolving guidelines emphasize biology-driven treatment strategies over age-based decisions for AML, as evidenced by trials demonstrating that molecular risk profiles and fitness metrics better predict response to therapies like venetoclax-based regimens than chronological age alone. This shift allows broader access to intensive options for biologically favorable elderly patients while reserving low-intensity paths for those with high-risk features, improving personalized care across populations.

Prognosis

Factors influencing outcome

Several factors influence the outcome in acute myeloid leukemia (AML), including cytogenetic abnormalities, molecular alterations, clinical characteristics at , treatment response, and emerging biomarkers. These variables help predict survival and risk of relapse, guiding individualized management while building on initial risk stratification performed at . Cytogenetic abnormalities play a central role in . Favorable , such as the t(8;21) translocation involving RUNX1::RUNX1T1, are associated with improved outcomes, with approximately 60% of patients achieving 5-year overall (OS). In contrast, adverse like a complex (defined as three or more unrelated abnormalities) confer poor , with 5-year OS rates typically ranging from 10% to 20%. Molecular markers further refine risk assessment. Mutations in NPM1 without concomitant FLT3-internal tandem duplication (FLT3-ITD) are favorable, linked to higher remission rates and better long-term survival. Conversely, TP53 mutations represent the worst prognostic factor, associated with resistance to therapy and markedly reduced OS, often below 10% at 5 years. Clinical features at presentation also impact survival. Age greater than 60 years approximately halves OS compared to younger patients, due to increased comorbidities and treatment intolerance. Secondary AML, arising from prior or therapy-related causes, carries a worse than disease, with median OS often limited to 6-10 months. Additionally, a (WBC) count exceeding 100,000/μL at is adverse, correlating with higher early mortality and risk. Post-treatment response, particularly measurable residual disease (MRD) status, strongly predicts outcome. Achieving MRD negativity after induction therapy improves 5-year OS by 20-30% compared to MRD-positive cases, reflecting lower potential. High CD123 expression on leukemic blasts is associated with hyperproliferation, therapy resistance, and inferior survival, emerging as an adverse prognostic marker in contemporary risk models.

Survival statistics

The overall 5-year relative for patients with acute myeloid leukemia (AML) is approximately 32%, based on recent , , and End Results (SEER) Program data. This rate varies significantly by age, with approximately 50-60% of patients under 60 years achieving 5-year , compared to 10-20% for those over 60 years. Survival outcomes differ markedly by AML subtype. (APL), a distinct subtype, has a cure rate exceeding 80-90% with targeted therapies like all-trans and . Core-binding factor AML, characterized by favorable cytogenetic abnormalities such as t(8;21) or inv(16), achieves 5-year of around 60-70% with standard . Over the past two decades, AML survival has improved substantially, rising from about 15-20% in the early 2000s to 30-32% in recent years, largely attributable to the integration of targeted therapies and better supportive care. remains common, occurring in 40-50% of patients within 2 years of achieving complete remission, with median overall survival post- typically ranging from 6-9 months. In pediatric patients, long-term survival rates are higher, reaching 65-70% at 5 years, reflecting advances in risk-adapted and transplantation.

Epidemiology

Incidence and prevalence

Acute myeloid leukemia (AML) has an estimated global incidence of approximately 145,000 new cases annually as of recent data, corresponding to an age-standardized incidence rate of about 1.8 to 2.0 per 100,000 population worldwide. In the United States, the incidence rate is higher at 4.3 new cases per 100,000 individuals per year, with an estimated 22,010 new diagnoses projected for 2025. The disease predominantly affects older adults, with a median age at of 68 years, and cases in individuals under 20 years are rare, accounting for less than 5% of all AML diagnoses. Prevalence remains low due to the aggressive nature of AML and limited long-term , with approximately 79,000 living with the disease as of 2022, or roughly 1 in 4,200 individuals. Globally, data are similarly constrained by high mortality rates shortly after . AML exhibits a slight predominance, with a of about 1.2:1 (s to females). The incidence of AML is rising globally at a rate of approximately 1-2% per year, largely attributable to population aging, which increases the absolute number of cases in older demographics.

Geographic and demographic variations

Acute myeloid leukemia (AML) exhibits notable geographic variations in incidence, with higher rates observed in developed regions compared to less developed areas. In , the age-standardized incidence rate (ASIR) for AML stands at approximately 4.0 per 100,000 population, contrasting with lower rates of around 1.5-2.0 per 100,000 in , potentially linked to differences in environmental exposures, aging populations, and diagnostic capabilities. Similarly, reports an ASIR of 4.2 per 100,000, while South-Central Asia shows rates closer to 2.5 per 100,000, highlighting a pattern where high-income regions bear a disproportionate burden. Demographic factors, particularly and , also influence AML incidence. , non-Hispanic individuals experience the highest rates at 4.1 new cases per 100,000, compared to 3.0 per 100,000 among Asian/Pacific Islanders—a difference of about 37%—with populations at an intermediate 3.5 per 100,000. These disparities persist even after adjusting for age, suggesting underlying genetic or environmental influences specific to ethnic groups. individuals have rates of 3.2 per 100,000, further underscoring the gradient where face elevated risk relative to Asians and other minorities. Socioeconomic status contributes to variations in AML presentation and diagnosis. Lower socioeconomic groups often experience delayed diagnoses, leading to presentation at older ages and a higher proportion of certain subtypes, such as (APL), which comprises up to 20-30% of AML cases in low-resource settings like , compared to 5-10% in high-income countries. This may stem from limited access to advanced diagnostics and environmental factors prevalent in underserved areas, though survival outcomes are also adversely affected by socioeconomic barriers. In pediatric populations, AML risk is markedly elevated in children with , who face a 150-fold increased likelihood compared to the general , with this association observed uniformly across ethnic and geographic groups due to the genetic basis of the syndrome. Globally, GLOBOCAN projections indicate that AML cases will rise to approximately 200,000 annually by 2040, driven by population aging despite stable or declining ASIRs in many regions.

History

Early descriptions

The recognition of acute myeloid leukemia (AML) emerged in the mid-19th century amid early investigations into abnormal blood conditions. In 1845, Scottish physician John Hughes Bennett provided one of the first clinical descriptions of based on a case of what he termed "leucocythæmia" or "white cell blood," marking the initial observation of the disease. In 1847, , a pathologist, reported a similar case based on findings in a patient with and abnormal proliferation, coining the term "leukämie" to denote the white blood appearance of the fluid extracted from the . By the mid-19th century, efforts to classify subtypes began, with Nikolaus Friedreich, a pathologist, introducing the first distinction between acute and forms in 1857. Friedreich's report on a case of rapidly fatal with extensive infiltration in organs highlighted the aggressive nature of the acute variant, which he differentiated from slower-progressing cases based on clinical course and postmortem findings. In the early , morphological analysis advanced the understanding of AML through microscopic examination of blood and bone marrow cells. In 1900, Swiss pathologist Otto Naegeli identified myeloblasts as the characteristic immature myeloid cells in what he termed "acute myeloblastic leukemia," distinguishing them from lymphoid blasts and establishing the myeloid lineage as central to the disease. This was further refined in 1913 when German physicians Hilmar Reschad and Victor Schilling-Torgau described monocytic leukemia as a variant featuring monocyte-like cells, adding to the morphological distinctions within acute leukemias. A major milestone in subclassifying AML occurred in 1976 with the French-American-British (FAB) cooperative group's proposal for a standardized system based on morphological, cytochemical, and clinical features. Led by John M. Bennett and colleagues, the FAB classification divided AML into subtypes (M1 through M6), such as acute myeloblastic leukemia without maturation (M1) and , providing the first widely adopted framework for and . Early treatments for AML were limited and largely palliative, with rudimentary approaches like compounds showing anecdotal benefits in leukemias, such as chronic myeloid leukemia, during the 1930s, though mechanisms were unknown and efficacy was inconsistent without modern supportive care. Prior to the advent of cytogenetic techniques in the late , diagnosing AML subtypes posed significant challenges, relying solely on light microscopy of Romanowsky-stained blood and bone marrow smears to identify and . This approach often led to inconsistencies in , as subtle differences between myeloid and lymphoid blasts or among myeloid variants were difficult to discern without genetic or immunophenotypic , complicating and selection.

Modern developments

The cytogenetic era in acute myeloid leukemia (AML) research began in the late 1970s with the identification of recurrent chromosomal abnormalities that provided critical insights into leukemogenesis. In 1977, Janet Rowley and colleagues reported the t(15;17) translocation in (APL), a subtype of AML, marking one of the first consistent cytogenetic markers associated with a specific . This discovery paved the way for understanding genes like PML-RARA, which disrupt normal myeloid . By the 1980s, these findings led to the development of all-trans (ATRA), a that induces in APL cells by counteracting the effects of the PML-RARA ; ATRA was first clinically tested in 1987 and revolutionized APL management, transforming it from a highly fatal disease to one with cure rates exceeding 90%. The 1990s and 2000s shifted focus to , uncovering recurrent gene that refined AML classification and . FLT3 , particularly internal tandem duplications (ITD), were first identified in 1998 as activating alterations in up to 30% of AML cases, correlating with poor due to enhanced signaling. Similarly, were discovered in 2005, found in approximately 30% of normal AML and often conferring a favorable outcome when without concomitant FLT3-ITD. These molecular insights culminated in the European LeukemiaNet (ELN) guidelines, first published in 2009 for and management, which integrated cytogenetic and molecular data for ; subsequent updates in 2017 and 2022 further incorporated like and RUNX1, while the 2025 ELN recommendations emphasize measurable residual disease (MRD) monitoring and refined genetic categories. The 2010s heralded a boom in targeted therapies, driven by the molecular targets identified earlier, leading to several U.S. (FDA) approvals that improved outcomes in genetically defined AML subsets. Midostaurin, a multi-kinase targeting FLT3, received FDA approval in 2017 for addition to standard in newly diagnosed FLT3-mutated AML, based on the RATIFY trial showing improved overall survival. , a selective FLT3 , was approved in 2018 for relapsed/refractory FLT3-mutated AML, demonstrating superior survival compared to salvage in the ADMIRAL trial. , a , gained accelerated approval in 2018 (full in 2020) for combination with hypomethylating agents in older or unfit patients, achieving complete remission rates of 60-70% in trials like VIALE-A. IDH inhibitors, such as enasidenib (IDH2, approved 2017) and (IDH1, approved 2018), targeted mutant enzymes in 15-20% of AML cases, inducing durable remissions by restoring . By 2025, advancements integrated next-generation sequencing (NGS) into routine practice, with ELN and (NCCN) guidelines recommending comprehensive genomic profiling at diagnosis to guide , including identification of rare actionable . trials, such as the study presented in 2025, employed AI-driven genomic classification across over 4,000 patients to refine AML subtypes and explore bispecific antibodies and CAR-T cells, showing promising response rates in MRD-positive disease. These developments have driven survival improvements, with 5-year overall survival rising from approximately 10-15% in the to around 30% by the mid-2020s, particularly in younger patients and favorable-risk groups, though challenges persist in older adults.

References

  1. [1]
    Acute Myeloid Leukemia - StatPearls - NCBI Bookshelf
    Apr 27, 2024 · Acute myeloid leukemia (AML) is a rapidly progressing myeloid neoplasm characterized by the clonal expansion of immature myeloid-derived cells, ...
  2. [2]
    Acute myelogenous leukemia - Symptoms and causes - Mayo Clinic
    Dec 20, 2024 · Acute myelogenous leukemia, also called AML, is a cancer of the blood and bone marrow. Bone marrow is the soft matter inside bones where blood cells are made.
  3. [3]
    Acute Myeloid Leukemia Treatment - NCI - National Cancer Institute
    May 16, 2025 · Signs and symptoms of AML include fever, feeling tired, and easy bruising or bleeding. Tests that examine the blood and bone marrow are used to ...
  4. [4]
    Acute Myeloid Leukemia (AML) Subtypes and Prognostic Factors
    Mar 4, 2025 · If there are no major gene or chromosome changes in the AML cells, usually at least 20% of the cells in the bone marrow or blood must be blasts ...
  5. [5]
    Childhood Acute Myeloid Leukemia Treatment (PDQ®) - NCI
    Apr 15, 2025 · Acute myeloid leukemia (AML). AML is a clonal disorder caused by malignant transformation of a bone marrow–derived, self-renewing stem cell or ...Juvenile Myelomonocytic... · Childhood Myelodysplastic...
  6. [6]
    Acute Myeloid Leukemia Treatment (PDQ®) - National Cancer Institute
    Mar 14, 2025 · Bone marrow blasts <5%; absence of blasts with Auer rods; absence of extramedullary disease; no hematologic recovery required. PR, All ...
  7. [7]
    Key Statistics for Acute Myeloid Leukemia (AML)
    Mar 4, 2025 · About 22,010 people will be diagnosed with AML. Most cases will be in adults. About 11,090 people will die from AML. Again, most of these deaths ...
  8. [8]
    What's new in AML Classification… | College of American Pathologists
    The defining genetic abnormalities for AML in WHO 2022 and the recurrent genetic abnormalities ICC 2022 are largely the same as WHO 2017 with mild differences ...
  9. [9]
    Acute Myeloid Leukemia: 2025 Update on Diagnosis, Risk ...
    Feb 12, 2025 · In 2022 the WHO fifth edition [22] and the International consensus criteria (ICC) [23] AML classification systems were published, each ...
  10. [10]
    International Consensus Classification of Myeloid Neoplasms and ...
    This report summarizes the new International Consensus Classification (ICC) with a focus on myeloid neoplasms and acute leukemia (Table 1). A separately ...
  11. [11]
    FLT3-ITD Mutation and Allogeneic Stem Cell Transplantation ... - NIH
    Patients may also present with infections or constitutional symptoms, such as weight loss and night sweats [4]. The diagnosis of AML is based on the ...
  12. [12]
    Signs and Symptoms of Acute Myeloid Leukemia (AML)
    Mar 4, 2025 · Symptoms caused by high numbers of leukemia cells: Bleeding and clotting problems, Bone or joint pain, Swelling in the abdomen.Symptoms caused by low... · Symptoms caused by high...
  13. [13]
    Gingival hyperplasia: An initial oral manifestation of acute myeloid ...
    [15] Gingival infiltration of leukemic cells is most commonly seen in acute myelomonocytic leukemia (M4) and acute monocytic leukemia (M5).[16] All subtypes of ...
  14. [14]
    Cutaneous presentation preceding acute monocytic leukemia - NIH
    Mar 10, 2017 · We report 4 cases of AML cutis, where skin infiltration precedes any blood or bone marrow evidence of leukemia.
  15. [15]
    Leukemia Cutis - StatPearls - NCBI Bookshelf
    Jul 17, 2023 · Leukemia cutis is skin involvement in patients with peripheral leukemia; it is relatively rare and typically signifies an advanced disease stage.Missing: hypertrophy | Show results with:hypertrophy
  16. [16]
    Oral Manifestation as the Main Sign of an Advanced Stage Acute ...
    Oral manifestations, mainly spontaneous bleeding, are a common finding in acute myelocytic leukemia and may represent the initial evidence of the disease.Missing: respiratory distress
  17. [17]
    Infections in Patients with Acute Leukemia - PMC - PubMed Central
    Upper respiratory tract infections (URTI) predominate, with rhinorrhea being the most common manifestation. Progression to lower respiratory tract infection ( ...Missing: ulcers | Show results with:ulcers
  18. [18]
    Nordic Guidelines for Germline Predisposition to Myeloid...
    Individuals with MNs due to germline predisposition exhibit increased risk for the development of MNs, mainly acute myeloid leukemia (AML) and myelodysplastic ...
  19. [19]
    Germline Predisposition in Hematologic Malignancies: Testing ...
    May 20, 2024 · The frequency of an underlying germline predisposition syndrome in patients presenting with a HM is currently estimated at 5%-10%,11,12,32,33 ...
  20. [20]
    Li-Fraumeni Syndrome - GeneReviews® - NCBI Bookshelf
    May 1, 2025 · The lifetime risks of cancer for women and men with classic LFS are 90% and 70%, respectively, and 50% of cancers occur prior to age 40 years.Table 1. [Molecular Genetic... · p.Arg337His · Genes and Databases]. · Table 4
  21. [21]
    Hematologic Malignancy Spectrum Characterized in Li-Fraumeni ...
    Aug 22, 2025 · Hematologic malignancies account for 4%-10% of cancers in individuals with Li-Fraumeni syndrome, but their phenotypic spectrum and clinical ...
  22. [22]
    Myeloid Leukemia of Down Syndrome - PMC - NIH
    Jun 21, 2023 · DS children have a 50-fold increased incidence of acute leukemia during the first 5 years of life. The acute leukemias in approximately 60% of ...
  23. [23]
    The link between Down syndrome and childhood leukaemia
    Sep 1, 2022 · Children with Down syndrome are 150 times more likely to develop acute myeloid leukaemia (AML) than children without Down syndrome.
  24. [24]
    Estimation of the prevalence of Fanconi anemia among patients with ...
    Analysis of several FA cohorts indicates that FA patients are more than 500-fold more likely to develop AML than the general population [2,3]. In addition, AML ...
  25. [25]
    How I treat MDS and AML in Fanconi anemia | Blood
    Jun 16, 2016 · The cumulative incidence of AML ranges from 10% to 37% by age 50 years, with the most common time to develop leukemia being in the teenage years ...Abstract · Clonal evolution, MDS, and... · Outcome, current management...
  26. [26]
    Cancer in dyskeratosis congenita - PMC - NIH
    All of these syndromes have an increased risk of acute myeloid leukemia (AML), and FA, DC, and DBA appear to have increased risks of solid tumors. A brief ...
  27. [27]
    Dyskeratosis congenita: natural history of the disease through the ...
    Jul 31, 2023 · Dyskeratosis congenita (DC) is a multisystem and ultra-rare hereditary disease characterized by somatic involvement, bone marrow failure, and predisposition to ...
  28. [28]
    CEBPA-Associated Familial Acute Myeloid Leukemia (PDQ®)
    Nov 25, 2024 · Germline CEBPA pathogenic variants appear to be highly penetrant, conferring greater than a 90% lifetime risk to develop AML.
  29. [29]
    CEBPA-Associated Familial Acute Myeloid Leukemia (AML) - NCBI
    Oct 21, 2010 · Germline CEBPA pathogenic variants exhibit complete or near-complete penetrance for the development of AML in families reported to date. Each ...Diagnosis · Clinical Characteristics · Management · Genetic Counseling
  30. [30]
    Germline RUNX1 Intragenic Deletion: Implications for Accurate ... - NIH
    Patients harbor mild to moderate thrombocytopenia and a risk of around 35% to develop AML or myelodysplasia. This disease (OMIM 601399) is due to germline ...
  31. [31]
    How I diagnose myeloid neoplasms with germline predisposition
    Jul 18, 2023 · Pathologists play a crucial role in the initial diagnosis of germline predisposition to myeloid neoplasia and subsequent surveillance for disease progression.CASE PRESENTATIONS · DEFINING GERMLINE... · QUESTIONS FOR THE...<|control11|><|separator|>
  32. [32]
    Routine Screening for Germline Variants in Patients With AML
    Nov 23, 2021 · The researchers suggest these findings support the need for routine screening of patients with AML for pathogenic germline variants.
  33. [33]
    Chemical Exposures and Risk of Acute Myeloid Leukemia and ...
    Known risk factors for AML include increasing age, male sex, prior chemotherapy, cigarette smoking, obesity, exposure to benzene and other chemicals including ...
  34. [34]
    Risk Factors for Acute Myeloid Leukemia (AML)
    Mar 4, 2025 · AML can occur at any age, but it becomes more common as people get older. Being born male. AML is slightly more common in men than in women. The ...Missing: incidence | Show results with:incidence
  35. [35]
    Benzene and acute myeloid leukemia - PubMed
    Used in the production of ubiquitous items such as plastics, lubricants, rubbers, dyes, and pesticides, benzene may be responsible for the higher risk of acute ...
  36. [36]
    Occupational pesticide exposure increases risk of acute myeloid ...
    Jan 21, 2021 · A higher risk to develop chronic lymphoid malignancies has been demonstrated to be associated with occupational pesticide exposure (OPE).
  37. [37]
    3.5.6 Acute myeloid leukaemia - Tobacco in Australia
    A 2016 meta-analysis of 27 studies showed that people who currently smoke had a 1.36-fold higher odds of developing AML than those who never smoked, and that ...
  38. [38]
    Leukemia Risks among Atomic-bomb Survivors – Radiation Effects ...
    Excess leukemia was the earliest delayed effect of radiation exposure seen in A-bomb survivors. Japanese physician Takuso Yamawaki in Hiroshima first noted ...
  39. [39]
    Late effect of atomic bomb radiation on myeloid disorders - PubMed
    Leukemia was the first malignancy linked to radiation exposure in atomic bomb survivors. Clear evidence of the dose-dependent excess risk of three major types ...
  40. [40]
    Therapy-related myelodysplasia and acute myeloid leukemia - PMC
    Alkylating agent-related t-MDS/AML usually appears 4 to 7 years after exposure to the mutagenic agent. Approximately two-thirds of patients present with MDS and ...
  41. [41]
    Diagnosis and treatment of therapy-related acute myeloid leukemia
    t-AML accounts for 7 %–8 % of AML cases and primarily occurs in elderly patients. •. t-AML is associated with adverse cytogenetics and shorter survival than de ...
  42. [42]
    Acute myeloid leukemia in a farmer with long term exposure...
    Van Maele-Fabry et al reported a significantly increased AML risk related to occupational pesticide exposure in cohort studies, although no significant ...
  43. [43]
    A Systematic Review and Meta-analysis of Childhood Leukemia and ...
    Childhood leukemia was associated with prenatal maternal occupational pesticide exposure in analyses of all studies combined and in several subgroups.
  44. [44]
    The incidence of secondary leukemias - PubMed
    Secondary leukemias account for 10-30% of all AML. The majority of secondary leukemias resulting from the use of cytotoxic drugs can be divided into two well ...
  45. [45]
    Secondary Acute Myeloid Leukemia: Pathogenesis and Treatment
    Secondary acute myeloid leukemia is characterized by a worse prognosis than its de novo counterparts, with a 5-year overall survival of <30%.Abstract · INTRODUCTION · PATHOGENESIS · TREATMENT OF PATIENTS...
  46. [46]
    Navigating the contested borders between myelodysplastic ...
    It is now known that approximately 20-30% of cases of MDS progress to overt AML while approximately 30% of cases of AML are thought to arise out of an ...
  47. [47]
    Acute Myeloid Leukemia Following Myeloproliferative Neoplasms
    In general, risk for LT varies by MPN subtype. PMF has the highest risk of LT (5.8-20.6% risk in 10 years) followed by PV (2.3-8.7% risk in 10 years) ...
  48. [48]
    Clinical and Molecular Determinants of Clonal Evolution in Aplastic ...
    A trend toward a higher risk of malignant progression was observed in AA (12.8%) and AA/PNH (13.1%) as compared with classical hemolytic PNH (3.4%, both P = .06 ...
  49. [49]
    Characterization of therapy-related acute myeloid leukemia
    Apr 1, 2023 · Between 5-10% of all AML cases have been reported to be therapy-related, and treatment for a solid cancer increases the risk of AML up to 10- ...
  50. [50]
    Acute Myeloid Leukemia Stem Cells: Origin, Characteristics ... - NIH
    Here we review leukemia stem cell (LSC) biology, particularly as it relates to the very heterogeneous nature of AML and to its high disease relapse rate.
  51. [51]
    The Impact of the Cellular Origin in Acute Myeloid Leukemia - NIH
    Here, we review how different mouse models, despite their inherent limitations, have functionally demonstrated that cellular origin plays a critical role in ...
  52. [52]
    Update on acute myeloid leukemia stem cells - PubMed Central - NIH
    Experimental evidence in mice shows that LSCs may arise either through neoplastic changes initiated in normal self-renewing HSCs or downstream progenitors cells ...
  53. [53]
    Acute Myeloid Leukemia: 2025 Update on Diagnosis, Risk ... - NIH
    AML with myelodysplasia‐related gene mutations (as delineated by ICC) is now defined as an adverse risk entity, which is delineated by the presence of a ...
  54. [54]
    A Mutational Landscape in Acute Myeloid Leukemia - MDPI
    NPM1, located on chromosome 5q35, is the most commonly mutated gene in AML. Mutations are commonly seen in exon 12, accounting for 75–80% of these cases, and ...
  55. [55]
    Acute myelogenous leukemia - Diagnosis and treatment - Mayo Clinic
    Dec 20, 2024 · Acute myeloid leukemia diagnosis often begins with an exam that checks for bruising, bleeding in the mouth or gums, infection, and swollen lymph nodes.Missing: definition | Show results with:definition
  56. [56]
    Tests for Acute Myeloid Leukemia (AML) - American Cancer Society
    Oct 28, 2025 · During the physical exam, the doctor will check your eyes, mouth, skin, lymph nodes, liver, spleen, and nervous system, and will look for areas ...Bone Marrow Samples · Lab Tests To Diagnose Aml... · Chromosome And Gene Tests
  57. [57]
    Bone marrow biopsy and aspiration - Mayo Clinic
    the spongy tissue inside some of your larger ...
  58. [58]
    Diagnosis and management of AML in adults - ASH Publications
    Sep 22, 2022 · The 4 categories described above are designated as AML/MDS if the bone marrow or blood blast count is 10% to 19% and as AML with ≥20% blasts ( ...Abstract · AML classification · European LeukemiaNet... · Therapy for AML
  59. [59]
    What is new in acute myeloid leukemia classification? - PMC - NIH
    Apr 15, 2024 · The blast count diagnostic criteria in ICC, consistent with other entities, is ≥ 10%. In contrast, WHO2022 suggests a blast count of ≥ 20%.
  60. [60]
    Acute Myeloid Leukemia (AML) - Hematology and Oncology
    Bone marrow examination (aspiration and needle biopsy) is routinely done. Blast cells in the bone marrow are typically between 25 and 95% in patients with AML.
  61. [61]
    Flow Cytometry in the Diagnosis of Leukemias - NCBI
    Flow cytometry has become a powerful immunophenotyping tool, and plays a critically important role in the diagnosis of various leukemias.
  62. [62]
    Immunophenotypic pattern of myeloid populations by flow cytometry ...
    Mature monocytes are positive for CD11b, CD11c, CD13, CD14, CD33, and CD64, and may express CD2 and CD4. Blasts in acute myeloid leukemias (AML) with minimal ...
  63. [63]
    Acute Myeloid Leukemia: Diagnosis and Evaluation by Flow Cytometry
    Flow cytometry offers rapid and sensitive immunophenotyping through a multiparametric approach and is a pivotal laboratory tool for the classification of AML.
  64. [64]
    Cytogenetics and gene mutations influence survival in older patients ...
    Oct 1, 2018 · Deletions of part or all of chromosomes 5, 7, or 17 occur in 5–10% of all patients with AML, are often associated with complex and monosomal ...
  65. [65]
    Cytogenetic heterogeneity negatively impacts outcomes in patients ...
    Mar 1, 2015 · Acquired cytogenetic abnormalities can be detected in approximately 50% of patients and are important independent predictors of initial response ...Missing: prevalence | Show results with:prevalence
  66. [66]
    An update on the molecular pathogenesis and potential therapeutic ...
    Jan 14, 2020 · RUNX1-RUNX1T1, one of the core-binding factor leukemias, is one of the most common subtypes of AML with recurrent genetic abnormalities and is associated with ...
  67. [67]
    Molecular characterization of AML with RUNX1-RUNX1T1 at ...
    Feb 4, 2019 · We show that at diagnosis patients who relapse following MR have a higher burden of co-mutated genes than patients that do not relapse.
  68. [68]
    Bone marrow biopsy morbidity and mortality - PubMed
    The most frequent and most serious adverse event was haemorrhage, reported in 14 patients, necessitating blood transfusion in six patients and leading to the ...
  69. [69]
  70. [70]
    Genetic risk classification for adults with AML receiving less ...
    Nov 21, 2024 · The 2024 ELN Less-Intensive genetic risk classification is applicable to patients receiving HMA monotherapy, HMA/VEN, or AZA/IVO (for IDH1mut ...
  71. [71]
    A scoring system for AML patients aged 70 years or older, eligible ...
    Jul 11, 2022 · The European Scoring System ≥70, easy for routine calculation, predicts long-term survival in older AML patients considered for intensive chemotherapy.
  72. [72]
    Acute Myeloid Leukemia - Guidelines Detail - NCCN
    Guidelines for Patients · Acute Myeloid Leukemia-English Version 2025 · Acute Myeloid Leukemia-Spanish Version 2025 ...
  73. [73]
  74. [74]
    Latest NCCN Guidelines on the Standard of Care for Patients With ...
    Oct 14, 2025 · For younger patients suitable for intensive treatment, the standard approach involves induction chemotherapy with cytarabine and daunorubicin (7 ...
  75. [75]
    Initial FLAG-Ida outperforms 7+3 for high-risk AML - MDEdge
    Feb 24, 2020 · “FLAG-Ida is a more efficacious regimen when used for initial induction compared to 3+7. It appears to produce better survival and improved ...
  76. [76]
    Improved Post remission survival of non- favorable risk Acute ...
    The fludarabine, high dose cytarabine and G-CSF with or without idarubicin combination regimen, referred to as FLAG+/-Ida, is commonly used as a salvage regimen ...
  77. [77]
    [PDF] NCCN Guidelines for Patients: Acute Myeloid Leukemia
    Dec 20, 2024 · Accurate testing is needed to diagnose and treat AML. A diagnosis of AML is based on the presence of myeloid blasts in the bone marrow or blood ...
  78. [78]
    The treatment of acute promyelocytic leukemia in 2023
    Jan 18, 2023 · Although ATRA with ATO and an anthracycline is the preferred induction regimen for patients with high-risk APL at most centers, GO remains an ...
  79. [79]
    Tumor lysis syndrome in induction therapy for acute myeloid ...
    Apr 5, 2024 · Guidelines recommend rasburicase for high-risk patients to prevent tumor lysis syndrome (TLS). However, little information is available on ...
  80. [80]
    Tumor lysis syndrome in patients with acute myeloid leukemia
    Jan 1, 2008 · 4 The standard management of TLS includes generous hyperhydration, urine alkalinization and uric acid reduction with allopurinol. Despite these ...Abstract · Introduction · Design and Methods · Discussion
  81. [81]
    Tumor Lysis Syndrome Treatment & Management
    Sep 16, 2024 · Conservative management and prevention of tumor lysis syndrome are similar, focusing on hydration and control of serum uric acid.
  82. [82]
    Typical Treatment of Acute Myeloid Leukemia (Except APL)
    May 4, 2025 · Induction. The first phase of treatment for AML is aimed at quickly getting rid of as many leukemia cells as possible. The intensity of the ...
  83. [83]
    Maintenance therapy in AML: Treatment options and clinical ...
    Nov 8, 2024 · We discuss the role, therapeutic options, and clinical considerations of a maintenance strategy in the treatment of AML.
  84. [84]
  85. [85]
    The Graft-Versus-Leukemia Effect in AML - Frontiers
    Crucially, alloreactivity of donor T cells against the patient's leukemia is responsible for the graft-versus-leukemia (GvL) effect, which is a major mechanism ...Evidence for a GvL Effect · NK Cells and the GvL Effect · Improving Transplant...
  86. [86]
    Indications for haematopoietic cell transplantation and CAR-T for ...
    Sep 9, 2025 · Overall, allo-HCT in first complete remission (CR1) is recommended for adverse-risk AML and the majority of intermediate-risk AML defined by ...
  87. [87]
    long term analysis of GITMO AML-R2 trial | Blood Cancer Journal
    Aug 21, 2024 · For decades, the association between busulfan and cyclophosphamide (BuCy2) has been the standard of care conditioning regimen for HSCT in AML ...
  88. [88]
    Acute myeloid leukemia management and research in 2025 - PMC
    Dec 19, 2024 · Allogeneic HSCT should be an integral part of the continuum approach that considers chemotherapy, targeted therapies, and post‐HSCT maintenance.
  89. [89]
    Targeted and epigenetic therapies for acute myeloid leukemia ...
    Sep 26, 2025 · The article discusses specific targeted agents like IDH, FLT3, BCL2, and Menin inhibitors that address molecular abnormalities in AML. It ...2 Targeted Therapy In Aml · 4 Overview Of Aml Treatments · 4.1 Gene Therapy For Aml
  90. [90]
    Midostaurin reduces relapse in FLT3-mutant acute myeloid leukemia
    The prospective randomized, placebo-controlled CALGB 10603/RATIFY trial (Alliance) demonstrated a statistically significant overall survival benefit from ...
  91. [91]
    Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3 ...
    Oct 30, 2019 · Our trial showed a survival advantage for FLT3-targeted therapy in patients with relapsed or refractory AML after data were censored for ...
  92. [92]
    Full article: IDH1-mutated relapsed or refractory AML
    Jun 27, 2019 · Ivosidenib in monotherapy achieved complete remission (CR) of 24%, overall response of 42%, and median overall survival of 9 months in R/R AML, ...
  93. [93]
    Targeting IDH1 and IDH2 Mutations in Acute Myeloid Leukemia
    Overall response rates in the primary efficacy populations were 41.6% for ivosidenib and 38.8% for enasidenib, with 34.4% and 28.9% of patients, respectively, ...Idh1 And Idh2 Mutations In... · Idh-Mutated Aml And Response... · Idh Inhibitors In...
  94. [94]
    Newly diagnosed acute myeloid leukemia in unfit patients - NIH
    Aug 16, 2025 · A total of 23 patients were treated with ivosidenib-azacitidine combination therapy (median age: 76 years), CR was achieved in 61% (median time ...
  95. [95]
    Clinical outcomes of hypomethylating agents plus Venetoclax as ...
    Feb 11, 2024 · In a study conducted at Mayo Clinic, HMA plus VEN induced CR/CRi in 83% of patients with an ASXL1 mutation. The high response rates observed in ...
  96. [96]
  97. [97]
    Ziftomenib in Relapsed or Refractory NPM1-Mutated AML
    Sep 25, 2025 · Ziftomenib—a potent, highly selective, oral menin inhibitor—disrupts formation of the menin-KMT2A complex and blocks the oncogenic activity of ...
  98. [98]
    Flotetuzumab as salvage immunotherapy for refractory acute ... - NIH
    Although CD123 is not specifically expressed in putative AML progenitor and stem cells, its targeting could lead to the eradication of leukemia stem cell pools.
  99. [99]
    New Type of CAR T Cells Successfully Target AML in Pilot Study
    Aug 29, 2025 · The first-ever clinical trial at MSK to evaluate a novel CAR T cell therapy for AML is showing promise.What Is CAR T Therapy? · What Makes These CAR T...
  100. [100]
    CAR-T and CAR-NK cell therapies in AML: breaking barriers ... - NIH
    Oct 23, 2025 · Recent clinical trials of CAR-T/NK therapies in AML have yielded cautiously optimistic results. For instance, CD123-directed CAR-T cells ...
  101. [101]
    How I treat leukemia during pregnancy | Blood - ASH Publications
    The management of the leukemias in pregnancy requires close collaboration with obstetric and neonatology colleagues as both the maternal and fetal outcomes must ...Introduction · Acute leukemia · Supportive care · Chronic leukemias
  102. [102]
    Leukemia in pregnancy: Diagnosis and therapeutic approach ...
    Management of AML in the second and third trimesters of pregnancy. Chemotherapy and pregnancy maintenance are recommended between the 13 and 24th weeks, while ...
  103. [103]
    Older Adults With Newly Diagnosed AML - ASCO Publications
    May 8, 2023 · However, pooled data from VIALE-A and a phase Ib HMA/venetoclax study demonstrated that patients with FLT3-ITD had a median OS of only 9.9 ...
  104. [104]
    Comparative efficacy and safety in low-intensity treatment for acute ...
    Apr 15, 2025 · Historical studies using intensive chemotherapy in older AML patients reported CR rates of 40–50%, 4- to 8 week mortality rates of 26–36%, ...
  105. [105]
    Acute Myeloid Leukemia–Composite Model for Mortality Risk ...
    Sep 7, 2017 · An AML composite model of augmented HCT-CI, age, and cytogenetic/molecular risks has a strong AUC of 0.76 for 1-year mortality.
  106. [106]
    Assessing eligibility for treatment in acute myeloid leukemia in 2023
    The HCT-CI has been shown to predict early death and survival among patients >60 years old and undergoing induction therapy for AML [Citation44]. It is ...
  107. [107]
    Fitness assessment in acute myeloid leukemia: recommendations ...
    Apr 29, 2025 · Supportive therapy should be initiated at AML presentation to revert disease-induced conditions that hamper accurate fitness assessment,102 ...
  108. [108]
    Prognostic Factors and Survival Rates for Childhood Leukemia
    The overall 5-year survival rate for children with AML is about 65-70%. Here are general survival statistics based on risk groups. These numbers come from past ...
  109. [109]
    Treatment Outcomes of Adolescents Compared to Younger Pediatric ...
    Mar 14, 2024 · Survival improved regardless of age; however, treatment outcomes for adolescents and young adults are still worse compared to younger children, ...
  110. [110]
    Alliance Global Study Challenges Age-Based Treatment Decisions ...
    Oct 14, 2025 · “Our findings support a more flexible, biology-driven approach to AML treatment and trial design. Age alone should not be a gatekeeper to ...
  111. [111]
    New Alliance Global Study Questions Age-Based Approaches in ...
    Oct 14, 2025 · Instead, these findings advocate for an integrative, biology-centric model to tailor treatment strategies, potentially broadening patient access ...
  112. [112]
    Prognostic significance of karyotype in de novo adult acute myeloid ...
    A good prognostic impact was found for t(8;21), t(15;17) and inv(16). The best factor proved to be t(8;21) (5-year survival probability: 50%), followed by t(15; ...Missing: OS | Show results with:OS
  113. [113]
    Acute Myeloid Leukemia with a Complex Karyotype - PMC - NIH
    CR durations are short, with medians of 6 to 8 months, and the probabilities or remaining in CR at 3 years are between 0 and 11%, and at 5 years between 0 and 9 ...
  114. [114]
    How I treat older patients with acute myeloid leukemia
    May 29, 2018 · 2 Outcomes of AML in older adults (aged >60 years) are dismal, with survival rates reported to be <10% at 3 years and <5% at 5 years.3, 4 ...
  115. [115]
    Long-term real-world evidence of CPX-351 of high-risk patients with ...
    Feb 25, 2025 · A meta-analysis of 81 studies reported 11 151 patients with AML with MRD-negative CR and showed an improvement of 5-year OS (68% vs 34%) and 5- ...
  116. [116]
    Acute Myeloid Leukemia — Cancer Stat Facts - SEER
    Death rates from acute myeloid leukemia are higher among older adults, or those 65 and older. People with leukemia have many treatment options, and ...
  117. [117]
    Acute promyelocytic leukemia: long-term outcomes from the ...
    Jan 9, 2025 · In conclusion, based on the largest APL patient cohort reported so far, we show that the ATRA-ATO treatment is associated with >80% survival at ...
  118. [118]
    Core-binding factor acute myeloid leukemia: long-term outcome of ...
    Apr 7, 2022 · The 1-, 3-, 5- and 10-year overall survival (OS) rates were 90%, 70 ... 1: Overall and Relpase Free Survival in 70 patients with Core-Binding ...
  119. [119]
    Acute myeloid leukaemia statistics - Cancer Council Victoria
    It demonstrates that five-year relative survival has increased for AML between 1983-1987 and 2018-2022 from 14% to 32%. Figure 6: Trend in five year relative ...<|control11|><|separator|>
  120. [120]
    Outcomes and genetic dynamics of acute myeloid leukemia at first ...
    May 2, 2024 · The median overall survival duration after relapse was 5.3 months, with a 1-year overall survival rate of 17.6%. Complex karyotype (hazard ...Abstract · Introduction · Methods · Results
  121. [121]
    Global, national, and regional burden of acute myeloid leukemia ...
    Jan 11, 2024 · It is estimated that in 2020, there were 19,940 new cases of AML diagnosed in the United States, with 11,180 resulting in death (6–8).
  122. [122]
    Global, regional, and national burden of acute myeloid leukemia ...
    Sep 11, 2024 · According to our study, the incidence of AML has continued to rise globally from 79,372 in 1990 to 144,645 in 2021 and AML affected the male and ...
  123. [123]
    Global, regional, and national burden of acute leukemia and its risk ...
    Jun 10, 2025 · In 2021 (Tables S1–S4), the number of global AML incidence was 144,645.60 cases. AML caused 130,189.07 death cases and 4,135,056.14 disability- ...
  124. [124]
    Are Race and Ethnicity Risk Factors for Leukemia?
    Jan 28, 2022 · White people appear to be most at risk for developing leukemia, whereas Black Americans appear to have the lowest risk.
  125. [125]
    Socioeconomic Status and Overall Survival Among Patients With ...
    Mar 4, 2024 · For AML, a negative association was observed between low SES and survival (HR, 1.49 [95% CI, 1.25-1.76]), but the association was attenuated in ...
  126. [126]
    Full article: The impact of medical education and networking on the ...
    Citation9 The higher APL frequency may be due to the age distribution of the population, since APL is known to be more frequent among young adults, whereas the ...<|separator|>
  127. [127]
    Leukemia Risk in a Cohort of 3.9 Million Children With and Without ...
    Children with Down syndrome had higher risk of AML before age 5 than other children (HR=399, 95%CI=281–566). Largest HRs were for megakaryoblastic leukemia ...Missing: variations | Show results with:variations
  128. [128]
    Consortium on leukemia in children with Down syndrome offers ...
    Nov 2, 2023 · Current statistics put the risk of developing ALL at 20-fold higher and the risk of AML at 150-fold higher in children with Down syndrome ...
  129. [129]
    The discovery and early understanding of leukemia - PubMed
    Oct 26, 2011 · That same year, Rudolf Virchow defined a reversed white and red blood cell balance. He introduced the disease as leukämie in 1847. Henry Fuller ...
  130. [130]
    Acute Myeloid Leukemia; Decided Victories, Disappointments, and ...
    Jan 1, 2008 · John Hughes Bennett (1812–1875) was only 33 years old and Rudolf Virchow (1821–1902) only 24 in 1845 when each described patients at autopsy
  131. [131]
    First contributors in the history of leukemia
    Virchow noted the enlarged spleen and liver, but also described blood vessels filled with material resembling pus. Virchow described the disparity between white ...
  132. [132]
    Acute Myeloid Leukemia History - News-Medical
    Nov 13, 2019 · First discussed in medical literature in the 1800s, acute myeloid leukemia is a progressive and aggressive type of cancer affecting the white blood cells.
  133. [133]
    Proposals for the classification of the acute leukaemias ... - PubMed
    Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group ... Br J Haematol. 1976 Aug;33(4):451-8. doi: 10.1111/j ...
  134. [134]
    From an old remedy to a magic bullet: molecular mechanisms ... - NIH
    Arsenic had been used in treating malignancies from the 18th to mid-20th century. In the past 3 decades, arsenic was revived and shown to be able to induce ...
  135. [135]
    Acute Promyelocytic Leukemia: A History over 60 Years—From the ...
    At the genetic level, APL was found to correspond to a unique t(15;17) chromosomal translocation. All-Trans Retinoic Acid Era. The Beginnings of ATRA Therapy.
  136. [136]
    History of Developing Acute Promyelocytic Leukemia Treatment and ...
    Cloning of the t(15;17) breakpoint in APL, later identified the RARA gene fusion to an unknown locus named myeloid leukemia (MYL), later renamed promyelocytic ...
  137. [137]
    FLT3 mutations in acute myeloid leukemia: Therapeutic paradigm ...
    14, 15 More than 20 years after the discovery of the FLT3 gene mutation ... mutations in FLT3, NPM1 and DNMT3A genes are frequently identified in AML patients.Missing: timeline | Show results with:timeline
  138. [138]
    The discovery of NPM1 mutations in acute myeloid leukemia - NIH
    A search for NPM1 mutations (in combination with other mutations) represents a critical step in the genetic-based risk stratification of AML patients according ...Missing: timeline | Show results with:timeline
  139. [139]
    2025 European LeukemiaNet recommendations for the ... - Nature
    Jul 11, 2025 · In this 5th version of the European LeukemiaNet guidance for adult patients, there are important changes in several areas of management based on evidence ...
  140. [140]
    Recent drug approvals for acute myeloid leukemia
    Sep 18, 2019 · In this review article, we will discuss these new agents approved for the treatment of AML within the last 2 years, and will outline the mechanistic features ...Missing: timeline | Show results with:timeline
  141. [141]
    New agents in acute myeloid leukemia (AML) - PMC - NIH
    Jul 30, 2020 · Venetoclax, in combination with azacitidine or decitabine or low dose cytarabine, received accelerated FDA approval in Nov 2018 for newly ...Missing: timeline | Show results with:timeline
  142. [142]
    #EHA2025: HARMONY and PETHEMA trials redefine AML ...
    Jul 22, 2025 · One abstract that stood out was S148, a HARMONY Alliance study that used AI-based, unsupervised genomic classification in over 4,000 patients ...Missing: NCCN | Show results with:NCCN
  143. [143]
    Acute Myeloid Leukemia -- Historical Perspective and Progress in ...
    We are witnessing an ongoing “slow-motion” revolution in AML reseach and therapy, with the approval of nine agents for different AML indications since 2017 : ...Missing: timeline | Show results with:timeline