advances in acute myeloid leukemia

20
STATE OF THE ART REVIEW the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 1 Advances in acute myeloid leukemia Laura F Newell, Rachel J Cook Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR, USA Correspondence to: L F Newell [email protected] Cite this as: BMJ 2021;375:n2026 http://dx.doi.org/10.1136/bmj.n2026 Series explanation: State of the Art Reviews are commissioned on the basis of their relevance to academics and specialists in the US and internationally. For this reason they are written predominantly by US authors. Introduction Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy characterized by the proliferation of myeloid blasts or progranulocytes that fail to undergo normal differentiation. Outcomes for patients with AML are often described as “dire” or “poor,” despite the fact that patients are sometimes cured and life can often be prolonged with treatment. Standard intensive chemotherapy for AML (combining an anthracycline and cytarabine) has been in use for more than 40 years. Recently, major advances in molecular and cell biology have improved our understanding of the pathophysiology of AML, expanded treatment options, and provided a deeper understanding of how and why individual patients may be at increased risk for development of leukemia. These advances provide personalized treatment options for many patients and may provide future opportunities to preempt the development of AML. We have not included a discussion of acute promyelocytic leukemia (APL), which has unique disease biology, diagnosis, treatment, and monitoring algorithms. Epidemiology An estimated 21 450 new cases of AML occurred in the US in 2019 (1.2% of all new cancer diagnoses), with an increasing incidence with advanced age: 25.1% of new AML cases were among adults aged 65-74 years and 33.7% among those aged 75 and older. The five year survival for patients with AML is a dismal 28.3%, with an estimated 10 920 deaths from AML in 2019. As with the incidence, the percentage of deaths also increases with age, being highest in patients aged 75 or older (43.7%). 1 Sources and selection criteria For this review, we searched PubMed and Medline databases from 2010 to 2020. We also included studies written before 2010 that were highly cited or of historical significance, and we selected only articles from peer reviewed journals written in English. We used the search terms “acute myeloid leukemia”, “AML”, “AML and decision making”, “FLT3”, “genetic and AML”, “bone marrow transplant and AML”, “stem cell transplant and AML”, “CHIP and AML”, “CAR-T and AML”, and “treatment and AML”. We prioritized studies by their relevance to the search criteria, by study year and publication date, and by study design and number of patients included. We excluded case reports but included selected small case series. We prioritized larger, more recent studies, as well as landmark and novel trials in the field, when available. As expected, in the section on emerging therapies, we found many early phase trials with low numbers of patients. We took into account the fact that the review process was inherently limited by the prevalence of the disease (many of the pivotal phase III registration studies enrolled only a few hundred patients, and more recent studies of novel agents often enroll <100). Additionally, patient outcomes are often censored at time of hematopoietic stem cell transplant (HSCT), leading to abbreviated time spent on the new therapies. Consequently, this review is not intended to be formally systematic, but our evaluation of combined datasets from trials allows for reinterpretation of risk stratification and deeper understanding of initial metrics used for overall prognostication and prediction of responses to specific therapies. 2 3 Clinical presentation The presenting signs and symptoms can vary from patients without symptoms identified, through routine screening blood work, to those with critical illness due to sepsis in the setting of severe bone ABSTRACT Acute myeloid leukemia (AML) is an uncommon but potentially catastrophic diagnosis with historically high mortality rates. The standard of care treatment remained unchanged for decades; however, recent discoveries of molecular drivers of leukemogenesis and disease progression have led to novel therapies for AML. Ongoing research and clinical trials are actively seeking to personalize therapy by identifying molecular targets, discovering patient specific and disease specific risk factors, and identifying effective combinations of modalities and drugs. This review focuses on important updates in diagnostic and disease classifications that reflect new understanding of the biology of AML, its mutational heterogeneity, some important genetic and environmental risk factors, and new treatment options including cytotoxic chemotherapy, novel targeted agents, and cellular therapies. on 28 January 2022 by guest. Protected by copyright. http://www.bmj.com/ BMJ: first published as 10.1136/bmj.n2026 on 6 October 2021. Downloaded from

Upload: others

Post on 28-Jan-2022

5 views

Category:

Documents


0 download

TRANSCRIPT

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 1

Advances in acute myeloid leukemiaLaura F Newell, Rachel J Cook

Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR, USACorrespondence to: L F Newell [email protected] this as: BMJ 2021;375:n2026 http://dx.doi.org/10.1136/bmj.n2026

Series explanation: State of the Art Reviews are commissioned on the basis of their relevance to academics and specialists in the US and internationally. For this reason they are written predominantly by US authors.

IntroductionAcute myeloid leukemia (AML) is a heterogeneous hematologic malignancy characterized by the proliferation of myeloid blasts or progranulocytes that fail to undergo normal differentiation. Outcomes for patients with AML are often described as “dire” or “poor,” despite the fact that patients are sometimes cured and life can often be prolonged with treatment. Standard intensive chemotherapy for AML (combining an anthracycline and cytarabine) has been in use for more than 40 years. Recently, major advances in molecular and cell biology have improved our understanding of the pathophysiology of AML, expanded treatment options, and provided a deeper understanding of how and why individual patients may be at increased risk for development of leukemia. These advances provide personalized treatment options for many patients and may provide future opportunities to preempt the development of AML. We have not included a discussion of acute promyelocytic leukemia (APL), which has unique disease biology, diagnosis, treatment, and monitoring algorithms.

EpidemiologyAn estimated 21 450 new cases of AML occurred in the US in 2019 (1.2% of all new cancer diagnoses), with an increasing incidence with advanced age: 25.1% of new AML cases were among adults aged 65-74 years and 33.7% among those aged 75 and older. The five year survival for patients with AML is a dismal 28.3%, with an estimated 10 920 deaths from AML in 2019. As with the incidence, the percentage of deaths also increases with age, being highest in patients aged 75 or older (43.7%).1

Sources and selection criteriaFor this review, we searched PubMed and Medline databases from 2010 to 2020. We also included

studies written before 2010 that were highly cited or of historical significance, and we selected only articles from peer reviewed journals written in English. We used the search terms “acute myeloid leukemia”, “AML”, “AML and decision making”, “FLT3”, “genetic and AML”, “bone marrow transplant and AML”, “stem cell transplant and AML”, “CHIP and AML”, “CAR-T and AML”, and “treatment and AML”. We prioritized studies by their relevance to the search criteria, by study year and publication date, and by study design and number of patients included. We excluded case reports but included selected small case series.

We prioritized larger, more recent studies, as well as landmark and novel trials in the field, when available. As expected, in the section on emerging therapies, we found many early phase trials with low numbers of patients. We took into account the fact that the review process was inherently limited by the prevalence of the disease (many of the pivotal phase III registration studies enrolled only a few hundred patients, and more recent studies of novel agents often enroll <100). Additionally, patient outcomes are often censored at time of hematopoietic stem cell transplant (HSCT), leading to abbreviated time spent on the new therapies. Consequently, this review is not intended to be formally systematic, but our evaluation of combined datasets from trials allows for reinterpretation of risk stratification and deeper understanding of initial metrics used for overall prognostication and prediction of responses to specific therapies.2 3

Clinical presentationThe presenting signs and symptoms can vary from patients without symptoms identified, through routine screening blood work, to those with critical illness due to sepsis in the setting of severe bone

ABSTRACT

Acute myeloid leukemia (AML) is an uncommon but potentially catastrophic diagnosis with historically high mortality rates. The standard of care treatment remained unchanged for decades; however, recent discoveries of molecular drivers of leukemogenesis and disease progression have led to novel therapies for AML. Ongoing research and clinical trials are actively seeking to personalize therapy by identifying molecular targets, discovering patient specific and disease specific risk factors, and identifying effective combinations of modalities and drugs. This review focuses on important updates in diagnostic and disease classifications that reflect new understanding of the biology of AML, its mutational heterogeneity, some important genetic and environmental risk factors, and new treatment options including cytotoxic chemotherapy, novel targeted agents, and cellular therapies.

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

2 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

marrow failure. Early manifestations of disease are non-specific and may be initially seen by care givers of disparate types (fig 1). Assessing a patient’s medical history, social history, and general fitness early in the evaluation is important, as these factors weigh heavily in the decisions about treatment. Recent studies have shown the prognostic value of fitness evaluations, particularly in older adults, and several reviews provide clear roadmaps for completing such evaluations.4-6

DiagnosisFormal diagnosis of AML can be made either from a peripheral blood sample or from a bone marrow biopsy, depending on the white blood count and the presence or absence of circulating blasts. Although some leukemic disorders can be diagnosed by the detection of pathognomonic chromosomal abnormalities—for example, APL with chromosome 15;17 translocation (t(15;17) and core binding factor leukemias (t(8;21), inv(16), or t(16;16)—ordinarily the diagnosis of AML requires the presence of an abnormal fraction of immature myeloid cells (either myeloblasts or promyelocytes). Histologically, myeloid leukemias require the presence of ≥20% blasts in either a blood or a bone marrow sample. A bone marrow assessment typically includes both an aspiration of the liquid portion of the marrow and a core biopsy. Immunophenotyping is done on the aspirate or, if the marrow is inaspirable, can be done on a core that has been crushed.

Flow cytometric analyses for certain cell surface and cytoplasmic markers confirm the diagnosis of AML, and the specific immunophenotype at diagnosis is useful after treatment to evaluate for persistence of disease. Cytogenetic analysis of metaphase chromosomes reveals the “karyotype,” the most prognostic element of the patient’s leukemia evaluations. The World Health Organization requires evaluations of AML to include clinical history (for example, a patient who has previously undergone treatment with cytotoxic chemotherapy or radiation will automatically be given a diagnosis of a “treatment related AML”), cytogenetics, molecular genetics, morphology, and immunophenotype.7 8 Molecular testing with next generation sequencing (NGS) has gained importance at diagnosis, for risk stratification, to identify targets for treatment, and as a baseline with which future assessments of disease burden will be compared to assess for depth of response.9-11 All patients should be assessed for comorbidities, functional status, and social support, as well as for their goals and values.12

AML classification and risk stratificationBox 1 reviews classification schemes for AML.13-17 19

Box 2 provides details about AML disease monitoring and “day 14” mid-induction bone marrow assessment,20-27 and box 3 describes the nuances of monitoring for minimal/measurable residual disease (MRD).28-40

“Risk factors” for AMLFamilial pre-disposition syndromesThat families exist in which multiple members (in both childhood and adulthood) are affected by a myeloid malignancy has been long recognized. In recent decades, specific genotypes associated with heritable hematologic malignancies have been defined. In the most recent WHO categorization,7 provisional recognition of this phenomenon includes multiple categories for myeloid neoplasms with germline predispositions including those that are isolated abnormalities (CEBPA, DDX41) or associated with additional abnormalities (either a platelet disorder or organ dysfunction), including RUNX1, ANKRD26, ETV6, GATA2, Li Fraumeni syndrome (TP53), and bone marrow failure syndromes such as Fanconi anemia and dyskeratosis congenita. Awareness and recognition of these conditions are increasing, which allows for both tailored treatment and genetic counseling.

Some affected families are identified by astute clinicians who note multiple affected family members or phenotypic abnormalities in the patient, whereas others are identified because of increasingly large NGS panels including myeloid predisposition genes. In the latter case, the presence of a suggestive family history may or may not be known. In a study of 360 patients with hematologic malignancies, NGS was completed using a panel including nine genes implicated in hereditary hematologic malignancies, resulting in the identification of 74 (21%) pathogenic variants of which six were ultimately confirmed to be germline.41 The increasing use of NGS that includes germline predisposition mutations allows for identification of at risk individuals. Confirmation of germline mutations should be completed on genetic material obtained by skin biopsy or buccal swab. A recent detailed review by the University of Chicago Hematopoietic Malignancies Cancer Risk Team describes clinical and laboratory features suggestive of a hereditary myeloid malignancy syndrome.42

Identifying these conditions to avoid treating the wrong disease (for example, presuming refractory immune thrombocytopenic purpura in a patient with an underlying platelet disorder predisposing to AML) is critically important, to avoid using affected family members with no symptoms as donors for HSCT and to recognize associated conditions for which the patient may need screening or modification of leukemia treatment (for example, patients with Fanconi anemia in whom cytotoxic chemotherapy should be minimized). Expert consensus recommendations are available for management of family members who have been identified as having the causal mutation but who have not developed the malignancy, which include consultation with a disease expert, genetic counseling, and screening for associated malignancies/conditions in addition to blood work and a baseline bone marrow biopsy to ensure that no hematologic malignancy has developed and as a baseline for future studies.43

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 3

Acquired risk factorsEnvironmental exposuresThe adverse effect of environmental exposures on development of leukemia has been well described among the survivors of the 1945 Hiroshima and Nagasaki atomic bombs. Survivors were noted to have an increased risk for the development of leukemia (and other malignancies) following radiation exposure, with increasing risk dependent on age at exposure and dose received. For radiation associated AML, the risk persisted throughout the study follow-up period, out to 55 years after the bomb detonations.44 Additionally, hematologic malignancies including AML and myelodysplastic syndrome associated with the Chernobyl nuclear power plant accident have been described, particularly in the clean-up workers with high exposures to ionizing radiation.45

Secondary AMLSecondary AML refers to myeloid leukemia that develops after an antecedent hematologic disorder or newly diagnosed AML presenting with myelodysplasia related changes and represents approximately 20% of all AML.46 The WHO classification of AML with myelodysplasia related changes (AML-MRC) requires the presence of at least 50% dysplastic cells in at least two cell lineages, excluding cases with NPM1 or biallelic CEBPA mutations.7 Myelodysplastic syndrome and secondary AML often have overlapping cytogenetic and molecular abnormalities but are formally distinguished by the percentage of myeloblasts (<20% equals myelodysplastic syndrome, ≥20% blasts equals AML). Approximately 30% of patients with myelodysplastic syndrome develop secondary AML in the course of their disease. AML-MRC is associated with poor prognosis and lower complete remission rates after conventional induction chemotherapy.47-51

Risk factors for the transformation from myelodysplastic syndrome to secondary AML include increased circulating peripheral blood blasts, complex cytogenetics, high risk molecular abnormalities, and certain epigenetic and DNA methylation changes.52 As opposed to de novo AML, secondary AML is characterized by multilineage dysplasia, complex karyotypes, and aberrant expression of multidrug resistance proteins and antiapoptotic proteins, as well as subclonal heterogeneity.53 The overall prognosis is poor, with low response rates to cytotoxic chemotherapy and a reported overall survival of six to 12 months.54

Disease progression to secondary AML can evolve stepwise, with the accumulation of somatic mutations and subclonal disease progression occurring over time (box 4).55 56 The pathogenesis of secondary AML is complex and dynamically evolving, with the acquisition of mutations in dominant and subclonal populations that respond differentially to chemotherapy. This may affect responses to current therapies and contribute to disease relapse, as well as provide the potential for targeted therapeutic

Fig 1 | Clinical presentation of acute myeloid leukemia (AML). Patients with a new AML diagnosis may present for care across the healthcare system, with patients seeking care for varied manifestations of AML: dentists for gingival hyperplasia, dermatologists for new rashes, cardiologists for cardiac symptoms related to anemia, neurologists for new weakness or cranial nerve abnormality, or primary care providers for progressive fatigue, bruising, fevers, lymphadenopathy/organomegaly, palpable masses (myeloid sarcoma), and recurrent infections. Patients range from mildly to catastrophically ill but benefit from early identification

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

4 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

intervention for secondary AML and at even earlier stages to delay or even prevent disease progression.

Therapy related AMLTherapy related AML develops in patients with previous chemotherapy (particularly alkylating agents and topoisomerase II inhibitors), radiation exposure, or both; it is associated with an extremely poor prognosis, with historically reported median

overall survival of six to eight months and up to 14.6 months in a recent series from the Italian Network on secondary leukemias.57 The only curative treatment option is HSCT. Box 5 provides details of the incidence, classification, and risk of therapy related AML.58 59 62-65 The pathogenesis of and possible genetic susceptibility for the development of therapy related AML is complex and involves the acquisition of somatic mutations

Box 1: Classification schemes for acute myeloid leukemia (AML)French-American-British (FAB) classification• The FAB classification scheme was first proposed in 1976.13 Subtypes of AML were defined on the basis of morphologic features and cytochemical

methods, categorized as M0 through M7 ○M0-M5: include leukemia involving myeloid blasts with varying degree of maturation ○M6: acute erythroid leukemia ○M7: acute megakaryocytic leukemia ○M3: represents the distinct subtype of acute promyelocytic leukemia (APL)

• Using FAB criteria, the most common subtypes were M2 (25%) and M4 (20%) disease• Subsequent modifications to the FAB classification scheme were made to incorporate emerging genetic findings14

• However, more recent classification schemes, based on molecular and prognostic scoring systems, have largely replaced use of the FAB scheme

World Health Organization classification• WHO introduced a new classification scheme for myeloid neoplasms in 2001 and revised it in 2008.15 16 The WHO categorization reflected

increasing emphasis on morphologic, genetic, and clinical factors ○Separation of de novo AML, from secondary AML evolving from myelodysplastic syndrome, was an important distinction, as well as subtypes of therapy related disease ○The initial classification required 30% myeloblasts for diagnosis of AML, but the revised WHO guidelines lowered the threshold for AML diagnosis to 20% blasts (notable exceptions being APL and core binding factor AML)

• The most recent 2016 WHO revision updated the previously defined disease categories, incorporating new diagnostic markers and clinical impact of specific gene mutations.7 Many of these revisions involved prognostication derived from gene expression analyses and next generation sequencing studies

○Categories include AML with recurrent genetic abnormalities, AML with myelodysplasia related changes, therapy related myeloid neoplasms, myeloid sarcoma, myeloid proliferations related to Down syndrome, and AML not otherwise specified ○Additionally, a new category, the myeloid neoplasms with germline predisposition, was added to incorporate the subgroup of familial AML associated with germline mutations (eg, germline RUNX1, GATA2, CEBPA)

European Leukemia-Net (ELN) classification• The ELN categories were developed to correlate genetic abnormalities with clinical variables and prognostic impact17

• Most recently updated in 2017,18 these guidelines provide classifications to assess disease risk or resistance. The categories are divided into favorable, intermediate, and adverse subtypes based on presence of cytogenetic abnormalities and mutations

• The favorable category includes: ○Core binding factor acute myeloid leukemia (AML), defined by the cytogenetic abnormalities t(8;21) and inv(16) ○This subtype also reflects the favorable prognosis of AML with NPM1 or biallelic CEBPA mutations, regardless of coexisting gene mutations, as well as the similar favorable impact of NPM1 with low allelic ratio (<0.5) FLT3-ITD versus NPM1 without FLT3-ITD

• The intermediate classification includes: ○Those cytogenetic abnormalities not classified as favorable or adverse ○Mutated NPM1 with high allelic ratio FLT3-ITD ○Low allelic ratio FT3-ITD without mutated NPM1 ○ t(9;11) abnormalities

• The adverse risk subtype includes: ○The high risk mutation TP53 and mutations in RUNX1 and ASXL1 (not coexisting with favorable risk subtypes) ○Monosomal or complex karyotypes ○Specific cytogenetic markers of high risk disease including t(6;9), t(9;22), inv(3), KMT2A rearrangement with t(v;11q23.3), and monosomy 5 or del(5q) ○FLT3-ITD with high allelic ratio (>0.5) carries a poor prognosis and is included in the adverse risk group

• National Comprehensive Cancer Network (NCCN) guidelines• The NCCN guidelines provide AML disease risk classifications of better risk, intermediate risk, and poor risk, based on genetic risk and aligned with

evidence based treatment recommendations19

• The NCCN subtypes are similar to ELN’s, but some differences exist in the classification schemes, including: ○The distinction between wild type NPM1 with low (ELN intermediate) and high (ELN adverse) allelic ratio FLT3-ITD ○The NCCN risk stratification does not delineate between allelic ratio with any FLT3-ITD within the poor risk subtype

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 5

driven by chemotherapy/radiation induced DNA damage in the hematopoietic stem cells, which can be influenced by inherited risk factors such as alterations in telomere shortening and single nucleotide polymorphisms in genes linked to drug metabolism and DNA repair (fig 2).62 63

Clonal hematopoiesisClonal hematopoiesis, defined as the expansion of a clone of blood cells with one or more somatic mutations,66 has been described in older adults and linked to specific acquired gene mutations.70-72 Different nomenclature has been used to describe clonal hematopoiesis, including clonal hematopoiesis of indeterminate potential (CHIP), age related clonal hematopoiesis, and idiopathic or clonal cytopenias of undetermined significance. More recently, age related clonal hematopoiesis has been identified as a risk factor for the development of hematologic malignancies,73-75 as well as for adverse cardiovascular outcomes such as coronary heart disease and ischemic stroke.76 Whole exome sequencing of peripheral blood DNA from large cohorts of adults without known malignancies was used to detect somatic mutations in genes that are epigenetic regulators of hematopoiesis and known potential drivers for hematologic disease. The frequency of clonal hematopoiesis increased with age, ranging from <1% in adults aged <50 years to >10% in adults >65 years of age.73-75

Most mutations were found to occur in genes DNMT3A, TET2, and ASXL1, as well as JAK2 and PPM1D. As opposed to patients with myelodysplastic syndrome or AML, most of whom have mutations in multiple driver genes,77 most adults with clonal hematopoiesis had only one detectable mutation.73 74 The presence of clonal hematopoiesis was identified as a risk factor for subsequent development of a hematologic malignancy and for mortality, and also as a risk for therapy related myeloid disease (box 6).73 74 78-81

Because a significant fraction of patients with CHIP have clones that do not expand or progress

to myeloid malignancy, somatic clonal mutations clearly are not sufficient to function as “drivers” of leukemia. Whether the presence of such mutations alone would be actionable for leukemia prevention trials or whether pre-treatment CHIP status might affect therapeutic decisions for patients with non-hematologic malignancies remains to be determined. Additionally, although the current definition of CHIP specifies a somatic mutation frequency of greater than 2% in a defined set of genes linked with leukemogenesis, the cut-off for variant allele frequency used to define the presence of this somatic mutation will clearly evolve.66 82

Treatment paradigmsThe care of a patient with AML involves treatment of the primary malignancy and extensive supportive care measures (table 1).90-96

Cytotoxic chemotherapyFor decades, the standard intensive treatment for AML has been the combination of seven days of cytarabine with three days of an anthracycline (idarubicin, daunorubicin), referred to as the “7+3” regimen.97-101 Rates of complete remission and cure vary according to age, ranging from 60-85% complete remission and 35-40% cure for adults ≤60 years of age to 40-60% complete remission and 5-15% cure for adults >60 years of age.99 Patients with mutations in genes defined as “secondary type” or TP53 mutations have been shown to have more chemoresistant disease requiring multiple cycles of induction therapy, compared with those with de novo or pan-AML genes.56Box 7 details induction chemotherapy regimens.19 83-86 102-106

Consolidation therapyEssentially all patients who achieve complete remission after induction ultimately relapse without post-induction consolidation therapy. Consolidation options include intensive chemotherapy and stem cell transplantation (discussed below). Details of consolidation chemotherapy are in box 8 (109-111).

Box 2: Disease monitoring and mid-induction bone marrow assessment (nadir marrow)• Disease response has historically been monitored at predetermined time points during treatment, although the timing of disease re-evaluation

varies considerably between centers, clinical trials, treatment regimens, and providers, and re-evaluation is sometimes foregone completely, such as in regimens FLAG-Ida and CLAG-M

○ In patients undergoing intensive cytotoxic chemotherapy, a “nadir” bone marrow biopsy has often been performed around day 14 to determine whether residual leukemia is present that would warrant a “re-induction” course of either the same or a different chemotherapy regimen

• Considerations for completion of mid-induction bone marrow and re-induction are: ○Fitness of the patient at the time of the nadir marrow and ability to tolerate additional therapy ○Likelihood of obtaining blast clearance based on cytogenetic and molecular risk stratification

• The day 14-21 marrow biopsy remains a practice recommended by the NCCN, with determination of the need for additional therapy based on a combination of hypocellularity and blast clearance or as recommended by a clinical trial protocol

○Clearance of blasts (<5% blasts in the marrow) at this early time point is prognostically favorable20 21

○For patients with a partial response (blasts 5-19%), whether additional chemotherapy at the day 14 time point is able to improve outcomes is unclear22-24

○ In large cohorts of patients with acute myeloid leukemia on clinical trials, approximately 50% of patients with residual disease at day 14 undergo re-induction, suggesting that the bone marrow biopsy may be used as much for prognostication as for treatment decisions at day 14 and that strict adherence to guidelines risks overtreatment in this population25-28

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

6 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

Box 3: Monitoring for minimal/measurable residual disease (MRD)MRDNo uniform approach exists for MRD detection in acute myeloid leukemia (AML)• Current methods rely on multi-parameter flow cytometry or molecular platforms (quantitative polymerase chain reaction (qPCR), next generation

sequencing), and are estimated to cover approximately 60% of AML in patients <60 years of age28

Post-induction MRDThe 2018 consensus document from the European Leukemia-Net (ELN) MRD Working Party on minimal/measurable residual disease in AML provided recommendations for MRD assessment by flow cytometry10

• A retrospective analysis of 245 adult patients with AML quantified MRD status by 10 color flow cytometry in marrow samples taken at time of bone marrow recovery after induction therapy and correlated with remission responses29

• Frequency of MRD was lowest in patients who achieved complete remission (19%), whereas detection of MRD was significantly higher in patients with complete remission with incomplete platelet recovery (54.2%) and with complete remission with incomplete blood count recovery) (60.9%) (P<0.01)

Mutation specific monitoringFor patients harboring mutations targetable by current standardized qPCR assays, the PCR approach has been shown to have high sensitivity.10 Various mutations have also been shown to have differential impact, as described below:• PCR based MRD testing was shown to have an independent prognostic effect, using samples from patients with NPM1 mutations receiving

treatment in the UK National Cancer Research Institute’s AML17 trial.30 Residual presence of NPM1 mutated transcripts after the second chemotherapy cycle was associated with increased risk of disease relapse as well as decreased overall survival. Most patients who relapsed had detectable NPM1 mutations at time of relapse

• In a series of 430 patients with at least one mutation at diagnosis, MRD assessment in post-treatment marrows at time of morphologic complete remission detected persistent mutations in 51.4%.31 Mutations in DNMT3A, TET2, and ASXL1 were the most common (78.7%, 54.2%, and 51.6%, respectively) but did not correlate with an increased risk of relapse. By contrast, molecular MRD in mutations other than the above (eg, NRAS, PTPN11, KIT, KRAS) was associated with significantly greater risk of disease relapse (55.4% v 31.9%) and lower overall survival (41.9% v 66.1%) compared with no MRD

• Whether specific residual mutations (eg, DNMT3A, TET2, and ASXL1) represent persistence of the antecedent clone of pre-malignant cells,32 reflective of clonal hematopoiesis,33 is unclear, as is whether they might contribute to relapse

Pre-transplant MRDStudies have also shown a negative prognostic impact of MRD positivity before hematopoietic stem cell transplant (HSCT) on post-transplant survival and risk of relapse34-36

• In a retrospective analysis of 359 adult patients with AML undergoing myeloablative HSCT, three year overall survival and progression-free survival estimates were similar for patients with pre-transplant MRD positivity (26% and 12%) and those with pre-transplant active AML disease (23% and 13%) and inferior to those of patients entering transplant with MRD negative status (73% and 67%)

• Relapse estimates were similar between patients with MRD positive and active disease (67% and 65%, respectively), compared with patients in MRD negative remission (22%)37

Monitoring for MRD in the era of personalized medicineQuestions remain about the optimal method or combination of methods for MRD assessment, sample source (peripheral blood, bone marrow), most impactful time point(s) for analysis, differential impact of MRD by various mutations and assays, and how best to integrate MRD assessment and status into treatment algorithms• All genetic testing for MRD assessment are not the same, and nor are all assays capable of detecting MRD38

• Further standardization and validation of assays across centers or in centralized laboratories will be needed, as well as continued incorporation into response assessments in clinical trials

• How best to target post-therapy MRD with mutation directed therapy is also an area of active ongoing investigation, with novel strategies recently reviewed39 40

Box 4: Evidence for stepwise progression to secondary acute myeloid leukemia (AML)• Whole genome sequencing studies of paired bone marrow samples at time of diagnosis of myelodysplastic syndrome, as well as time of secondary

AML, found persistence of the antecedent founding clone and progression of subclone(s) at disease evolution to secondary AML55

○Additionally, the number of secondary AML specific mutations was greater in patients with slower disease progression (defined as ≥20 months), compared with patients with more rapid development of secondary AML (defined as <6 months)

• Mutational analysis comparing samples from patients with de novo AML, secondary AML, and therapy related AML by next generation sequencing was recently reported56

○ In secondary AML, there was >95% specificity for mutations occurring in eight genes (SRSF2, SF3B1, U2AF1, ZRSR2, ASXL1, EZH2, BCOR, STAG2), with an under-representation of mutations in NPM1 and rearrangements in MLL/11q23 and CBF ○Persistence of disease mutations was found in the post-induction therapy marrow samples of 69% of patients achieving morphologic complete remission, including a subset with persistence of the founder clone, suggesting a differential sensitivity to chemotherapy and showing conceptually that pre-leukemic clones likely existed before the onset of overt leukemia as well

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 7

Targeted therapy/novel agentsThe development of lower intensity treatments has increased the therapeutic options for all patients, but in particular may offer additional treatments that are more palatable to providers and to patients who are older or unfit or wish to avoid prolonged hospital admission, given the lack of mucosal toxicity and barrier disruption traditionally associated with cytotoxic chemotherapies. Comprehension of complex treatment algorithms and previously unfathomable medical complications may be difficult for patients grappling with a new life threatening diagnosis and can contribute to discordance between patients’ and providers’ perceptions of the risks and potential benefits of AML directed therapy; this can make “informed” consent difficult.107 The importance and difficulty of shared decision making in patients with AML has been well described, and the challenges are evident in population studies showing the high percentage of patients who historically have received no treatment despite data suggesting that most would benefit from some treatment.12 108 The intensity of treatment requiring time spent in clinics and hospitals in addition to up-front risks of treatment related morbidity and mortality has been, historically, a barrier to treatment for AML.109 A recent review of therapies in development for targeting pathways in AML with both novel therapies and combination therapies shows the extraordinary progress and opportunity in novel treatments.110 Table 2 lists several emerging and novel therapies for AML.111 112

Hypomethylating agents and low dose chemotherapyHypomethylating agents (azacitidine and decitabine) are administered in the clinic as an intravenous or subcutaneous (azacitidine only) injection and are appropriate for all subtypes of AML. Hypomethylating agents enhance and inhibit expression of sets of genes in cells by influencing the methylome. Many possible mechanisms of action have been proposed for the drugs, and no clear cytogenetic or molecular predictors of response are available.125

TP53 mutated AML is known to have particularly poor outcomes and is seen more commonly in older adults, patients with complex karyotypes, and patients with treatment related AML.126 127 A phase II study found that a 10 day course of decitabine improved overall survival of patients with TP53 mutated AML enough to resemble that of intermediate risk AML,128 and a five day course was sufficient in patients with newly diagnosed AML.129 The ideal hypomethylating agent regimen has not been established in the setting of TP53 mutated AML. Some data from retrospective studies support withholding traditional cytotoxic chemotherapies if the TP53 mutation status is known in advance of the time at which definitive chemotherapy must be started.130 Although the likelihood of obtaining complete remission is lower when using the hypomethylating agents, the survival benefit of being in complete remission was just as potent in patients who obtained complete remission with azacitidine as those who obtained complete remission with a “7+3”

Box 5: Therapy related acute myeloid leukemia (AML)Incidence and characteristics of therapy related AML• The incidence varies from <1% to 10%, depending on underlying disease and previous therapies, and represents <10% of all AML cases46

• Characteristic features of therapy related AML include58: ○Complex cytogenetics ○Abnormalities of chromosomes 5 and/or 7 (usually partial or complete loss) ○Translocations involving the MLL gene and chromosomes 11q23 or 21q22 (particularly after topoisomerase II inhibitors) ○Chemotherapy refractory disease ○Mutations in TP53 that are associated with adverse cytogenetics and particularly poor outcomes ○Less frequently, therapy related AML can present with cytogenetic markers of traditionally “good risk” disease—t(8;21), inv(16), or t(15;17)

Classification, onset, and risk of therapy related AML• The WHO classification of myeloid neoplasms and acute leukemia categorizes therapy related myeloid neoplasms as a distinct entity, subdivided

into therapy related myelodysplastic syndrome and therapy related AML7

• In population based cancer registries, therapy related AML has been well described in patients with Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, and multiple myeloma; after autologous hematopoietic stem cell transplantation; and after treatment for solid tumor malignancies including breast, ovarian, bone, gastrointestinal, and other cancers59

• Mutational analyses have suggested that therapy related myelodysplastic syndrome and therapy related AML have distinct molecular profiles compared with de novo AML, with differences in the frequency of mutated genes such as TP53, epigenetic regulators, and spliceosome genes60

○However, more recently, next generation sequencing showed that acquired mutations in therapy related AML and therapy related myelodysplastic syndrome were more similar to de novo AML with the same alterations than to the therapy related AML cohort as a whole56

○Thus the classification of therapy related AML may not be distinct on the basis of mutational profile alone, but driven by the complexity of karyotypic abnormalities61

• The timing of leukemia presentation varies according to previous exposures: ○Therapy related AML after topoisomerase II inhibitors occurs earlier, often within 2-3 years after treatment ○By contrast, therapy related AML after alkylating agents typically occurs later, more commonly within 5-7 years of therapy, and is often preceded by the development of therapy related myelodysplastic syndrome62

• The risk of developing therapy related AML varies depending on the alkylating agent, with a dose-response effect based on cumulative exposure63

• Therapy related AML has also been described after exposure to fludarabine and azathioprine and after solid organ transplantation64 65

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

8 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

based regimen.131 132 Azacitidine has been studied in a phase III randomized trial and showed both hematologic response rates and complete remission rates that provide benefit above conventional care and low dose cytarabine (LDAC).133 The relative benefits in terms of quality of life of complete remission versus complete remission with incomplete blood count recovery versus hematologic improvement remain understudied. LDAC given subcutaneously either once or twice daily for 10 days is also low intensity chemotherapy that does not require admission to hospital for administration and has been used in combination with novel agents.134 135

B cell lymphoma-2 inhibitionThe B cell lymphoma-2 (BCL-2) family of proteins is involved in multiple regulatory processes of apoptosis. BCL-2 inhibition was identified as a drug target for the purposes of re-enabling activation of apoptosis in malignant cells. Venetoclax is an oral BCL-2 inhibitor first developed in chronic lymphocytic leukemia (CLL), a disease in which BCL-

2 is usually significantly overexpressed leading to impaired apoptosis.136 Venetoclax proved effective in heavily pre-treated patients with CLL, occasionally leading to tumor lysis syndrome.137 Although less BCL-2 overexpression is seen in AML than in CLL, pre-clinical data suggested efficacy and the initial phase II trial of single agent venetoclax for treatment of relapsed/refractory AML showed a marginal overall response rate of 19%.138 139 Subsequent trials studied combinations of venetoclax with LDAC or hypomethylating agents, with improved results exceeding those of historic controls.140 The trial VIALE-A randomized older adult patients with newly diagnosed AML who were not candidates for induction chemotherapy to either single agent azacitidine or azacitidine plus venetoclax. Results for the combination arm showed higher complete remission and overall response rates (36.7% and 66.4%) compared with single agent azacitidine (17.9% and 28.3%).141

Initial real world data using a hypomethylating agent in combination with venetoclax continue to

Fig 2 | Pathogenesis of therapy related acute myeloid leukemia (t-AML).60 61 66-69 Experimental evidence supports the notion that chemotherapy and/or radiation selects for the expansion of a mutant clone that is more resistant to DNA damage.66 68 Normal hematopoietic cells ordinarily thrive in contact with a complex microenvironment that includes non-hematopoietic (stromal) cells and well differentiated (auxiliary) hematopoietic cells. The microenvironment normally supports stem cell function but, if challenged with genotoxic agents, can inhibit stem cell survival providing an opportunity for the selection of pre-leukemic founder mutations resistant to such agents.69 The stem cells with founder mutations marked with “?” are meant to emphasize that mutations can develop either before or as a direct result of DNA damage inflicted by the genotoxic stress. Specifically, in leukemic cells of patients with t-AML, mutations of TP53 were not only found in the post-chemotherapy leukemic cells but were also detectable at low concentrations years before initial chemotherapy.67 Finally, founder mutations alone are insufficient to “drive” the expansion of fully malignant AML clones but provide the right cellular context for the acquisition of additional mutations that subsequently drive the emergence of fully leukemic cells on 28 January 2022 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.n2026 on 6 October 2021. D

ownloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 9

support favorable response rates, though as expected they are lower than those reported in clinical trials.142 A parallel trial of venetoclax with LDAC showed a complete remission rate of 26% and overall response rate of 54% in previously untreated older adults.134 Additional details on molecular sub-populations of patients with AML who are most likely to benefit from venetoclax are emerging, showing high response rates and durable remissions in patients with NPM1 or IDH2 mutations.143 Although uncommon, tumor lysis syndrome can develop in patients starting therapy with a regimen containing venetoclax, particularly in those with white blood cell counts greater than 25×109/L. Recommended prophylaxis, monitoring, and management of tumor lysis syndrome have been well described in recent publications, in addition to practical recommendations on management of drug interactions with commonly used “azole” antifungals.144 145 The role of venetoclax in the post-transplant setting is being studied.

Myeloid cell leukemia-1 (MCL-1) is another member of the anti-apoptotic family of BCL-2 proteins for which inhibitory drugs are being developed for treatment of AML.146 MCL-1 is highly expressed in patients with untreated AML, and several MCL-1 inhibitors are in development, some of which

are being used in combination with venetoclax. Additional therapies affecting the MCL-1 apoptotic pathway include cyclin dependent kinase inhibitors, many of which are in varying stages of development and clinical trials.146

FLT3 inhibitionThe increasing recognition of genomic heterogeneity in AML has led to the development of therapies that directly target the mutations. Targeted therapies are increasingly available for specific driver mutations, although the number of patients for whom these drugs (FLT3 and isocitrate dehydrogenase inhibitors) are in routine clinical use remains a minority. Multiple novel agents target the FLT3 pathway, both FLT3-ITD mutations within the juxtamembrane domain and FLT3-TKD mutations in the tyrosine kinase domain. FLT3-ITD mutations are associated with a poor prognosis because of the increased frequency of relapse after a first complete remission has been obtained. FLT3-ITD mutations are seen in approximately 25% of patients with newly diagnosed AML and should be tested for at diagnosis.18 Testing should be quantitative and include the variant allele frequency or allelic ratio, as these are prognostic, and additional co-mutations such as NPM1 that

Box 6: Clonal hematopoiesis and myeloid diseaseClonal hematopoiesis as a risk factor for hematologic malignancy and mortality• In one study,73 the hazard ratio was:

○12.9 (95% confidence interval 5.8 to 28.7) for development of a hematologic malignancy ○1.4 (1.0 to 1.8) for risk of death

• In another study,74 the hazard ratio was: ○11.1 (3.9 to 32.6) for risk of hematologic cancer ○1.4 (1.1 for 1.8) for all cause mortality

• The risk of developing a hematologic malignancy among patients with clonal hematopoiesis was 0.5-1% per year depending on the variant allele fraction (proportion of mutated alleles), compared with <0.1% for those without clonal hematopoiesis, and these mutations persisted over time73 74 78

Therapy related clonal hematopoiesis• Clonal hematopoiesis has also been described in patients with non-hematologic malignancies, such as therapy related clonal hematopoiesis• Use of the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) to sequence paired blood derived

versus tumor derived DNA samples showed that79: ○25.1% of patients had at least one somatic mutation, with presumptive driver mutations in 4.5% of patients ○TP53 and PPM1D mutations were associated with previous chemotherapy and radiation ○Statistically significant associations were found between clonal hematopoiesis and older age, tobacco use, and previous radiation therapy but not chemotherapy ○Patients with clonal hematopoiesis had an increased risk of developing a hematologic malignancy (18 month estimate of 1% (95% confidence interval 0.5% to 1.8%) versus 0.3% (0.1% to 0.5%) for those without clonal hematopoiesis). ○Additionally, overall survival was inferior among patients with clonal hematopoiesis with a presumptive driver mutation, with the most common cause of death being disease progression of the non-hematologic malignancy

Clonal hematopoiesis and therapy related myelodysplastic syndrome and AML• Clonal hematopoiesis has also been associated with the development of therapy related AML and myelodysplastic syndrome• A nested case-control study determined the prevalence of clonal hematopoiesis in patients >70 years of age with chemotherapy treated cancer, who

subsequently did or did not develop therapy related myelodysplastic syndrome or therapy related AML80

○The prevalence of clonal hematopoiesis of indeterminate potential (CHIP) among the entire cohort was 33%, and was significantly higher in patients who developed therapy related myeloid disease (62%) compared with those who did not (27%) ○Patients with CHIP were at greater risk of developing therapy related myelodysplastic syndrome/AML than those without CHIP (adjusted odds ratio 5.75, 95% confidence interval 1.52 to 25.09; P=0.013)

• In another case-control study,81 clonal hematopoiesis with driver mutations was detected in 71% of patients who ultimately developed therapy related myeloid malignancies, compared with 31% in age matched controls.

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

10 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

further modify risk stratification.147 148 Midostaurin and gilteritinib are now approved by the US Food and Drug Administration (FDA) for up-front and relapsed/refractory FLT3-ITD AML respectively.149 150 First generation FLT3 inhibitors including sorafenib and midostaurin are yielding to next generation FLT3 inhibitors including crenolanib, quizartinib, and gilteritinib in ongoing clinical trials designed to establish the benefits of early and prolonged FLT3 inhibition for patients with newly diagnosed AML and in relapsed disease. Several recent reviews have summarized the evolving FLT3 landscape and

identified important unanswered questions.151-153 Additional tyrosine kinase inhibitors are being studied for targets other than FLT3 and are in various stages of development, as recently described.154

Isocitrate dehydrogenase inhibitionThe isocitrate dehydrogenase inhibitors ivosidenib and enasidenib target mutations in the enzymes isocitrate dehydrogenase 1 and 2 respectively that are involved in the Krebs cycle. Mutations in the isocitrate dehydrogenase enzymes cause an accumulation of oncometabolites that lead myeloid

Table 1 | Considerations for supportive careCategory Trials/referencesTransfusion supportBlood products leukocyte depleted and irradiated for all patients receiving immunosuppressive therapy (including fludarabine containing regimens, stem cell transplant)

NCCN

Prophylactic versus therapeutic transfusion regimens: increased grade 2-4 bleeding with therapeutic platelet transfusion and higher (although small) numbers of fatal CNS hemorrhages

ELN, 83-85

Conventional transfusion thresholds: prophylactic platelet transfusion for platelet count <10×109/L or with any evidence of bleeding; prophylactic RBC transfusion for hemoglobin ≤8 g/dL or for symptoms of anemia

ELN, NCCN

For coagulopathy and/or overt bleeding: maintain platelets ≥50×109/L; maintain fibrinogen >150 mg/dL with cryoprecipitate; normalize PT, PTT concentrations with fresh frozen plasma

ELN, NCCN

Tranexamic acid 83, NCT03136445, NCT02578901Growth factor supportNo clear recommendation for routine use of myeloid growth factors during initial induction therapy ELN, NCCN, 86Growth factors may be considered with post-remission therapy NCCNInfection prophylaxisAntibacterial: quinolone (or other) prophylaxis ELN, NCCN, 87 88Pneumocystis jirovecii prophylaxis for patients at increased risk, including recipients of purine analogs and recipients of ≥20 mg prednisone equivalent for ≥1 month)

88

Antiviral: aciclovir prophylaxis for herpes simplex virus seropositive patients; prophylaxis with entecavir or tenofovir for patients at high risk of hepatitis B virus reactivation

88

Antifungal: posaconazole (anti-mold activity); micafungin when azoles strictly prohibited ELN, NCCN, 89Other considerationsHydroxyurea for management of leukocytosis ELN, NCCNTumor lysis prophylaxis/monitoring: hydration with diuresis as needed; allopurinol; electrolyte monitoring; consideration for rasburicase if rapidly increasing white blood cells, elevated uric acid, renal impairment

NCCN

Steroid eye drops for patients receiving high dose cytarabine NCCNCNS=central nervous system; ELN=European Leukemia-Net; NCCN=National Comprehensive Cancer Network; PT=prothrombin time; PTT=partial thromboplastin time; RBC=red blood cell.

Box 7: Induction chemotherapyConventional chemotherapy• Fludarabine, high dose cytarabine, and idarubicin (FLAG-ida) has been shown to be an effective regimen102

○Superior rates of remission and reduced risk of relapse compared with a 10 day cytarabine plus daunorubicin regimen were shown, but also with increased toxicity and thus no improvement in overall survival

• Other intensive cytotoxic regimens incorporating clofarabine have been investigated103-105

• Cladribine, another nucleoside analog, has also been investigated in combination with daunorubicin and cytarabine (eg, the “DAC regimen”), with response rates of up to 70% in de novo acute myeloid leukemia (AML) and 40-50% in relapsed/refractory AML83 84 86 106

○ Induction regimens containing cladribine are included as category 1 recommendations in the National Comprehensive Cancer Network guidelines for AML patients <60 years, as well as in the salvage setting19

CPX-351• Recently, the US Food and Drug Administration and European Medicines Agency approved CPX-351 (Vyxeos), a liposomal dual drug encapsulation

of cytarabine and daunorubicin at a fixed 5:1 molar ratio, for the treatment of adults with newly diagnosed therapy related AML or AML with myelodysplasia related changes, on the basis of a randomized phase III study in adults aged 60-75 years85

• Patients were randomized to a 7+3 regimen versus CPX-351 ○Higher response rates were seen with CPX-351 than with conventional 7+3 chemotherapy (complete remission/complete remission with incomplete blood count recovery 47.7% v 33.3%; P=0.016) ○Median overall survival was improved with CPX-351 versus 7+3 (9.56 v 5.95 months; hazard ratio 0.69; P=0.003) ○No significant differences were seen in early mortality rates though day 30 and day 60

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 11

blasts to linger in an immature state.8 The addition of the isocitrate dehydrogenase inhibitors blocks this process and allows for resumption of more normal differentiation, sometimes leading to the complication of “differentiation syndrome,” in which the rapid maturation of many immature white blood cells leads to dyspnea, fever, pulmonary infiltrates, and hypoxia.155 156 Isocitrate dehydrogenase 1 mutations are present in approximately 7-14% of patients with AML and isocitrate dehydrogenase 2 mutations in 8-19%.157 Both drugs were initially tested as single agents in the relapsed/refractory setting to demonstrate efficacy sufficient to warrant FDA approval. Ivosidenib is also approved for use as initial therapy and is now in phase III combination studies with azacitidine for newly diagnosed disease. The availability of novel agents with potential to improve outcomes emphasizes the importance of initial diagnostic testing to identify patient specific mutations that affect not only prognosis but also choice of therapy in both the up-front and relapsed/refractory setting.

Hedgehog pathway inhibitionGlasdegib is an additional new agent in the treatment of AML that inhibits the hedgehog pathway and smoothened protein. A 2:1 randomized trial in 132 patients who were not candidates for intensive chemotherapy compared glasdegib plus LDAC versus LDAC alone. It found that the combination achieved a 17% remission rate compared with 2.3% in the single agent LDAC arm (P< 0.05).135 Ongoing trials are examining its role in the up-front setting in combination with induction chemotherapy.

TP53 pathway modulationMutations in the TP53 tumor suppressor gene occur in about 10% of new AML diagnoses and are more common in patients with secondary AML.158 Survival is markedly worse for patients with TP53 mutations and is commonly seen in patients with complex karyotypes.159 The presence of a TP53 mutation is inherently problematic for patients being treated with cytotoxic chemotherapy, as mutated p53 causes dysregulation of the apoptotic pathway which decreases the efficacy of DNA damaging chemotherapy agents.130 New agents targeting the TP53 pathway are in development, including APR-

246, which restores wild type p53 function and has been advanced to a phase III clinical trial after the phase Ib/II study of patients with myelodysplastic syndrome and oligoblastic AML showed promising results.160

Cellular therapies and immunotherapy for AMLAllogeneic hematopoietic stem cell transplantWhen the goal of treatment for AML is cure, HSCT is often a consideration for post-remission therapy in patients with intermediate or adverse risk disease for whom relapse is most likely. The efficacy of HSCT relies variably on the anti-leukemic effect of the “conditioning” (pre-transplant chemotherapy and radiation) and the graft versus leukemia effect mediated primarily through donor T cells. Candidates for HSCT may be identified shortly after diagnosis of AML on the basis of risk stratification and the patient’s characteristics or later when leukemia persists in an MRD positive state or with overt recurrence or refractory disease.10

Eligibility to proceed with a transplant is based on a favorable response to induction chemotherapy, patient’s fitness, and availability of a stem cell donor. HSCT has higher up-front treatment related mortality than chemotherapy based treatment, but for selected patient populations it can provide improved long term survival. Decision making about the risks and benefits of transplant are complicated, although clear indications of disease states that most strongly require transplant for potential cure exist.161 With increasing “alternative” donor options (donors who are less well HLA matched or from conventional HLA matched donors), transplants are becoming more available to patients. Important strides have been made in recent years in identifying options for patients who previously would not have had an identified stem cell donor, with the ready availability of umbilical cord blood units and successful transplantation of haplo-identical stem cells.162 With improvements in prophylaxis for graft versus host disease and the use of post-transplant cyclophosphamide, further improvement in outcomes with mismatched transplants may be possible in the future.163

Relapse of AML post-transplant remains the major obstacle for patients who undergo HSCT and is mediated by both persistence of disease and immune evasion. Recent studies have elegantly described mechanisms of relapse via down-regulation of HLA class II, effectively allowing the leukemia to “hide” from the donor immune system.164 165 T cell exhaustion (both CD4+ and CD8+) is an additional mechanism of relapse, for which donor lymphocyte infusions have shown efficacy.166 Ongoing studies are examining ways in which the post-transplant immune system and leukemia cells can be modulated to re-engage an effective immune response.

Maintenance therapy after transplant is also an area of active study. The randomized trial of post-transplant gilteritinib maintenance (US CTN 1506) for patients with FLT3 ITD positive disease recently

Box 8: Consolidation chemotherapy• High doses of cytarabine (up to 3000 mg/m2), six doses per cycle, given for up to four

cycles, are commonly used (dose reduced in older patients)• However, some data suggest that intermediate dose (2000 mg/m2) cytarabine may

be an alternative regimen, particularly for patients with intermediate risk AML.19 Additionally, recent studies have failed to show a benefit for high dose cytarabine regimens in favorable risk AML with younger age (<65 years),87-89 101 102 as is also discussed in the European Leukemia-Net (ELN) guideline18

• Whether higher doses of cytarabine are more effective than intermediate dosing, what the optimal number of cycles of consolidation should be, what the impact of age is on dose selection, and whether adding other agents (including additional anthracyclines) is beneficial are not yet known18 101

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

12 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

Treatment class Mechanism/target Trials/referencesSelective agentsApoptotic pathways: S64315 Mcl-1 NCT02979366 (AML, MDS) + Venetoclax NCT03672695 (AML) AMG 176 Mcl-1 NCT02675452 (R/R AML, R/R myeloma) + Venetoclax NCT03797261 (R/R AML, R/R NHL) AZD5991 +/– venetoclax Mcl-1 NCT03218683 (R/R hematologic malignancies) Alvocidib CDK9 NCT03969420 (R/R after venetoclax + HMA) + Cytarabine/mitoxantrone or cytarabine/daunorubicin

NCT03563560—2 cohorts: relapsed, refractory AML; newly diagnosed AML

+ Venetoclax NCT03441555 (R/R AML) Dinaciclib + venetoclax CDK1,2,5,9 NCT03484520 (R/R AML) Voruciclib CDK9 NCT03547115 (R/R AML, B cell malignancy) AZD4573 CDK9 NCT03263637 (R/R hematologic malignancies)TP53 pathway: APR-246 (eprenetapopt) + azacitidine Induces apoptosis by shifting TP53 toward wild-type

conformationNCT03072043 (oligoblastic TP53 mutated AML, MDS, MPN, or CMML)

FLT3 inhibition: Gilteritinib v midostaurin + induction + consolidation

FLT3-TKD or ITD NCT04027309 (newly diagnosed FLT3+ AML); NCT03836209 (newly diagnosed FLT3+ AML)

Crenolanib v midostaurin + cytarabine + daunorubicin

FLT3-TKD or ITD NCT03258931 (newly diagnosed FLT3 TKD or ITD AML)

Quizartinib + 7+3 + continuation therapy FLT3-ITD NCT02668653 (newly diagnosed FLT3-ITD+ AML) Gilteritinib + azacitidine v azacitidine FLT3-TKD or ITD NCT02752035 (newly diagnosed FLT3+ TKD or ITD AML, not

eligible for standard induction chemotherapy) CPX-351 + quizartinib FLT3-ITD NCT04209725 (R/R AML, FLT3-ITD+)Antibody based treatmentsUnconjugated/monoclonal antibodies: 1-7F9 Anti-KIR NCT01256073 (elderly AML ≥60 not eligible for HCT) BI 835858 monotherapy CD33 111; NCT01690624 (R/R AML); NCT03207191 (relapsed

post-HCT) + Decitabine NCT02632721 (R/R AML, AML ineligible for standard

intensive therapy) Cusatuzumab (JNJ-74494550) +/– azacitidine CD70 NCT04023526 (newly diagnosed AML ineligible for intensive

chemotherapy) + Venetoclax +/– azacitidine NCT04150887 (newly diagnosed AML ineligible for intensive

chemotherapy) Daratumumab CD38 NCT03067571 (R/R AML, high risk MDS); NCT03537599

(relapsed post-HCT) FLYSYN FLT3 NCT02789254 (AML with MRD) MEN1112 Bst1/CD157 NCT02353143 (R/R AML) Pembrolizumab (in combination with intensive chemotherapy)

PD-1 NCT04214249 (newly diagnosed AML)

PF-04518600 OX40 agonist NCT03390296 (R/R AML)Multivalent antibodies: AMG 330 CD33, CD3 BiTE 11; NCT02520427 (R/R AML) AMV-564 CD33, CD3 bispecific TandAb NCT03144245 (R/R AML) APVO436 CD123, CD3 bispecific NCT03647800 (R/R AML, MDS) ESK1-BiTE WT1 112 Flotetuzumab CD123, CD3 bispecific DART NCT02152956 (PIF, early relapse AML) GEM333 CD33, CD3 bispecific NCT03516760 (CD33+ R/R AML) GTB-3550 CD16, IL-15, CD33 TriKE NCT03214666 (CD33+ R/R AML, high risk MDS) JNJ-63709178 CD123, CD3 duobody NCT 02715011 (R/R AML) JNJ-67571244 CD33, CD3 bispecific NCT03915379 (R/R AML, high risk MDS) MCLA-117 CLEC12A (CLL-1), CD3 bispecific NCT03038230 (R/R AML and elderly untreated high risk

AML)Toxin conjugated antibodies: ASP1235/AGS62P1 FLT3 targeted antibody conjugated to cytotoxic payload

(microtubule disrupting agent)NCT02864290 (R/R AML)

Gemtuzumab ozogamicin CD33 targeted antibody conjugated to calicheamicin 113-120; NCT03737955 (MRD+ AML, high risk MDS/MPN); NCT03737955 (MRD+ MDS, MPN, AML)

IMGN632 CD123 targeted antibody conjugated to novel DNA alkylating agent

NCT03386513 (CD123+ R/R AML, hematologic malignancies)

IMGN779 CD33 targeted antibody conjugated to novel DNA alkylating agent

NCT02674763 (CD33+ R/R AML)

INA03 CD71 targeted antibody conjugated to MMAE NCT03957915 (R/R acute leukemia)(Continued)

Table 2 | Emerging therapies and selected clinical trials for acute myeloid leukemia

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 13

met accrual, and results are awaited. Studies of post-transplant hypomethylating agents and targeted therapies including venetoclax and immunotherapies are all ongoing with the goal of mitigating the risk of relapse. Some studies have shown that post-transplant azacitidine was safe and may prolong event-free and overall survival,167 but others have not observed such an effect.168 Additional studies are needed to identify which patients are candidates for which therapies and on what schedules in the post-transplant setting.

Chimeric antigen receptor T cell (CAR-T) therapyThe field of cellular therapy has been revolutionized by the development of CAR-T therapies, in which

autologous or allogeneic T cells are isolated and genetically modified to express chimeric antigen receptors targeting specific cell surface antigens. The re-programmed T cells possess effector T cell function, combined with the ability of antibodies to recognize targeted antigens, in a major histocompatibility complex (MHC) independent manner.169 170 One method of immune evasion used by tumor cells is the loss of MHC class I expression, leading to avoidance by the tumor of T cell surveillance and recognition171; thus the ability of CAR-T cells to function in an MHC independent manner is important in efforts to improve the anti-tumor effect.170 The first approvals by the FDA were for the anti-CD19 CAR-T product tisagenlecleucel (Kymriah) for relapsed or refractory

Treatment class Mechanism/target Trials/referencesCheckpoint blockade combination: Magrolimab (Hu5F9-G4) +/– azacitidine CD47 monoclonal antibody and macrophage checkpoint

inhibitor (SIRPα receptor)NCT03248479 (R/R AML or AML ineligible for standard intensive therapy)

+ Azacitidine + venetoclax NCT04435691 (AML)Cytotoxic agents Cladribine based regimens Purine analog (resistant to adenosine deaminase) 83 84 86 106 Clofarabine based regimens Purine analog (resistant to adenosine deaminase) 103-105 CPX-351 Liposomal cytarabine and daunorubicin 85 CPX-351 + gemtuzumab ozogamicin Liposomal cytarabine and daunorubicin + CD33 targeted

antibody conjugated to calicheamicinNCT03878927 (AML >55 years of age); NCT03904251 (relapsed AML)

G-CLAM + gemtuzumab ozogamicin Cytotoxic chemotherapy + CD33 targeted antibody conjugated to calicheamicin

NCT03531918 (untreated AML, high grade myeloid neoplasm)

Cellular therapiesCAR-T: 121 122 CART-19 CD19 CAR NCT03896854 (R/R CD19+ AML) CAR-T CD19 CD19 CAR NCT04257175 (R/R AML, t8;21 and CD19 expression) CART-38 CD38 CAR NCT04351022 (R/R CD38+ AML) CD33CART CD33 CAR NCT03971799 (R/R AML) CART123 CD123 CAR NCT04678336 (AML in ≥2nd relapse, aged 1-29 years) CD123 CAR-T CD123 CAR NCT03796390 (R/R AM, with ≥80% CD123+) CD123-CAR T CD123 CAR NCT04318678 (R/R CD123+ AML, age ≤21 years) CLL-1 CAR T CLL-1 CAR NCT04219163 (R/R AML, with ≥30% CLL-1 blasts) Compound CAR-T CD33/CD123, CD123/CLL1 123 MB-102 CD123 CAR NCT02159495 (R/R CD123+ AML) MLM-CAR44.1 CD44v6 CAR NCT04097301 (R/R AML expressing CD44v6) PRGN-3006 CD33 CAR NCT03927261 (R/R AML, high risk MDS) TAA6 CD276 CAR NCT04692948 (R/R AML) UCART123 CD123 CAR NCT03190278 (R/R AML)Natural killer (NK) cell: 124 Alloreactive NK cell infusion Alloreactive NK cells NCT03955848 (AML in morphologic +/– cytogenetic or

molecular CR) CIML-NK cells Cytokine induced memory-like NK cells NCT01898793 (R/R AML, high risk AML not undergoing

transplant, or MDS with excess blasts) CYNK-001 CD56+/CD3– NK cells expanded from human placental

CD34+ cellsNCT04310592 (AML in morphologic CR with MRD)

FT516 Expresses novel CD16 Fc receptor NCT04023071 (R/R AML) FT538 Expresses novel CD16 Fc receptor, IL-15 receptor fusion,

CD38 knockoutNCT04614636 (R/R AML)

Haploidentical NK cells Haploidentical allogeneic donor derived in vitro activated NK cells

NCT04209712 (AML with MRD after 2 courses of standard chemotherapy)

mbIL21 expanded NK cells Membrane bound IL-21 expanded haploidentical NK cells NCT01787474 (R/R AML) NKX101 Allogeneic CAR NK cells targeting NKG2D ligands NCT04623944 (R/R AML or intermediate, high, or very high

risk MDS) oNKord Off-the-shelf, ex vivo cultured allogeneic NK cells NCT04632316 (AML in morphologic remission with MRD) UCB-NK cells +/- IL-2 Ex vivo NK cells generated from allogeneic umbilical cord

bloodNCT04347616 (stable or non-rapidly progressive AML ineligible for transplant)

ADCC=antibody dependent, cell mediated cytotoxicity; AML=acute myeloid leukemia; BiTE=bispecific T cell engager; Bst1=bone marrow stromal cell antigen 1; CAR-T=chimeric antigen receptor T cells; CD=cluster of differentiation; CLEC12A=C type lectin domain family 12 member A; CR=complete remission; DART=dual affinity re-targeting; FLT3=fms-like tyrosine kinase 3; G-CLAM=granulocyte colony stimulating factor, cladribine, cytarabine, mitoxantrone; HCT=hematopoietic cell transplantation; IL=interleukin; MDS=myelodysplastic syndrome; MMAE=monomethyl auristatin E; MPN=myeloproliferative neoplasm; MRD=minimal residual disease; PD-1=programmed cell death protein 1; PIF=primary induction failure; R/R=relapsed/refractory; TandAb=tandem diabodies; TriKE=tri-specific killer engager; WT1=Wilms’ tumor 1.

Table 2 | Continued

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

14 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

pediatric B lineage acute lymphoblastic leukemia and for relapsed or refractory diffuse large B cell lymphoma (DLBCL) in adults and axicabtagene ciloleucel (Yescarta) for the treatment of relapsed or refractory DLBCL and for relapsed or refractory primary mediastinal B cell lymphoma.121 172

However, despite this success in B lineage acute lymphoblastic leukemia and DLBCL, the development of CAR-T therapy for AML has been more challenging, partly because of the lack of a suitable target antigen. Given that AML target antigens such as CD33 and CD123 are often expressed not just on leukemia blasts but also on normal hematopoietic stem and progenitor cells,173-175 concern exists that eradication of all such antigen expressing cells would lead to ablation of the myeloid cell pool and marrow failure.176 Efforts to circumvent this hematologic toxicity include the identification of novel target antigens such as CLL-1, Lewis Y, FLT3, and CD44v6; compound CAR-T targeting both CD33/CD123 or CD123/CLL1, which could bypass the problem of loss of target antigen seen on leukemia cells at the time of relapse; and the incorporation of HSCT as a stem cell rescue or in combination with CAR-T and a genetically modified allograft.122 123 172 177-181

Nineteen CAR-T studies are actively recruiting for AML, as well as six additional trials that are either not yet recruiting or of unknown status, as recently reviewed.17 21 22 The FDA has granted orphan drug designation to the CD123 targeted CAR-T product MB-102 (clinicaltrials.com, NCT02159495),182 as well as to the PRGN-3006 UltraCAR-T (NCT03927261),183 for patients with AML.

Natural killer cell therapyNatural killer cells are effector lymphocytes that play a role in the innate immune response and possess direct cytotoxic and anti-tumor effects without the requirement for antigen specificity.123 Natural killer cell therapies can be derived from both autologous and allogeneic sources, including haplo-identical and in combination with HSCT,184 and can be used in the allogeneic setting without evidence of graft versus host disease.185 Efforts are under way to optimize natural killer cell sources (for example, haplo-identical peripheral blood, umbilical cord blood, pluripotent stem cells, and CAR expressing natural killer cells)186-188 and in vivo natural killer cell expansion and persistence, including lymphodepletion chemotherapy before natural killer cell infusion and co-administration of cytokines such as interleukin 2 and interleukin 15.185 189 190

A recently published first in human study assessed lymphodepleting chemotherapy followed by haplo-identical natural killer cell infusion with recombinant human interleukin 15, to potentiate in vivo natural killer cell activation.189 Recombinant human interleukin 15 was administered either intravenously or subcutaneously, in escalating doses. Assessment of response by day 42 showed a complete remission/complete remission with incomplete blood count recovery rate of 35% in both

cohorts (32% for intravenous recombinant human interleukin 15, 40% for subcutaneous recombinant human interleukin 15), with median durations of response of 107 days (intravenous) and 278 days (subcutaneous), and one year overall survival of 19% (intravenous) and 21% (subcutaneous).

More than a dozen actively recruiting trials of natural killer cell and CAR-natural killer cell therapies for AML are registered on clinicaltrials.gov. Active studies include natural killer therapies including haplo-identical and third party off-the-shelf products, as consolidation therapy or for relapsed/refractory disease, in tandem with haplo-identical or cord blood transplantation, as well as in combination with other agents (for example, recombinant human interleukin 2, immune checkpoint inhibitors).191 Strategies to enhance natural killer cell activity, as well as natural killer cell based cellular therapies under commercial development, have been recently reviewed.124

Antibody based therapyHigh expression of CD33 on AML blasts has also been exploited for antibody based therapy.173 Gemtuzumab ozogamicin (Mylotarg), an immunoconjugate combining anti-CD33 antibody with the toxin calicheamicin, was initially granted accelerated approval on the basis of a phase II study with a reported 30% response rate192  193; subsequently, gemtuzumab ozogamicin was discontinued given safety concerns and disappointing efficacy results.113-115 Recent studies showed high efficacy in APL and AML and efficacy when given in fractionated doses in combination with chemotherapy.116-118 192 The US FDA, followed by the European Medicines Agency, reapproved the use of gemtuzumab ozogamicin combined with daunorubicin and cytarabine for patients with newly diagnosed CD33 positive AML in 2017.117 119 120 194 195 The FDA has also approved gemtuzumab ozogamicin for use as a single agent in relapsed or refractory AML and in children.

Additional antibody based therapies include unconjugated monoclonal antibodies that can enhance antibody dependent, cell mediated cytotoxicity mediated by natural killer cells; multivalent antibodies (including bi-specific and tri-specific) designed to link tumor associated antigens with T cell or natural killer effector cells; and radioimmunoconjugates with a radionuclide attached to a monoclonal antibody, as recently summarized.196 197

GuidelinesEuropean, American, and WHO guidelines for the diagnosis and management of AML are referenced throughout this review. The European Leukemia Net (ELN), European Society for Medical Oncology (ESMO), and National Comprehensive Cancer Network (NCCN) all provide detailed guidelines for the clinical management of patients with AML, starting with initial investigation, through diagnosis and

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 15

management, to end of life care, and are important resources for the practicing clinician.18 19 198 Because the guidelines cover a vast number of patient specific situations, they do not permit direct comparison. The ELN risk stratification algorithm, which may need updating in the era of new therapies that may differentially affect prognosis, is still used by both ESMO and NCCN in their more recent iterations, with differences as noted earlier in the review.199 The NCCN guidelines are developed by members from US Cancer Centers that are designated “comprehensive cancer centers” on the basis of recent FDA approvals, presentations at scientific meetings and congresses, third party submissions, PubMed literature review, clinical data review, and institutional review. The recommendations are rated according to categories of evidence and consensus. Concerns have been raised about recommendations for newer oncologic therapies in the absence of robust randomized clinical trial data.200 More recently, several focused guidelines have been completed, identifying a specific sub-group of patients for whom recommendations might differ.201 202 The rapidity of changing knowledge about AML is likely to require incremental updates of guidelines with specific diagnostic, risk stratification, and management recommendations rather than complete revisions in the future.

Emerging treatmentsEmerging treatments are incorporated throughout the text in the relevant sections, as well as in table 2.

ConclusionsAML remains a complicated and difficult to treat disease, but major developments in our understanding of the molecular pathogenesis have led to improvements in diagnosis, monitoring, and treatment. Targeted therapies, immunotherapies, and novel agents are increasing the number of therapeutic choices for patients and providers. Studies on the

impact of factors such as cost, quality of life, time spent in treatment, and patients’ preferences will now be more relevant given the increasing number of decisions that must be made. Pre-leukemic conditions and genetic predisposition states are also increasingly recognized at a genomic level and raise the possibility of ultimately considering studies of leukemia prevention in the future. Providers should be prepared to refer patients and family members for appropriate testing and genetic counseling as needed. Ongoing cooperative efforts in basic science, translational studies, and clinical trials will continue to advance improvements for patients with AML.

We gratefully acknowledge Grover Bagby for his review of the manuscript, tables, and figures.Contributors: LFN and RJC developed the outline for this manuscript, did the literature review, and wrote all sections of the manuscript.Funding: LFN is supported by the National Institute of Child Health and Human Development (1K23 HD091369-01).Competing interests: We have read and understood the BMJ policy on declaration of interests and declare the following interests: none.Provenance and peer review: Commissioned; externally peer reviewed.Patient involvement: No patients were involved in the creation of this article.

1  National Cancer Institute Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Leukemia — Acute Myeloid Leukemia (AML). https://seer.cancer.gov/statfacts/html/amyl.html.

2  Eisfeld AK, Kohlschmidt J, Mrózek K, et al. Mutation patterns identify adult patients with de novo acute myeloid leukemia aged 60 years or older who respond favorably to standard chemotherapy: an analysis of Alliance studies. Leukemia 2018;32:1338-48. doi:10.1038/s41375-018-0068-2 

3  Ostronoff F, Othus M, Lazenby M, et al. Prognostic significance of NPM1 mutations in the absence of FLT3-internal tandem duplication in older patients with acute myeloid leukemia: a SWOG and UK National Cancer Research Institute/Medical Research Council report. J Clin Oncol 2015;33:1157-64. doi:10.1200/JCO.2014.58.0571 

4  Bhatt VR. Personalizing therapy for older adults with acute myeloid leukemia: Role of geriatric assessment and genetic profiling. Cancer Treat Rev 2019;75:52-61. doi:10.1016/j.ctrv.2019.04.001 

5  Klepin HD. Ready for prime time: role for geriatric assessment to improve quality of care in hematology practice. Blood 2019;134:2005-12. doi:10.1182/blood.2019001299 

GLOSSARY OF ABBREVIATIONS• AML—acute myeloid leukemia• APL—acute promyelocytic leukemia• BCL-2—B cell lymphoma-2• CAR-T—chimeric antigen receptor T cell• CHIP—clonal hematopoiesis of indeterminate

potential• CLL—chronic lymphocytic leukemia• DLBCL—diffuse large B cell lymphoma• ELN—European Leukemia-Net• FAB—French-American-British• HSCT—hematopoietic stem cell transplant• LDAC—low dose cytarabine• MCL-1—myeloid cell leukemia-1• MHC—major histocompatibility complex• MRC—myelodysplasia related changes• MRD—minimal/measurable residual disease• NCCN—National Comprehensive Cancer Network• NGS—next generation sequencing• qPCR—quantitative polymerase chain reaction

RESEARCH QUESTIONS• In what clinical situations should treatment of

acute myeloid leukemia (AML) be delayed to obtain additional prognostic information?

• How has risk stratification changed on the basis of availability of novel therapies?

• What is the most practical, replicable tool to measure residual AML after treatment?

• Can patients at the highest risk for therapy related AML be predicted before deciding therapy for their first malignancy, so that additional consideration could be given to choice of treatment?

○Would the risks and benefits of chemotherapy and/or radiation be reconsidered in light of an increased risk of therapy related AML?

• Do targetable mechanisms exist that might be used as therapies to prevent drivers of leukemogenesis in patients at risk (eg, post-chemotherapy/radiation, clonal hematopoiesis of indeterminate potential)?

• How should quality of life and patient reported outcomes influence treatment choices?

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

16 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

6  Klepin HD, Ritchie E, Major-Elechi B, et al. Geriatric assessment among older adults receiving intensive therapy for acute myeloid leukemia: Report of CALGB 361006 (Alliance). J Geriatr Oncol 2020;11:107-13. doi:10.1016/j.jgo.2019.10.002 

7  Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016;127:2391-405. doi:10.1182/blood-2016-03-643544 

8  Arber DA. The 2016 WHO classification of acute myeloid leukemia: What the practicing clinician needs to know. Semin Hematol 2019;56:90-5. doi:10.1053/j.seminhematol.2018.08.002 

9  Bullinger L, Döhner K, Döhner H. Genomics of Acute Myeloid Leukemia Diagnosis and Pathways. J Clin Oncol 2017;35:934-46. doi:10.1200/JCO.2016.71.2208 

10  Schuurhuis GJ, Heuser M, Freeman S, et al. Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party. Blood 2018;131:1275-91. doi:10.1182/blood-2017-09-801498 

11  Ravandi F, Walter RB, Freeman SD. Evaluating measurable residual disease in acute myeloid leukemia. Blood Adv 2018;2:1356-66. doi:10.1182/bloodadvances.2018016378 

12  LeBlanc TW, Erba HP. Shifting paradigms in the treatment of older adults with AML. Semin Hematol 2019;56:110-7. doi:10.1053/j.seminhematol.2019.02.002 

13  Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. Br J Haematol 1976;33:451-8. doi:10.1111/j.1365-2141.1976.tb03563.x 

14  Bennett JM, Catovsky D, Daniel MT, et al. Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-American-British Cooperative Group. Ann Intern Med 1985;103:620-5. doi:10.7326/0003-4819-103-4-620 

15  Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 2002;100:2292-302. doi:10.1182/blood-2002-04-1199 

16  Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 2009;114:937-51. doi:10.1182/blood-2009-03-209262 

17  Döhner H, Estey EH, Amadori S, et al, European LeukemiaNet. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 2010;115:453-74. doi:10.1182/blood-2009-07-235358 

18  Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017;129:424-47. doi:10.1182/blood-2016-08-733196 

19  National Comprehensive Cancer Network. NCCN Guidelines: Acute Myeloid Leukemia. 2021. https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1411.

20  Kern W, Haferlach T, Schoch C, et al. Early blast clearance by remission induction therapy is a major independent prognostic factor for both achievement of complete remission and long-term outcome in acute myeloid leukemia: data from the German AML Cooperative Group (AMLCG) 1992 Trial. Blood 2003;101:64-70. doi:10.1182/blood-2002-02-0532 

21  Bertoli S, Bories P, Béné MC, et al, Groupe Ouest-Est d’Etude des Leucémies Aiguës et Autres Maladies du Sang (GOELAMS). Prognostic impact of day 15 blast clearance in risk-adapted remission induction chemotherapy for younger patients with acute myeloid leukemia: long-term results of the multicenter prospective LAM-2001 trial by the GOELAMS study group. Haematologica 2014;99:46-53. doi:10.3324/haematol.2013.091819 

22  Morris TA, DeCastro CM, Diehl LF, et al. Re-induction therapy decisions based on day 14 bone marrow biopsy in acute myeloid leukemia. Leuk Res 2013;37:28-31. doi:10.1016/j.leukres.2012.09.016 

23  Yezefski T, Xie H, Walter R, et al. Value of routine ‘day 14’ marrow exam in newly diagnosed AML. Leukemia 2015;29:247-9. doi:10.1038/leu.2014.268 

24  Campuzano-Zuluaga G, Deutsch Y, Salzberg M, et al. Routine interim disease assessment in patients undergoing induction chemotherapy for acute myeloid leukemia: Can we do better?Am J Hematol 2016;91:277-82. doi:10.1002/ajh.24271 

25  Ollila TA, Olszewski AJ, Butera JN, Quesenberry MI, Quesenberry PJ, Reagan JL. Marrow Hypocellularity, But Not Residual Blast Count or Receipt of Reinduction Chemotherapy, Is Prognostic on Day-14 Assessment in Acute Myeloid Leukemia Patients With Morphologic Residual Disease. Clin Lymphoma Myeloma Leuk 2018;18:204-9. doi:10.1016/j.clml.2018.01.007 

26  Othus M, Mukherjee S, Sekeres MA, et al. Prediction of CR following a second course of ‘7+3’ in patients with newly diagnosed acute myeloid leukemia not in CR after a first course. Leukemia 2016;30:1779-80. doi:10.1038/leu.2016.48 

27  Jamy O, Bodine C, Sampat D, et al. Observation Versus Immediate Reinduction for Acute Myeloid Leukemia Patients With Indeterminate Day 14 Bone Marrow Results. Clin Lymphoma Myeloma Leuk 2020;20:31-8. doi:10.1016/j.clml.2019.09.613 

28  Hourigan CS, Gale RP, Gormley NJ, Ossenkoppele GJ, Walter RB. Measurable residual disease testing in acute myeloid leukaemia. Leukemia 2017;31:1482-90. doi:10.1038/leu.2017.113 

29  Chen X, Xie H, Wood BL, et al. Relation of clinical response and minimal residual disease and their prognostic impact on outcome in acute myeloid leukemia. J Clin Oncol 2015;33:1258-64. doi:10.1200/JCO.2014.58.3518 

30  Ivey A, Hills RK, Simpson MA, et al, UK National Cancer Research Institute AML Working Group. Assessment of Minimal Residual Disease in Standard-Risk AML. N Engl J Med 2016;374:422-33. doi:10.1056/NEJMoa1507471 

31  Jongen-Lavrencic M, Grob T, Hanekamp D, et al. Molecular Minimal Residual Disease in Acute Myeloid Leukemia. N Engl J Med 2018;378:1189-99. doi:10.1056/NEJMoa1716863 

32  Steensma DP, Ebert BL. Clonal Hematopoiesis after Induction Chemotherapy for Acute Myeloid Leukemia. N Engl J Med 2018;378:1244-5. doi:10.1056/NEJMe1802610 

33  Hasserjian RP, Steensma DP, Graubert TA, Ebert BL. Clonal hematopoiesis and measurable residual disease assessment in acute myeloid leukemia. Blood 2020;135:1729-38. doi:10.1182/blood.2019004770 

34  Walter RB, Gyurkocza B, Storer BE, et al. Comparison of minimal residual disease as outcome predictor for AML patients in first complete remission undergoing myeloablative or nonmyeloablative allogeneic hematopoietic cell transplantation. Leukemia 2015;29:137-44. doi:10.1038/leu.2014.173 

35  Kayser S, Benner A, Thiede C, et al. Pretransplant NPM1 MRD levels predict outcome after allogeneic hematopoietic stem cell transplantation in patients with acute myeloid leukemia. Blood Cancer J 2016;6:e449. doi:10.1038/bcj.2016.46 

36  Candoni A, De Marchi F, Zannier ME, et al. High prognostic value of pre-allogeneic stem cell transplantation minimal residual disease detection by WT1 gene expression in AML transplanted in cytologic complete remission. Leuk Res 2017;63:22-7. doi:10.1016/j.leukres.2017.10.010 

37  Araki D, Wood BL, Othus M, et al. Allogeneic Hematopoietic Cell Transplantation for Acute Myeloid Leukemia: Time to Move Toward a Minimal Residual Disease-Based Definition of Complete Remission?J Clin Oncol 2016;34:329-36. doi:10.1200/JCO.2015.63.3826 

38  Voso MT, Ottone T, Lavorgna S, et al. MRD in AML: The Role of New Techniques. Front Oncol 2019;9:655. doi:10.3389/fonc.2019.00655 

39  Ball B, Stein EM. Which are the most promising targets for minimal residual disease-directed therapy in acute myeloid leukemia prior to allogeneic stem cell transplant?Haematologica 2019;104:1521-31. doi:10.3324/haematol.2018.208587 

40  Percival MM, Estey EH. Current treatment strategies for measurable residual disease in patients with acute myeloid leukemia. Cancer 2019;125:3121-30. doi:10.1002/cncr.32354 

41  Drazer MW, Kadri S, Sukhanova M, et al. Prognostic tumor sequencing panels frequently identify germ line variants associated with hereditary hematopoietic malignancies. Blood Adv 2018;2:146-50. doi:10.1182/bloodadvances.2017013037 

42  University of Chicago Hematopoietic Malignancies Cancer Risk Team. How I diagnose and manage individuals at risk for inherited myeloid malignancies. Blood 2016;128:1800-13. doi:10.1182/blood-2016-05-670240 

43  Godley LA, Shimamura A. Genetic predisposition to hematologic malignancies: management and surveillance. Blood 2017;130:424-32. doi:10.1182/blood-2017-02-735290 

44  Hsu WL, Preston DL, Soda M, et al. The incidence of leukemia, lymphoma and multiple myeloma among atomic bomb survivors: 1950-2001. Radiat Res 2013;179:361-82. doi:10.1667/RR2892.1 

45  Gluzman D, Imamura N, Sklyarenko L, Nadgornaya V, Zavelevich M, Machilo V. Patterns of hematological malignancies in Chernobyl clean-up workers (1996-2005). Exp Oncol 2006;28:60-3.

46  Granfeldt Østgård LS, Medeiros BC, Sengeløv H, et al. Epidemiology and Clinical Significance of Secondary and Therapy-Related Acute Myeloid Leukemia: A National Population-Based Cohort Study. J Clin Oncol 2015;33:3641-9. doi:10.1200/JCO.2014.60.0890 

47  Weinberg OK, Arber DA. Acute Myeloid Leukemia With Myelodysplasia-Related Changes: A New Definition. Surg Pathol Clin 2010;3:1153-64. doi:10.1016/j.path.2010.09.012 

48  Weinberg OK, Seetharam M, Ren L, et al. Clinical characterization of acute myeloid leukemia with myelodysplasia-related changes as defined by the 2008 WHO classification system. Blood 2009;113:1906-8. doi:10.1182/blood-2008-10-182782 

49  Devillier R, Gelsi-Boyer V, Brecqueville M, et al. Acute myeloid leukemia with myelodysplasia-related changes are characterized by a specific molecular pattern with high frequency of ASXL1 mutations. Am J Hematol 2012;87:659-62. doi:10.1002/ajh.23211 

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 17

50  Devillier R, Mansat-De Mas V, Gelsi-Boyer V, et al. Role of ASXL1 and TP53 mutations in the molecular classification and prognosis of acute myeloid leukemias with myelodysplasia-related changes. Oncotarget 2015;6:8388-96. doi:10.18632/oncotarget.3460 

51  Xu XQ, Wang JM, Gao L, et al. Characteristics of acute myeloid leukemia with myelodysplasia-related changes: A retrospective analysis in a cohort of Chinese patients. Am J Hematol 2014;89:874-81. doi:10.1002/ajh.23772 

52  Porwit A, Saft L. The AML–MDS interface—leukemic transformation in myelodysplastic syndromes. J Hematop 2011;4:69-79. doi:10.1007/s12308-011-0088-6

53  Kuykendall A, Duployez N, Boissel N, Lancet JE, Welch JS. Acute Myeloid Leukemia: The Good, the Bad, and the Ugly. Am Soc Clin Oncol Educ Book 2018;38:555-73. doi:10.1200/EDBK_199519 

54  Hulegårdh E, Nilsson C, Lazarevic V, et al. Characterization and prognostic features of secondary acute myeloid leukemia in a population-based setting: a report from the Swedish Acute Leukemia Registry. Am J Hematol 2015;90:208-14. doi:10.1002/ajh.23908 

55  Walter MJ, Shen D, Ding L, et al. Clonal architecture of secondary acute myeloid leukemia. N Engl J Med 2012;366:1090-8. doi:10.1056/NEJMoa1106968 

56  Lindsley RC, Mar BG, Mazzola E, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 2015;125:1367-76. doi:10.1182/blood-2014-11-610543 

57  Fianchi L, Pagano L, Piciocchi A, et al. Characteristics and outcome of therapy-related myeloid neoplasms: Report from the Italian network on secondary leukemias. Am J Hematol 2015;90:E80-5. doi:10.1002/ajh.23966 

58  Klimek VM. Recent advances in the management of therapy-related myelodysplastic syndromes and acute myeloid leukemia. Curr Opin Hematol 2013;20:137-43. doi:10.1097/MOH.0b013e32835d82e6 

59  Morton LM, Dores GM, Tucker MA, et al. Evolving risk of therapy-related acute myeloid leukemia following cancer chemotherapy among adults in the United States, 1975-2008. Blood 2013;121:2996-3004. doi:10.1182/blood-2012-08-448068 

60  Shih AH, Chung SS, Dolezal EK, et al. Mutational analysis of therapy-related myelodysplastic syndromes and acute myelogenous leukemia. Haematologica 2013;98:908-12. doi:10.3324/haematol.2012.076729 

61  McNerney ME, Godley LA, Le Beau MM. Therapy-related myeloid neoplasms: when genetics and environment collide. Nat Rev Cancer 2017;17:513-27. doi:10.1038/nrc.2017.60 

62  Bhatia S. Therapy-related myelodysplasia and acute myeloid leukemia. Semin Oncol 2013;40:666-75. doi:10.1053/j.seminoncol.2013.09.013 

63  Godley LA, Larson RA. Therapy-related myeloid leukemia. Semin Oncol 2008;35:418-29. doi:10.1053/j.seminoncol.2008.04.012 

64  Offman J, Opelz G, Doehler B, et al. Defective DNA mismatch repair in acute myeloid leukemia/myelodysplastic syndrome after organ transplantation. Blood 2004;104:822-8. doi:10.1182/blood-2003-11-3938 

65  Morrison VA, Rai KR, Peterson BL, et al. Therapy-related myeloid leukemias are observed in patients with chronic lymphocytic leukemia after treatment with fludarabine and chlorambucil: results of an intergroup study, cancer and leukemia group B 9011. J Clin Oncol 2002;20:3878-84. doi:10.1200/JCO.2002.08.128 

66  Bowman RL, Busque L, Levine RL. Clonal Hematopoiesis and Evolution to Hematopoietic Malignancies. Cell Stem Cell 2018;22:157-70. doi:10.1016/j.stem.2018.01.011 

67  Wong TN, Ramsingh G, Young AL, et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature 2015;518:552-5. doi:10.1038/nature13968 

68  Link DC, Walter MJ. ‘CHIP’ping away at clonal hematopoiesis. Leukemia 2016;30:1633-5. doi:10.1038/leu.2016.130 

69  Stoddart A, Wang J, Fernald AA, Karrison T, Anastasi J, Le Beau MM. Cell intrinsic and extrinsic factors synergize in mice with haploinsufficiency for Tp53, and two human del(5q) genes, Egr1 and Apc. Blood 2014;123:228-38. doi:10.1182/blood-2013-05-506568 

70  Busque L, Mio R, Mattioli J, et al. Nonrandom X-inactivation patterns in normal females: lyonization ratios vary with age. Blood 1996;88:59-65. doi:10.1182/blood.V88.1.59.59 

71  Busque L, Patel JP, Figueroa ME, et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat Genet 2012;44:1179-81. doi:10.1038/ng.2413 

72  Shlush LI, Zandi S, Mitchell A, et al, HALT Pan-Leukemia Gene Panel Consortium. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 2014;506:328-33. doi:10.1038/nature13038 

73  Genovese G, Kähler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014;371:2477-87. doi:10.1056/NEJMoa1409405 

74  Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014;371:2488-98. doi:10.1056/NEJMoa1408617 

75  Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014;20:1472-8. doi:10.1038/nm.3733 

76  Jaiswal S, Natarajan P, Silver AJ, et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med 2017;377:111-21. doi:10.1056/NEJMoa1701719 

77  Ley TJ, Miller C, Ding L, et al, Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013;368:2059-74. doi:10.1056/NEJMoa1301689 

78  Young AL, Challen GA, Birmann BM, Druley TE. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun 2016;7:12484. doi:10.1038/ncomms12484 

79  Coombs CC, Zehir A, Devlin SM, et al. Therapy-Related Clonal Hematopoiesis in Patients with Non-hematologic Cancers Is Common and Associated with Adverse Clinical Outcomes. Cell Stem Cell 2017;21:374-382.e4. doi:10.1016/j.stem.2017.07.010 

80  Gillis NK, Ball M, Zhang Q, et al. Clonal haemopoiesis and therapy-related myeloid malignancies in elderly patients: a proof-of-concept, case-control study. Lancet Oncol 2017;18:112-21. doi:10.1016/S1470-2045(16)30627-1 

81  Takahashi K, Wang F, Kantarjian H, et al. Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: a case-control study. Lancet Oncol 2017;18:100-11. doi:10.1016/S1470-2045(16)30626-X 

82  Steensma DP, Bejar R, Jaiswal S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood 2015;126:9-16. doi:10.1182/blood-2015-03-631747

83  Molica M, Breccia M, Capria S, et al. The role of cladribine in acute myeloid leukemia: an old drug up to new tricks. Leuk Lymphoma 2020;61:536-45. doi:10.1080/10428194.2019.1672060 

84  Anžej Doma S, Škerget M, Pajič T, Sever M. Improved survival of AML patients by addition of cladribine to standard induction chemotherapy. Ann Hematol 2020;99:519-25. doi:10.1007/s00277-020-03923-9 

85  Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia. J Clin Oncol 2018;36:2684-92. doi:10.1200/JCO.2017.77.6112 

86  Zhou A, Han Q, Song H, Zi J, Ma J, Ge Z. Efficacy and toxicity of cladribine for the treatment of refractory acute myeloid leukemia: a meta-analysis. Drug Des Devel Ther 2019;13:1867-78. doi:10.2147/DDDT.S207425 

87  Thomas X, Elhamri M, Raffoux E, et al. Comparison of high-dose cytarabine and timed-sequential chemotherapy as consolidation for younger adults with AML in first remission: the ALFA-9802 study. Blood 2011;118:1754-62. doi:10.1182/blood-2011-04-349258 

88  Miyawaki S, Ohtake S, Fujisawa S, et al. A randomized comparison of 4 courses of standard-dose multiagent chemotherapy versus 3 courses of high-dose cytarabine alone in postremission therapy for acute myeloid leukemia in adults: the JALSG AML201 Study. Blood 2011;117:2366-72. doi:10.1182/blood-2010-07-295279 

89  Schaich M, Röllig C, Soucek S, et al. Cytarabine dose of 36 g/m2 compared with 12 g/m2 within first consolidation in acute myeloid leukemia: results of patients enrolled onto the prospective randomized AML96 study. J Clin Oncol 2011;29:2696-702. doi:10.1200/JCO.2010.33.7303 

90  Loke J, Lowe DM, Miller LJ, et al, National Cancer Research Institute Acute Myeloid Leukaemia Working Party on Supportive Care/Transfusion. Supportive care in the management of patients with acute myeloid leukaemia: where are the research needs?Br J Haematol 2020;190:311-3. doi:10.1111/bjh.16708 

91  Gurion R, Belnik-Plitman Y, Gafter-Gvili A, et al. Colony-stimulating factors for prevention and treatment of infectious complications in patients with acute myelogenous leukemia. Cochrane Database Syst Rev 2012;(6):CD008238. doi:10.1002/14651858.CD008238.pub3 

92  Stanworth SJ, Estcourt LJ, Powter G, et al, TOPPS Investigators. A no-prophylaxis platelet-transfusion strategy for hematologic cancers. N Engl J Med 2013;368:1771-80. doi:10.1056/NEJMoa1212772 

93  Wandt H, Schaefer-Eckart K, Wendelin K, et al, Study Alliance Leukemia. Therapeutic platelet transfusion versus routine prophylactic transfusion in patients with haematological malignancies: an open-label, multicentre, randomised study. Lancet 2012;380:1309-16. doi:10.1016/S0140-6736(12)60689-8 

94  Cornely OA, Maertens J, Winston DJ, et al. Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia. N Engl J Med 2007;356:348-59. doi:10.1056/NEJMoa061094 

95  Gafter-Gvili A, Fraser A, Paul M, et al. Antibiotic prophylaxis for bacterial infections in afebrile neutropenic patients following chemotherapy. Cochrane Database Syst Rev 2012;1:CD004386. doi:10.1002/14651858.CD004386.pub3 

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

18 doi: 10.1136/bmj.n2026 | BMJ 2021;375:n2026 | the bmj

96  Taplitz RA, Kennedy EB, Bow EJ, et al. Antimicrobial Prophylaxis for Adult Patients With Cancer-Related Immunosuppression: ASCO and IDSA Clinical Practice Guideline Update. J Clin Oncol 2018;36:3043-54. doi:10.1200/JCO.18.00374 

97  Fernandez HF, Sun Z, Yao X, et al. Anthracycline dose intensification in acute myeloid leukemia. N Engl J Med 2009;361:1249-59. doi:10.1056/NEJMoa0904544 

98  Röllig C, Bornhäuser M, Thiede C, et al. Long-term prognosis of acute myeloid leukemia according to the new genetic risk classification of the European LeukemiaNet recommendations: evaluation of the proposed reporting system. J Clin Oncol 2011;29:2758-65. doi:10.1200/JCO.2010.32.8500 

99  Döhner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med 2015;373:1136-52. doi:10.1056/NEJMra1406184 

100  Löwenberg B, Ossenkoppele GJ, van Putten W, et al, Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON), German AML Study Group (AMLSG), Swiss Group for Clinical Cancer Research (SAKK) Collaborative Group. High-dose daunorubicin in older patients with acute myeloid leukemia. N Engl J Med 2009;361:1235-48. doi:10.1056/NEJMoa0901409 

101  Löwenberg B. Sense and nonsense of high-dose cytarabine for acute myeloid leukemia. Blood 2013;121:26-8. doi:10.1182/blood-2012-07-444851 

102  Burnett AK, Russell NH, Hills RK, et al. Optimization of chemotherapy for younger patients with acute myeloid leukemia: results of the medical research council AML15 trial. J Clin Oncol 2013;31:3360-8. doi:10.1200/JCO.2012.47.4874 

103  Becker PS, Kantarjian HM, Appelbaum FR, et al. Clofarabine with high dose cytarabine and granulocyte colony-stimulating factor (G-CSF) priming for relapsed and refractory acute myeloid leukaemia. Br J Haematol 2011;155:182-9. doi:10.1111/j.1365-2141.2011.08831.x 

104  Burnett AK, Russell NH, Hills RK, et al. A comparison of clofarabine with ara-C, each in combination with daunorubicin as induction treatment in older patients with acute myeloid leukaemia. Leukemia 2017;31:310-7. doi:10.1038/leu.2016.225 

105  Halpern AB, Othus M, Huebner EM, et al. Phase 1/2 trial of GCLAM with dose-escalated mitoxantrone for newly diagnosed AML or other high-grade myeloid neoplasms. Leukemia 2018;32:2352-62. doi:10.1038/s41375-018-0135-8 

106  Holowiecki J, Grosicki S, Giebel S, et al. Cladribine, but not fludarabine, added to daunorubicin and cytarabine during induction prolongs survival of patients with acute myeloid leukemia: a multicenter, randomized phase III study. J Clin Oncol 2012;30:2441-8. doi:10.1200/JCO.2011.37.1286 

107  El-Jawahri A, Nelson-Lowe M, VanDusen H, et al. Patient-Clinician Discordance in Perceptions of Treatment Risks and Benefits in Older Patients with Acute Myeloid Leukemia. Oncologist 2019;24:247-54. doi:10.1634/theoncologist.2018-0317 

108  Kayastha N, Wolf SP, Locke SC, Samsa GP, El-Jawahri A, LeBlanc TW. The impact of remission status on patients’ experiences with acute myeloid leukemia (AML): an exploratory analysis of longitudinal patient-reported outcomes data. Support Care Cancer 2018;26:1437-45.

109  El-Jawahri AR, Abel GA, Steensma DP, et al. Health care utilization and end-of-life care for older patients with acute myeloid leukemia. Cancer 2015;121:2840-8. doi:10.1002/cncr.29430 

110  Carter JL, Hege K, Yang J, et al. Targeting multiple signaling pathways: the new approach to acute myeloid leukemia therapy. Signal Transduct Target Ther 2020;5:288. doi:10.1038/s41392-020-00361-x 

111  Vasu S, He S, Cheney C, et al. Decitabine enhances anti-CD33 monoclonal antibody BI 836858-mediated natural killer ADCC against AML blasts. Blood 2016;127:2879-89. doi:10.1182/blood-2015-11-680546 

112  Dao T, Pankov D, Scott A, et al. Therapeutic bispecific T-cell engager antibody targeting the intracellular oncoprotein WT1. Nat Biotechnol 2015;33:1079-86. doi:10.1038/nbt.3349 

113  Rowe JM, Löwenberg B. Gemtuzumab ozogamicin in acute myeloid leukemia: a remarkable saga about an active drug. Blood 2013;121:4838-41. doi:10.1182/blood-2013-03-490482 

114  Rajvanshi P, Shulman HM, Sievers EL, McDonald GB. Hepatic sinusoidal obstruction after gemtuzumab ozogamicin (Mylotarg) therapy. Blood 2002;99:2310-4. doi:10.1182/blood.V99.7.2310 

115  Petersdorf SH, Kopecky KJ, Slovak M, et al. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. Blood 2013;121:4854-60. doi:10.1182/blood-2013-01-466706 

116  Lo-Coco F, Cimino G, Breccia M, et al. Gemtuzumab ozogamicin (Mylotarg) as a single agent for molecularly relapsed acute promyelocytic leukemia. Blood 2004;104:1995-9. doi:10.1182/blood-2004-04-1550 

117  Castaigne S, Pautas C, Terré C, et al, Acute Leukemia French Association. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a

randomised, open-label, phase 3 study. Lancet 2012;379:1508-16. doi:10.1016/S0140-6736(12)60485-1 

118  Taksin AL, Legrand O, Raffoux E, et al. High efficacy and safety profile of fractionated doses of Mylotarg as induction therapy in patients with relapsed acute myeloblastic leukemia: a prospective study of the alfa group. Leukemia 2007;21:66-71. doi:10.1038/sj.leu.2404434 

119  US Food and Drug Administration. FDA approves Mylotarg for treatment of acute myeloid leukemia. 2017. https://www.fda.gov/news-events/press-announcements/fda-approves-mylotarg-treatment-acute-myeloid-leukemia.

120  European Medicines Agency. Mylotrag. 2020. https://www.ema.europa.eu/en/medicines/human/EPAR/mylotarg-0.

121  Cummins KD, Gill S, et al. Will CAR T cell therapy have a role in AML? Promises and pitfalls. Semin Hematol 2019;56:155-63. doi:10.1053/j.seminhematol.2018.08.008 

122  Hofmann S, Schubert ML, Wang L. Chimeric Antigen Receptor (CAR) T Cell Therapy in Acute Myeloid Leukemia (AML). J Clin Med 2019;8:200. doi:10.3390/jcm8020200 

123  Shang Y, Zhou F. Current Advances in Immunotherapy for Acute Leukemia: An Overview of Antibody, Chimeric Antigen Receptor, Immune Checkpoint, and Natural Killer. Front Oncol 2019;9:917. doi:10.3389/fonc.2019.00917 

124  Cooley S, Parham P, Miller JS. Strategies to activate NK cells to prevent relapse and induce remission following hematopoietic stem cell transplantation. Blood 2018;131:1053-62. doi:10.1182/blood-2017-08-752170 

125  Stomper J, Lübbert M. Can we predict responsiveness to hypomethylating agents in AML?Semin Hematol 2019;56:118-24. doi:10.1053/j.seminhematol.2019.02.001 

126  Kadia TM, Jain P, Ravandi F, et al. TP53 mutations in newly diagnosed acute myeloid leukemia: Clinicomolecular characteristics, response to therapy, and outcomes. Cancer 2016;122:3484-91. doi:10.1002/cncr.30203 

127  Bowen D, Groves MJ, Burnett AK, et al. TP53 gene mutation is frequent in patients with acute myeloid leukemia and complex karyotype, and is associated with very poor prognosis. Leukemia 2009;23:203-6. doi:10.1038/leu.2008.173 

128  Welch JS, Petti AA, Miller CA, et al. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes. N Engl J Med 2016;375:2023-36. doi:10.1056/NEJMoa1605949 

129  Short NJ, Kantarjian HM, Loghavi S, et al. Treatment with a 5-day versus a 10-day schedule of decitabine in older patients with newly diagnosed acute myeloid leukaemia: a randomised phase 2 trial. Lancet Haematol 2019;6:e29-37. doi:10.1016/S2352-3026(18)30182-0 

130  Hunter AM, Sallman DA. Current status and new treatment approaches in TP53 mutated AML. Best Pract Res Clin Haematol 2019;32:134-44. doi:10.1016/j.beha.2019.05.004 

131  Othus M, Sekeres MA, Nand S, et al. Relative survival following response to 7 + 3 versus azacytidine is similar in acute myeloid leukemia and high-risk myelodysplastic syndromes: an analysis of four SWOG studies. Leukemia 2019;33:371-8. doi:10.1038/s41375-018-0275-x 

132  Estey E, Gale RP. How good are we at predicting the fate of someone with acute myeloid leukaemia?Leukemia 2017;31:1255-8. doi:10.1038/leu.2017.56 

133  Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood 2015;126:291-9. doi:10.1182/blood-2015-01-621664 

134  Wei AH, Strickland SAJr, Hou JZ, et al. Venetoclax Combined With Low-Dose Cytarabine for Previously Untreated Patients With Acute Myeloid Leukemia: Results From a Phase Ib/II Study. J Clin Oncol 2019;37:1277-84. doi:10.1200/JCO.18.01600 

135  Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia 2019;33:379-89. doi:10.1038/s41375-018-0312-9 

136  Vogler M, Walter HS, Dyer MJS. Targeting anti-apoptotic BCL2 family proteins in haematological malignancies - from pathogenesis to treatment. Br J Haematol 2017;178:364-79. doi:10.1111/bjh.14684 

137  Stilgenbauer S, Eichhorst B, Schetelig J, et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study. Lancet Oncol 2016;17:768-78. doi:10.1016/S1470-2045(16)30019-5 

138  Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and Biological Correlates of Response in a Phase II Study of Venetoclax Monotherapy in Patients with Acute Myelogenous Leukemia. Cancer Discov 2016;6:1106-17. doi:10.1158/2159-8290.CD-16-0313 

139  Pollyea DA, Amaya M, Strati P, Konopleva MY. Venetoclax for AML: changing the treatment paradigm. Blood Adv 2019;3:4326-35. doi:10.1182/bloodadvances.2019000937 

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

STATE OF THE ART REVIEW

the bmj | BMJ 2021;375:n2026 | doi: 10.1136/bmj.n2026 19

140  DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 2019;133:7-17. doi:10.1182/blood-2018-08-868752 

141  DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N Engl J Med 2020;383:617-29. doi:10.1056/NEJMoa2012971 

142  Winters AC, Gutman JA, Purev E, et al. Real-world experience of venetoclax with azacitidine for untreated patients with acute myeloid leukemia. Blood Adv 2019;3:2911-9. doi:10.1182/bloodadvances.2019000243 

143  DiNardo CD, Tiong IS, Quaglieri A, et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood 2020;135:791-803. doi:10.1182/blood.2019003988 

144  Jonas BA, Pollyea DA. How we use venetoclax with hypomethylating agents for the treatment of newly diagnosed patients with acute myeloid leukemia. Leukemia 2019;33:2795-804. doi:10.1038/s41375-019-0612-8 

145  DiNardo CD, Wei AH. How I treat acute myeloid leukemia in the era of new drugs. Blood 2020;135:85-96. doi:10.1182/blood.2019001239 

146  Wei AH, Roberts AW, Spencer A, et al. Targeting MCL-1 in hematologic malignancies: Rationale and progress. Blood Rev 2020;44:100672. doi:10.1016/j.blre.2020.100672 

147  Sakaguchi M, Yamaguchi H, Najima Y, et al. Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in acute myeloid leukemia. Blood Adv 2018;2:2744-54. doi:10.1182/bloodadvances.2018020305 

148  Boddu P, Kantarjian H, Borthakur G, et al. Co-occurrence of FLT3-TKD and NPM1 mutations defines a highly favorable prognostic AML group. Blood Adv 2017;1:1546-50. doi:10.1182/bloodadvances.2017009019 

149  Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N Engl J Med 2017;377:454-64. doi:10.1056/NEJMoa1614359 

150  Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N Engl J Med 2019;381:1728-40. doi:10.1056/NEJMoa1902688 

151  Daver N, Schlenk RF, Russell NH, Levis MJ. Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia 2019;33:299-312. doi:10.1038/s41375-018-0357-9 

152  Majothi S, Adams D, Loke J, Stevens SP, Wheatley K, Wilson JS. FLT3 inhibitors in acute myeloid leukaemia: assessment of clinical effectiveness, adverse events and future research-a systematic review and meta-analysis. Syst Rev 2020;9:285. doi:10.1186/s13643-020-01540-1 

153  Pratz KW, Levis M. How I treat FLT3-mutated AML. Blood 2017;129:565-71. doi:10.1182/blood-2016-09-693648 

154  Megías-Vericat JE, Ballesta-López O, Barragán E, Martínez-Cuadrón D, Montesinos P. Tyrosine kinase inhibitors for acute myeloid leukemia: A step toward disease control?Blood Rev 2020;44:100675. doi:10.1016/j.blre.2020.100675 

155  Fathi AT, DiNardo CD, Kline I, et al, AG221-C-001 Study Investigators. Differentiation Syndrome Associated With Enasidenib, a Selective Inhibitor of Mutant Isocitrate Dehydrogenase 2: Analysis of a Phase 1/2 Study. JAMA Oncol 2018;4:1106-10. doi:10.1001/jamaoncol.2017.4695 

156  DiNardo CD. Ivosidenib in IDH1-Mutated Acute Myeloid Leukemia. N Engl J Med 2018;379:1186. doi:10.1056/NEJMc1809507 

157  Medeiros BC, Fathi AT, DiNardo CD, Pollyea DA, Chan SM, Swords R. Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia 2017;31:272-81. doi:10.1038/leu.2016.275 

158  Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med 2016;374:2209-21. doi:10.1056/NEJMoa1516192 

159  Rücker FG, Schlenk RF, Bullinger L, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood 2012;119:2114-21. doi:10.1182/blood-2011-08-375758 

160  Sallman DA, DeZern AE, Garcia-Manero G, et al. Eprenetapopt (APR-246) and Azacitidine in TP53-Mutant Myelodysplastic Syndromes. J Clin Oncol 2021;39:1584-94. doi:10.1200/JCO.20.02341 

161  Cornelissen JJ, Blaise D. Hematopoietic stem cell transplantation for patients with AML in first complete remission. Blood 2016;127:62-70. doi:10.1182/blood-2015-07-604546 

162  Ruggeri A, Labopin M, Sanz G, et al, Eurocord, Cord Blood Committee of Cellular Therapy and Immunobiology working party-EBMT, ALWP-EBMT study. Comparison of outcomes after unrelated cord blood and unmanipulated haploidentical stem cell transplantation in adults with acute leukemia. Leukemia 2015;29:1891-900. doi:10.1038/leu.2015.98 

163  Battipaglia G, Labopin M, Kröger N, et al. Posttransplant cyclophosphamide vs antithymocyte globulin in HLA-mismatched unrelated donor transplantation. Blood 2019;134:892-9. doi:10.1182/blood.2019000487 

164  Toffalori C, Zito L, Gambacorta V, et al. Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation. Nat Med 2019;25:603-11. doi:10.1038/s41591-019-0400-z 

165  Christopher MJ, Petti AA, Rettig MP, et al. Immune Escape of Relapsed AML Cells after Allogeneic Transplantation. N Engl J Med 2018;379:2330-41. doi:10.1056/NEJMoa1808777 

166  Liu L, Chang YJ, Xu LP, et al. Reversal of T Cell Exhaustion by the First Donor Lymphocyte Infusion Is Associated with the Persistently Effective Antileukemic Responses in Patients with Relapsed AML after Allo-HSCT. Biol Blood Marrow Transplant 2018;24:1350-9. doi:10.1016/j.bbmt.2018.03.030 

167  de Lima M, Giralt S, Thall PF, et al. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogenous leukemia or myelodysplastic syndrome: a dose and schedule finding study. Cancer 2010;116:5420-31. doi:10.1002/cncr.25500 

168  Oran B, de Lima M, Garcia-Manero G, et al. A phase 3 randomized study of 5-azacitidine maintenance vs observation after transplant in high-risk AML and MDS patients. Blood Adv 2020;4:5580-8. doi:10.1182/bloodadvances.2020002544 

169  Feins S, Kong W, Williams EF, Milone MC, Fraietta JA. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol 2019;94(S1):S3-9. doi:10.1002/ajh.25418 

170  June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science 2018;359:1361-5. doi:10.1126/science.aar6711 

171  Garrido F, Aptsiauri N, Doorduijn EM, Garcia Lora AM, van Hall T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr Opin Immunol 2016;39:44-51. doi:10.1016/j.coi.2015.12.007 

172  Cummins KD, Gill S. Chimeric antigen receptor T-cell therapy for acute myeloid leukemia: how close to reality? Haematologica 2019;104:1302-8. doi:10.3324/haematol.2018.208751 

173  Ehninger A, Kramer M, Röllig C, et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J 2014;4:e218. doi:10.1038/bcj.2014.39 

174  Testa U, Pelosi E, Frankel A. CD 123 is a membrane biomarker and a therapeutic target in hematologic malignancies. Biomark Res 2014;2:4. doi:10.1186/2050-7771-2-4 

175  ten Cate B, de Bruyn M, Wei Y, Bremer E, Helfrich W. Targeted elimination of leukemia stem cells; a new therapeutic approach in hemato-oncology. Curr Drug Targets 2010;11:95-110. doi:10.2174/138945010790031063 

176  Gill S, Tasian SK, Ruella M, et al. Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood 2014;123:2343-54. doi:10.1182/blood-2013-09-529537 

177  Ritchie DS, Neeson PJ, Khot A, et al. Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Mol Ther 2013;21:2122-9. doi:10.1038/mt.2013.154 

178  Wang Y, Xu Y, Li S, et al. Targeting FLT3 in acute myeloid leukemia using ligand-based chimeric antigen receptor-engineered T cells. J Hematol Oncol 2018;11:60. doi:10.1186/s13045-018-0603-7 

179  Casucci M, Nicolis di Robilant B, Falcone L, et al. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood 2013;122:3461-72. doi:10.1182/blood-2013-04-493361 

180  Kim MY, Yu KR, Kenderian SS, et al. Genetic Inactivation of CD33 in Hematopoietic Stem Cells to Enable CAR T Cell Immunotherapy for Acute Myeloid Leukemia. Cell 2018;173:1439-1453.e19. doi:10.1016/j.cell.2018.05.013 

181  Borot F, Wang H, Ma Y, et al. Gene-edited stem cells enable CD33-directed immune therapy for myeloid malignancies. Proc Natl Acad Sci U S A 2019;116:11978-87. doi:10.1073/pnas.1819992116 

182  Mustang Bio. FDA Clears IND for Mustang Bio’s MB-102 (CD123 CAR T). 2019. https://www.globenewswire.com/news-release/2019/08/05/1896824/0/en/FDA-Clears-IND-for-Mustang-Bio-s-MB-102-CD123-CAR-T.html.

183  Precigen. Precigen Receives FDA Orphan Drug Designation for PRGN-3006 UltraCAR-T™ in Patients with Acute Myeloid Leukemia (AML). 2020. https://www.prnewswire.com/news-releases/precigen-receives-fda-orphan-drug-designation-for-prgn-3006-ultracar-t-in-patients-with-acute-myeloid-leukemia-aml-300981401.html.

184  Rezvani K. Adoptive cell therapy using engineered natural killer cells. Bone Marrow Transplant 2019;54(Suppl 2):785-8. doi:10.1038/s41409-019-0601-6 

185  Woan KV, Miller JS. Harnessing Natural Killer Cell Antitumor Immunity: From the Bench to Bedside. Cancer Immunol Res 2019;7:1742-7. doi:10.1158/2326-6066.CIR-19-0404 

186  Spanholtz J, Tordoir M, Eissens D, et al. High log-scale expansion of functional human natural killer cells from umbilical cord blood CD34-positive cells for adoptive cancer immunotherapy. PLoS One 2010;5:e9221. doi:10.1371/journal.pone.0009221 

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from

No commercial reuse: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe

STATE OF THE ART REVIEW

187  Knorr DA, Ni Z, Hermanson D, et al. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl Med 2013;2:274-83. doi:10.5966/sctm.2012-0084 

188  Li Y, Hermanson DL, Moriarity BS, Kaufman DS. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity. Cell Stem Cell 2018;23:181-192.e5. doi:10.1016/j.stem.2018.06.002 

189  Cooley S, He F, Bachanova V, et al. First-in-human trial of rhIL-15 and haploidentical natural killer cell therapy for advanced acute myeloid leukemia. Blood Adv 2019;3:1970-80. doi:10.1182/bloodadvances.2018028332 

190  Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005;105:3051-7. doi:10.1182/blood-2004-07-2974 

191  ClinicalTrials.gov. Search. https://clinicaltrials.gov/ct2/results?recrs= ab&cond=AML&term=NK+cell&cntry=&state=&city=&dist=.

192  Sievers EL, Larson RA, Stadtmauer EA, et al, Mylotarg Study Group. Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. J Clin Oncol 2001;19:3244-54. doi:10.1200/JCO.2001.19.13.3244 

193  Bross PF, Beitz J, Chen G, et al. Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia. Clin Cancer Res 2001;7:1490-6.

194  Lambert J, Pautas C, Terré C, et al. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: final efficacy and safety updates from the open-label, phase III ALFA-0701 trial. Haematologica 2019;104:113-9. doi:10.3324/haematol.2018.188888 

195  Hitzler J, Estey E. Gemtuzumab ozogamicin in acute myeloid leukemia: act 2, with perhaps more to come. Haematologica 2019;104:7-9. doi:10.3324/haematol.2018.205948 

196  Walter RB. Investigational CD33-targeted therapeutics for acute myeloid leukemia. Expert Opin Investig Drugs 2018;27:339-48. doi:10.1080/13543784.2018.1452911 

197  Williams BA, Law A, Hunyadkurti J, Desilets S, Leyton JV, Keating A. Antibody Therapies for Acute Myeloid Leukemia: Unconjugated, Toxin-Conjugated, Radio-Conjugated and Multivalent Formats. J Clin Med 2019;8:1261. doi:10.3390/jcm8081261 

198  Heuser M, Ofran Y, Boissel N, et al, ESMO Guidelines Committee. Electronic address: [email protected]. Acute myeloid leukaemia in adult patients: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020;31:697-712. doi:10.1016/j.annonc.2020.02.018 

199  Estey EH. Acute myeloid leukemia: 2021 update on risk-stratification and management. Am J Hematol 2020;95:1368-98. doi:10.1002/ajh.25975 

200  Wagner J, Marquart J, Ruby J, et al. Frequency and level of evidence used in recommendations by the National Comprehensive Cancer Network guidelines beyond approvals of the US Food and Drug Administration: retrospective observational study. BMJ 2018;360:k668. doi:10.1136/bmj.k668 

201  Sekeres MA, Guyatt G, Abel G, et al. American Society of Hematology 2020 guidelines for treating newly diagnosed acute myeloid leukemia in older adults. Blood Adv 2020;4:3528-49. doi:10.1182/bloodadvances.2020001920 

202  Maertens JA, Girmenia C, Brüggemann RJ, et al, European Conference on Infections in Leukaemia (ECIL), a joint venture of the European Group for Blood and Marrow Transplantation (EBMT), the European Organization for Research and Treatment of Cancer (EORTC), the Immunocompromised Host Society (ICHS) and European Conference on Infections in Leukaemia (ECIL), a joint venture of the European Group for Blood and Marrow Transplantation (EBMT), the European Organization for Research and Treatment of Cancer (EORTC), the Immunocompromised Host Society (ICHS) and the European LeukemiaNet (ELN). European guidelines for primary antifungal prophylaxis in adult haematology patients: summary of the updated recommendations from the European Conference on Infections in Leukaemia. J Antimicrob Chemother 2018;73:3221-30. doi:10.1093/jac/dky286

on 28 January 2022 by guest. Protected by copyright.

http://ww

w.bm

j.com/

BM

J: first published as 10.1136/bmj.n2026 on 6 O

ctober 2021. Dow

nloaded from