Next Article in Journal
Young Myeloma Patients: A Systematic Review of Manifestations and Outcomes
Next Article in Special Issue
Management of Patients with Lower-Risk Myelodysplastic Neoplasms (MDS)
Previous Article in Journal
Multi-Institutional Patterns of Use of Tumor-Treating Fields for Patients with Malignant Pleural Mesothelioma
Previous Article in Special Issue
A Systematic Review Assessing the Impact of Vitamin D Levels on Adult Patients with Lymphoid Malignancies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Tools for Diagnosis and Monitoring of AML

1
Hematopathology Section, Pathology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
2
Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
3
Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
4
Hematology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2023, 30(6), 5201-5213; https://doi.org/10.3390/curroncol30060395
Submission received: 28 April 2023 / Revised: 11 May 2023 / Accepted: 18 May 2023 / Published: 23 May 2023
(This article belongs to the Special Issue Haematological Neoplasms: Diagnosis and Management)

Abstract

:
In recent years, major advances in the understanding of acute myeloid leukemia (AML) pathogenesis, together with technological progress, have led us into a new era in the diagnosis and follow-up of patients with AML. A combination of immunophenotyping, cytogenetic and molecular studies are required for AML diagnosis, including the use of next-generation sequencing (NGS) gene panels to screen all genetic alterations with diagnostic, prognostic and/or therapeutic value. Regarding AML monitoring, multiparametric flow cytometry and quantitative PCR/RT-PCR are currently the most implemented methodologies for measurable residual disease (MRD) evaluation. Given the limitations of these techniques, there is an urgent need to incorporate new tools for MRD monitoring, such as NGS and digital PCR. This review aims to provide an overview of the different technologies used for AML diagnosis and MRD monitoring and to highlight the limitations and challenges of current versus emerging tools.

1. Introduction

Acute myeloid leukemia (AML) is characterized by a high biological heterogeneity both at diagnosis and during disease evolution. In this context, an exhaustive characterization of immunophenotypic and molecular profiles is required to guide the clinical management of AML patients. In recent years, growing evidence has shown the prominent role of cytogenetic and mutational data in AML and, accordingly, the recently updated World Health Organization (WHO) [1] and International Consensus Classification (ICC) [2] classifications of myeloid neoplasms as well as the European LeukemiaNet (ELN) recommendations [3] have incorporated genetics into their diagnostic, prognostic and therapeutic algorithms. In this scenario, a combination of immunophenotyping and genetic studies are required for AML diagnosis and follow-up. Measurable residual disease (MRD) has strong prognostic and predictive value in AML, and its accurate monitoring is crucial for disease management. The plethora of aberrant phenotypic and molecular characteristics of AML cells represents a chance to track this disease but, at the same time, constitutes a major challenge for hematopathology laboratories. In this review, we discuss the most relevant technologies based on flow cytometry and molecular biology used for AML diagnosis and MRD monitoring in the context of recently updated diagnostic and prognostic guidelines.

2. Diagnosis

2.1. Flow Cytometry for Lineage Assessment

Multiparametric flow cytometry (MFC) consists of the recognition of cells based on antigen detection through a combination of fluorochrome-labeled monoclonal antibodies, which allows phenotypic characterization and the quantification of a specific cell population.
When acute leukemia is suspected, morphologic and immunophenotypic examinations of the bone marrow and peripheral blood are the quickest techniques to confirm the diagnosis (Figure 1). The latter is also essential when assigning the leukemic lineage: a crucial step that determines further diagnostic tests and therapeutic strategies. Lineage-assigning antigens are shown in Table 1. Myeloperoxidase (MPO) expression is the hallmark of myeloid commitment, though it is not always present. AML with minimal differentiation is defined by an absence of lineage-assigning antigens in combination with the expression of at least two other myeloid-related antigens (e.g., CD13, CD33, CD117). Monocytic AML often loses MPO expression and is characterized by specific markers such as CD11c, CD14, CD64 and lysozyme.
AML differentiation, maturation stage and the aberrant co-expression of lymphoid markers can be only addressed with a broad panel of antibodies. Morphologic and immunophenotypic criteria for cases without recurrent genetic abnormalities have been updated in the latest WHO classification [1]. Even when this information lacks its independent prognostic impact, it often correlates with particular genetic lesions. For example, acute promyelocytic leukemia (APL) exhibits a characteristic immunophenotype (CD34−, HLA-DR−, CD117+, CD15−) that can prompt PML::RARA gene fusion evaluation and consequent management until the molecular result is available. Another example is the association between the co-expression of CD25, CD123 and CD99 on myeloid blast and FLT3 mutations [4].
The Euroflow Consortium has developed, through serial testing, an acute leukemia orientation tube (ALOT), which includes most of the lineage-defining markers, and a panel for AML that covers the different myeloid lineages and most frequent lymphoid aberrancies [5]. Moreover, the standardization of MFC and the building of reference databases with normal and acute leukemia samples allows for automated gating identification of leukocyte subpopulations and classification of blast cells [6,7].
The use of a comprehensive panel of antibodies also allows for the recognition of leukemia-associated immunophenotype (LAIP) patterns that can be of interest during follow-up. Finally, new therapies targeting the surface antigens of myeloid blasts are under development. In the future, flow cytometry could be useful when identifying potential candidates for treatment against not-universally expressed antigens, as could be chimeric antigen receptor therapy against CD123 [8].

2.2. Molecular Biology

Over the last decade, genomic studies based on Next-Generation Sequencing (NGS) have dissected the molecular profile of AML, with the description of new mutations, copy number variations and recurrent fusion genes [9,10]. The growth in molecular knowledge has prompted the update of both diagnostic and management recommendations for AML patients. The recently updated WHO, ICC and ELN classifications have undertaken a major role in genetic data and have been integrated into diagnostic and prognostic algorithms (Table 2) [1,2,3]. In this context, genetic analyses currently mandatory in the evaluation of AML are conventional cytogenetics together with the screening of fusion genes and of gene mutations, whose number has significantly increased in comparison with prior classifications (Figure 1). Recommendations for molecular assessments in AML include the following genes: FLT3 (both internal tandem duplications, ITD, and tyrosine kinase domain, TKD, mutations), IDH1 and IDH2 as therapeutic targets, and NPM1, CEBPA, DDX41, TP53, ASXL1, BCOR, EZH2, RUNX1, SF3B1, SRSF2, STAG2, U2AF1 and ZRSR2 as diagnostic/prognostic disease markers. Furthermore, the screening of the most prevalent gene rearrangements include PML::RARA, RUNX1::RUNX1T1, CBFB::MYH11, KMT2A rearrangements, BCR::ABL1 and others and should be performed when rapid information is needed for the clinical management of a patient, or if the morphology and/or immunophenotype are highly suggestive of the presence of a particular fusion gene [3].
In this scenario, the screening of single genes is progressively being replaced by targeted NGS panels, which, in addition, could offer a combined solution for the parallel analysis of DNA for mutations and RNA for fusions in the same assay. However, the main limitation of NGS technology is that it requires batching to be cost-effective, making it unrealistic for most diagnostic laboratories to report NGS results within a week. In this context, conventional PCR-based techniques are still being used for the screening of relevant markers that require a rapid turn-around time (3–5 days), including NPM1, FLT3, IDH1/2, and fusion genes. In particular, FLT3-ITD evaluation is recommended to be performed by capillary electrophoresis [11] because it cannot be performed robustly by NGS.
Among the most relevant molecular changes included in the new classifications is the description of some new AML types with fusion genes such as DEK::NUP214, BCR::ABL1 or rearrangements involving KMT2A, MECOM and NUP98 (Table 2). Another remarkable change refers to the recognition of particular clinicobiological entities associated with adverse risk, AML with a TP53 mutation and the renamed AML with myelodysplasia-related gene mutations (ICC 2022)/AML myelodysplasia-related (WHO 2022), which harbor specific cytogenetic and molecular alterations beyond those previously considered ASXL1 and RUNX1 (Table 2). Regarding AML with CEBPA mutations, it remains a favorable prognostic entity, but a biallelic mutation is not required, according to several studies published; a single in-frame mutation in the basic leucine zipper (bZIP) region of CEBPA is sufficient for diagnosis [3,12,13]. At the prognostic level, all AML patients with FLT3-ITD are classified in the intermediate risk group irrespective of the FLT3-ITD ratio and the presence of the NPM1 mutation. Furthermore, the presence of adverse-risk cytogenetic aberrations in NPM1-mutated AML is now also indicative of high risk [3].
The study of an additional set of genes, including ANKRD26, BCORL1, BRAF, CBL, CSF3R, DNMT3A, ETV6, GATA2, JAK2, KIT, KRAS, NRAS, NF1, PHF6, PPM1D, PTPN11, RAD21, SETBP1, TET2 and WT1, is recommended at AML diagnosis, and some of them can also be used for MRD monitoring [3,14]. When AML with germline predisposition is suspected, mutational studies should be complemented with an extended NGS panel covering known predisposing genes together with genetic counseling. Both ICC and WHO updated classifications include specific categories of myeloid neoplasms with a germline predisposition characterized by the presence of germline inherited mutations and specific clinical features [1,2], but the discussion of these entities is beyond the scope of this review.

3. Monitoring

Though classical AML response criteria rely on morphological blast count, both MFC and molecular techniques offer higher sensitivity and specificity for the identification of malignant cells after therapy. Since 2017, ELN response criteria have included a complete response without MRD, recognizing the value of a deeper remission status. MRD detection is highly predictive of relapse [15,16] and allows for early intervention (MRD-directed therapy). Retrospective studies suggest pre-emptive treatment before overt morphological relapse and can improve the results of salvage treatment [17,18]: a hypothesis that is currently being addressed by prospective clinical trials (CT) [19]. In the allogeneic transplantation setting, the presence of MRD before this procedure also increases the risk of relapse [20] and could guide toward more intensive conditioning regimens [21]. Finally, MRD has been proposed as a surrogate endpoint for CT, which could accelerate drug approval.
Nonetheless, being aware of the sensitivity and limitations of each particular methodology is essential for the correct interpretation of MRD results. Thus, MRD negativity does not necessarily mean leukemia eradication but rather indicates a leukemic load below the limit of detection for each particular assay. Furthermore, MRD reliability is related to sample quality, as sensitivity is directly dependent on the number of processed cells, and specificity may be affected by sample viability. To guarantee as many viable cells as possible, it is recommended that BM aspiration is first pulled using the first 48 h for MRD studies while prioritizing MFC [22].

3.1. Flow Cytometry for MRD Detection in AML

Indeed, many studies have proven that the identification of leukemic cells by MFC after induction therapy or during consolidation treatment is a strong indicator of a higher relapse risk with an impact on overall survival [23,24,25,26]. There is also evidence of the MRD prognostic value in a low-intensity therapy setting [27,28]. The ELN MRD Working Party proposes an MFC-MRD assessment based on the detection of immunophenotypic anomalies of leukemic cells following LAIP [29] and is different from normal (DfN) [30] strategies. The LAIP strategy is based on identifying abnormal antigen patterns that are detected at diagnosis. The DfN approach is applicable to patients for whom the diagnostic phenotype is unknown and does not rely on its stability over time.
MFC is the most commonly used technology for MRD detection [22] because of its widespread use in most hematopathology laboratories and the fact that it is applicable to 85–90% of patients with AML [3] with a limited cost (Figure 1). Another advantage of MFC is that it is able to detect signs of hemodilution through the examination of other cell subsets [31]. Moreover, forward and side scatter properties hint at the overall sample quality, which can be more accurately assessed using a viability dye [31]. Modern cytometers can process several millions of cells in a few minutes, analyzing at least eight markers at a time. Assuming a tube composition is able to distinguish the aberrant phenotype from background noise, theoretical sensitivity lies between 10−3 and 10−5. However, the clinically validated threshold proposed by ELN recommendations is only 0.1%, which is lower than the sensitivity cut-off of many molecular techniques due to technical limitations and interpretation complexity. One of the main caveats of MFC is its low reproducibility, which is caused by differences in equipment, cytometer set-up, sample processing protocols and reagents across institutions. Even though there is a set of markers that are considered basic for AML MRD detection (Table 1), tube combination is highly variable and can greatly determine the gating strategy. In addition, AML is phenotypically highly heterogeneous, both in terms of the wide range of possible aberrancies as of intra-tumor complexity with different subclones, which may exhibit particular immunophenotypes. Moreover, LAIP shifts are not rare at relapse, and certain aberrancies can be found in healthy and regenerating bone marrow at low frequencies, which compels specificity thresholds for every specific assay [32]. Expertise is required for every examiner to overcome interpretation complexity [22]. Recently, some publications have addressed the standardization of MRD analysis by MFC in order to improve the inter-rater reliability between investigators [31,33]. Furthermore, automated analysis tools are being developed to achieve unbiased results [34,35,36]. With standardized assays, deeper sensitivity thresholds will probably be accepted for clinical use in the future.
The study of the leukemia stem cell (LSC) compartment is still considered investigational, though its prognostic value has been demonstrated in prospective studies [37,38] and, thus, may also be recommended for clinical routine analysis in the near future.
Spectral flow cytometry allows for the simultaneous analysis of more than 20 markers, which can greatly increase its capability to identify immunophenotypic aberrancies and increase the number of analyzed cells when a sample is scarce [39]. Nevertheless, this methodology is still investigational and far from being implemented in most laboratories.

3.2. Molecular Monitoring

The advent of precision medicine, together with technological advances, has led to a new era in molecular AML monitoring, with the challenge of choosing the most appropriate technique and the best molecular marker for each patient. At a technical level, molecular MRD should be assessed by methodologies with a limit of detection of at least 10−3. To achieve this value, the technologies currently recommended by the ELN in AML are real-time quantitative PCR (qPCR), digital PCR (dPCR) and NGS (Figure 1), taking into account that dPCR and NGS are still exploratory and need to be integrated with MFC-MRD data [22].

3.2.1. qPCR

Real-time quantitative PCR (qPCR) was the first molecular methodology to be established for MRD leukemia monitoring because of its capability to accurately quantify the mutational load of several genetic aberrations with high sensitivity (10−5–10−6) [40]. qPCR allows for the real-time measurement of PCR amplification products by means of fluorescent assays that are specifically designed for each alteration, including fusion genes as well as gene mutations. For many years and still today, qPCR has been the gold-standard technique for monitoring AML patients harboring the fusion genes PML::RARA in APL and RUNX1::RUNX1T1 and CBFB::MYH11 in core-binding factor (CBF) leukemias [41,42,43]. The methodology and interpretation of qPCR for these fusion genes have been standardized by Europe Against Cancer (EAC) Consortium [40,44], with a wide application in routine patients’ clinical management because of their simplicity, relatively low cost and high sensitivity. In addition to APL and CBF leukemias, the clinical value of MRD monitoring has been also well established and is strongly recommended in AML patients with NPM1 mutations [45,46]. The high gene expression of these fusion genes and NPM1 mutations allows for the analysis of RNA as a preferred starting material with excellent sensitivity and facilitates the study of fusion genes without requiring the particular location of each intronic breakpoint [47]. Therefore, in patients with these alterations, the molecular monitoring of aberrant transcripts should be performed at the time points at which MRD is considered clinically relevant, with a high predictive value for relapse, as defined in the latest ELN recommendations [22]. Although the analysis in the peripheral blood is currently accepted at some time points, it should be noted that MRD assessment in bone marrow has higher sensitivity by an order of magnitude compared with blood [48].
qPCR can also be used to monitor other MRD targets through the design of particular assays that are specific for each type of genetic alteration. Thus, qPCR is also applicable in the analysis of other recurrent fusion genes, such as KMT2A rearrangements or the DEK::NUP214 fusion resulting from t(6;9)(p22.3;q34.1), as well as atypical NPM1 mutations and KMT2A partial tandem duplications [46,49,50]. All these targets are clinically valuable MRD markers; however, the qPCR-based methodology used was not standardized given the lack of studies with large series of patients. Despite its high sensitivity and robustness, the main drawback of qPCR is its limited applicability. About 50% of AML patients do not carry NPM1 mutations or recurrent fusion genes, and in this context, there is an urgent need to introduce novel molecular MRD techniques in order to monitor a broader range of AML patients.

3.2.2. Digital PCR

Droplet digital PCR (ddPCR) is the most extensively used dPCR technology and has emerged as an attractive method to monitor MRD while overcoming some limitations of conventional techniques. ddPCR can be more accurate than qPCR, particularly in disease quantification at a very low level, as each sample is initially fractionated and the final analysis is based on thousands of individual measurements, obtaining an absolute quantification of the amplified target of interest without the need for a standard curve [51]. This provides ddPCR with the ability to repeatedly monitor patients’ MRD with high sensitivity. The limit of detection in this technique depends on several aspects, including the specific design assay or the number of replicates per sample, but in any case, should be at least 10−3 according to ELN recommendations. In recent years, several studies have shown the potential of ddPCR to analyze MRD in AML, both in monitoring somatic mutations and fusion genes [52]. The feasibility of ddPCR to track MRD in AML has been explored in different studies, in particular, for NPM1-mutated patients, with the advantage of atypical mutation screening through the design of specific assays [53]. Based on the published data, the last ELN update already included dPCR, together with qPCR, as one of the recommended technologies for the molecular MRD assessment in mutant NPM1, CBF AML or APL. Regarding the monitoring of other recurrent mutations, some studies have shown the potential of ddPCR for predicting AML relapse, including IDH1/2 [46,54]. However, the value of some of these mutations as potential MRD markers should be validated in additional studies with larger cohorts of patients.
For the MRD assessment of rare mutations, ddPCR could be useful due to its versatility when designing alteration-specific assays, but at the same time, this could also be a pitfall due to the lack of standardized assays. For this reason, to date, the potential of ddPCR disease tracking in cases with specific gene alterations is still under investigation, and its MRD results must be interpreted together with MFC data.

3.2.3. NGS

As discussed above, the use of targeted NGS panels is one of the most extended approaches when accurately diagnosing and stratifying risks in AML patients. In addition to initial diagnosis and relapse, NGS has recently become a potentially useful tool for monitoring MRD in AML due to the development of error-corrected NGS, which increases sensitivity through the use of random barcodes or unique molecule identifiers [55]. These strategies allow for the identification and removal of artifacts introduced by PCR amplification during library preparation, leading to the reliable and accurate tracking of genetic targets with a limit of detection of at least 10−3. Due to its intrinsic characteristics, NGS-MRD provides a potential solution to some limitations of the conventional molecular techniques due to its capability to monitor in the same run of sequencing specific and various gene alterations in virtually all AML patients. Thus, several studies have shown that MRD monitoring by NGS has a predictive value for AML relapse, as well as prognostic value, together with an excellent correlation with treatment efficacy [16,56,57]. However, despite being promising, these strategies require highly specialized bioinformatic tools that are not yet available in most routine laboratories. Moreover, some technical issues need to be solved and standardized before entering into clinical practice. In this scenario, there needs to be an increasing cooperative effort to harmonize NGS-MRD protocols. Partly due to this lack of standardization, the best strategy is to combine NGS with MFC for MRD assessment [58].

3.2.4. Selection of MRD Molecular Markers

Although all pathogenic/likely pathogenic variants detected at AML diagnosis could be considered potential MRD markers, some limitations should be taken into account (Table 3). First, confirmed or suspected germline mutations with a variant allele frequency higher than 30% in ANKRD26, CEBPA, DDX41, ETV6, GATA2, RUNX1 and TP53 genes, should be excluded as they are not informative for disease monitoring [22]. Among the somatically acquired mutations, it is crucial to distinguish those that are specific from leukemic cells with the potential to relapse and those related to pre-leukemic clonal hematopoiesis (CH). In this context, mutations known as DTA (affecting DNMT3A, TET2 or ASXL1 genes) often persist in complete remission (CR) without being associated with a higher risk of relapse and should be excluded from MRD tracking [16,59,60]. Regarding other less frequently mutated genes related to CH, including non-DTA, such as SRSF2, IDH1 or IDH2, preliminary studies seem to indicate that their detection at CR is not associated with AML relapse [61], although larger studies are needed to confirm this result.
On the contrary, late event mutations in genes with a transformative leukemic capacity, such as NPM1, FLT3, RUNX1 or RAS, are considered good MRD markers, as their detection in CR can be associated with an increased risk of relapse, representing residual leukemic cells when detected [59]. Nonetheless, mutations in signaling kinase pathways (FLT3, RAS, KIT) are usually subclonal and are not always present at relapse; therefore, it is highly recommended to conduct an analysis in combination with additional molecular markers. In patients treated with targeted agents (FLT3, IDH1/2 inhibitors), the molecular monitoring of targeted markers is recommended in combination with others [22]. In fact, the analysis of additional markers may also be applied even in NPM1 mutated AML patients, given the not negligible frequency of wild-type NPM1 when relapsed among AML patients with an NPM1 mutation [46,62]. All this accumulated evidence may favor the use of a multigene MRD panel as a strategy to target several mutations per patient, excluding DTA and genes related to germline predisposition.

4. Conclusions

Over the last decade, the growing knowledge of the pathogenesis of AML, together with new technological advances and the success of targeted therapies, have led to a new era in the diagnosis and follow-up of AML. In this context, guidelines for disease classification, risk stratification and MRD monitoring have been recently updated, prompting routine laboratories to incorporate new diagnostic and monitoring tools in order to optimally characterize and track leukemic cells. Due to the remarkable AML biological heterogeneity, both flow cytometry-based as well as molecular procedures require specialized expertise and, in this context, harmonization, which is of critical importance. Cooperative efforts are underway, which is particularly challenging for MRD studies, given their strong relevance in AML clinical practice. Automated unsupervised analysis strategies may improve the standardization of MFC-MRD assays and overcome the subjectivity associated with flow cytometry. Emerging technologies, such as NGS-MRD and dPCR, should be incorporated into the arsenal of MRD techniques, taking into account that, although promising, they are still complementary and need to be integrated with other laboratory data.

Author Contributions

Conceptualization, writing and supervision, F.G. and M.L.-G.; Review and editing, F.G., M.G., N.V., J.E., D.C. and M.L.-G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Fondo de Investigaciones Sanitarias/Instituto de Salud Carlos III (ISCIII) grant PI22/01660.

Institutional Review Board Statement

The study did not require ethical approval.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef] [PubMed]
  2. Arber, D.A.; Orazi, A.; Hasserjian, R.P.; Borowitz, M.J.; Calvo, K.R.; Kvasnicka, H.-M.; Wang, S.A.; Bagg, A.; Barbui, T.; Branford, S.; et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: Integrating morphologic, clinical, and genomic data. Blood 2022, 140, 1200–1228. [Google Scholar] [CrossRef] [PubMed]
  3. Döhner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef] [PubMed]
  4. Angelini, D.F.; Ottone, T.; Guerrera, G.; Lavorgna, S.; Cittadini, M.; Buccisano, F.; De Bardi, M.; Gargano, F.; Maurillo, L.; Divona, M.; et al. A leukemia-associated CD34/CD123/CD25/CD99+ immunophenotype identifies FLT3-mutated clones in acute myeloid Leukemia. Clinical Cancer Research. Clin. Cancer Res. 2015, 21, 3977–3985. [Google Scholar] [CrossRef] [PubMed]
  5. Kalina, T.; Flores-Montero, J.; van der Velden, V.H.J.; Martin-Ayuso, M.; Böttcher, S.; Ritgen, M.; Almeida, J.; Lhermitte, L.; Asnafi, V.; Mendonça, A.; et al. EuroFlow standardization of flow cytometer instrument settings and immunophenotyping protocols. Leukemia 2012, 26, 1986–2010. [Google Scholar] [CrossRef]
  6. Lhermitte, L.; on behalf of Mthe EuroFlow Consortium; Mejstrikova, E.; van der Sluijs-Gelling, A.J.; Grigore, G.E.; Sedek, L.; Bras, A.E.; Gaipa, G.; da Costa, E.S.; Novakova, M.; et al. Automated database-guided expert-supervised orientation for immunophenotypic diagnosis and classification of acute leukemia. Leukemia 2018, 32, 874–881. [Google Scholar] [CrossRef]
  7. Lhermitte, L.; Barreau, S.; Morf, D.; Fernandez, P.; Grigore, G.; Barrena, S.; de Bie, M.; Flores-Montero, J.; Brüggemann, M.; Mejstrikova, E.; et al. Automated identification of leukocyte subsets improves standardization of database-guided expert-supervised diagnostic orientation in acute leukemia: A EuroFlow study. Mod. Pathol. 2021, 34, 59–69. [Google Scholar] [CrossRef]
  8. Martínez, D.S.; Tirado, N.; Mensurado, S.; Martínez-Moreno, A.; Romecín, P.; Agüera, F.G.; Correia, D.V.; Silva-Santos, B.; Menéndez, P. Generation and proof-of-concept for allogeneic CD123 CAR-Delta One T (DOT) cells in acute myeloid leukemia. J. Immunother. Cancer 2022, 10, e005400. [Google Scholar] [CrossRef]
  9. The Cancer Genome Atlas Research Network; Ley, T.J.; Miller, C.; Ding, L.; Raphael, B.J.; Mungall, A.J.; Robertson, G.; Hoadley, K.; Triche, T.; Laird, P.; et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 2013, 368, 2059–2074. [Google Scholar]
  10. Papaemmanuil, E.; Gerstung, M.; Bullinger, L.; Gaidzik, V.I.; Paschka, P.; Roberts, N.D.; Potter, N.E.; Heuser, M.; Thol, F.; Bolli, N.; et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N. Eng. J. Med. 2016, 374, 2209–2221. [Google Scholar] [CrossRef]
  11. Thiede, C.; Steudel, C.; Mohr, B.; Schaich, M.; Schäkel, U.; Platzbecker, U.; Wermke, M.; Bornhäuser, M.; Ritter, M.; Neubauer, A.; et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: Association with FAB subtypes and identification of subgroups with poor prognosis. Blood 2002, 99, 4326–4335. [Google Scholar] [CrossRef] [PubMed]
  12. Taube, F.; Georgi, J.A.; Kramer, M.; Stasik, S.; Middeke, J.M.; Röllig, C.; Krug, U.; Krämer, A.; Scholl, S.; Hochhaus, A.; et al. CEBPA mutations in 4708 patients with acute myeloid leukemia: Differential impact of bZIP and TAD mutations on outcome. Blood 2022, 139, 87–103. [Google Scholar] [CrossRef] [PubMed]
  13. Wakita, S.; Sakaguchi, M.; Oh, I.; Kako, S.; Toya, T.; Najima, Y.; Doki, N.; Kanda, J.; Kuroda, J.; Mori, S.; et al. Prognostic impact of CEBPA bZIP domain mutation in acute myeloid leukemia. Blood Adv. 2022, 6, 238–247. [Google Scholar] [CrossRef] [PubMed]
  14. Schuurhuis, G.J.; Heuser, M.; Freeman, S.; Béné, M.-C.; Buccisano, F.; Cloos, J.; Grimwade, D.; Haferlach, T.; Hills, R.K.; Hourigan, C.S.; et al. Minimal/measurable residual disease in AML: A consensus document from the European LeukemiaNet MRD Working Party. Blood 2018, 131, 1275–1291. [Google Scholar] [CrossRef]
  15. Short, N.J.; Fu, C.; Berry, D.A.; Walter, R.B.; Freeman, S.D.; Hourigan, C.S.; Huang, X.; Gonzalez, G.N.; Hwang, H.; Qi, X.; et al. Association of hematologic response and assay sensitivity on the prognostic impact of measurable residual disease in acute myeloid leukemia: A systematic review and meta-analysis. Leukemia 2022, 36, 2817–2826. [Google Scholar] [CrossRef]
  16. Jongen-Lavrencic, M.; Grob, T.; Hanekamp, D.; Kavelaars, F.G.; al Hinai, A.; Zeilemaker, A.; Erpelinck-Verschueren, C.A.; Gradowska, P.L.; Meijer, R.; Cloos, J.; et al. Molecular Minimal Residual Disease in Acute Myeloid Leukemia. N. Engl. J. Med. 2018, 378, 1189–1199. [Google Scholar] [CrossRef]
  17. Bataller, A.; Oñate, G.; Diaz-Beyá, M.; Guijarro, F.; Garrido, A.; Vives, S.; Tormo, M.; Arnan, M.; Salamero, O.; Sampol, A.; et al. Acute myeloid leukemia with NPM1 mutation and favorable European LeukemiaNet category: Outcome after preemptive intervention based on measurable residual disease. Br. J. Haematol. 2020, 191, 52–61. [Google Scholar] [CrossRef]
  18. Short, N.J.; Macaron, W.; Kadia, T.; Dinardo, C.; Issa, G.C.; Daver, N.; Wang, S.; Jorgensen, J.; Nguyen, D.; Bidikian, A.; et al. Clinical outcomes and impact of therapeutic intervention in patients with acute myeloid leukemia who experience measurable residual disease (MRD) recurrence following MRD -negative remission. Am. J. Hematol. 2022, 97, E408–E411. [Google Scholar] [CrossRef]
  19. Tiong, I.S.; Loo, S. Targeting Measurable Residual Disease (MRD) in Acute Myeloid Leukemia (AML): Moving beyond Prognostication. Int. J. Mol. Sci. 2023, 24, 4790. [Google Scholar] [CrossRef]
  20. Buckley, S.A.; Wood, B.L.; Othus, M.; Hourigan, C.S.; Ustun, C.; Linden, M.A.; DeFor, T.E.; Malagola, M.; Anthias, C.; Valkova, V.; et al. Minimal residual disease prior to allogeneic hematopoietic cell transplantation in acute myeloid leukemia: A meta-analysis. Haematologica 2017, 102, 865–873. [Google Scholar] [CrossRef]
  21. Hourigan, C.S.; Dillon, L.W.; Gui, G.; Logan, B.R.; Fei, M.; Ghannam, J.; Li, Y.; Licon, A.; Alyea, E.P.; Bashey, A.; et al. Impact of Conditioning Intensity of Allogeneic Transplantation for Acute Myeloid Leukemia with Genomic Evidence of Residual Disease. J. Clin. Oncol. 2020, 38, 1273–1283. [Google Scholar] [CrossRef]
  22. Heuser, M.; Freeman, S.D.; Ossenkoppele, G.J.; Buccisano, F.; Hourigan, C.S.; Ngai, L.L.; Tettero, J.M.; Bachas, C.; Baer, C.; Béné, M.-C.; et al. 2021 Update on MRD in acute myeloid leukemia: A consensus document from the European LeukemiaNet MRD Working Party. Blood 2021, 138, 2753–2767. [Google Scholar] [CrossRef] [PubMed]
  23. Köhnke, T.; Sauter, D.; Ringel, K.; Hoster, E.; Laubender, R.P.; Hubmann, M.; Bohlander, S.K.; Kakadia, P.M.; Schneider, S.; Dufour, A.; et al. Early assessment of minimal residual disease in AML by flow cytometry during aplasia identifies patients at increased risk of relapse. Leukemia 2015, 29, 377–386. [Google Scholar] [CrossRef] [PubMed]
  24. Ravandi, F.; Jorgensen, J.; Borthakur, G.; Jabbour, E.; Kadia, T.; Pierce, S.; Brandt, M.; Wang, S.; Konoplev, S.; Wang, X.; et al. Persistence of minimal residual disease assessed by multiparameter flow cytometry is highly prognostic in younger patients with acute myeloid leukemia. Cancer 2017, 123, 426–435. [Google Scholar] [CrossRef]
  25. Buldini, B.; Rizzati, F.; Masetti, R.; Fagioli, F.; Menna, G.; Micalizzi, C.; Putti, M.C.; Rizzari, C.; Santoro, N.; Zecca, M.; et al. Prognostic significance of flow-cytometry evaluation of minimal residual disease in children with acute myeloid leukaemia treated according to the AIEOP-AML 2002/01 study protocol. Br. J. Haematol. 2017, 177, 116–126. [Google Scholar] [CrossRef] [PubMed]
  26. Terwijn, M.; Van Putten, W.L.; Kelder, A.; Van Der Velden, V.H.; Brooimans, R.A.; Pabst, T.; Maertens, J.; Boeckx, N.; De Greef, G.E.; Valk, P.J.; et al. High Prognostic Impact of Flow Cytometric Minimal Residual Disease Detection in Acute Myeloid Leukemia: Data From the HOVON/SAKK AML 42A Study. J. Clin. Oncol. 2013, 31, 3889–3897. [Google Scholar] [CrossRef] [PubMed]
  27. Pratz, K.W.; Jonas, B.A.; Pullarkat, V.; Recher, C.; Schuh, A.C.; Thirman, M.J.; Garcia, J.S.; DiNardo, C.D.; Vorobyev, V.; Fracchiolla, N.S.; et al. Measurable Residual Disease Response and Prognosis in Treatment-Naïve Acute Myeloid Leukemia with Venetoclax and Azacitidine. J. Clin. Oncol. 2022, 40, 855–865. [Google Scholar] [CrossRef] [PubMed]
  28. Montesinos, P.; Bergua, J.; Infante, J.; Esteve, J.; Guimaraes, J.E.; Sierra, J.; Sanz, M. Update on management and progress of novel therapeutics for R/R AML: An Iberian expert panel consensus. Ann. Hematol. 2019, 98, 2467–2483. [Google Scholar] [CrossRef]
  29. Zeijlemaker, W.; Kelder, A.; Cloos, J.; Schuurhuis, G.J. Immunophenotypic Detection of Measurable Residual (Stem Cell) Disease Using LAIP Approach in Acute Myeloid Leukemia. Curr. Protoc. Cytom. 2019, 91, e66. [Google Scholar] [CrossRef]
  30. Wood, B.L. Acute Myeloid Leukemia Minimal Residual Disease Detection: The Difference from Normal Approach. Curr. Protoc. Cytom. 2020, 93, e73. [Google Scholar] [CrossRef]
  31. Tettero, J.M.; Freeman, S.; Buecklein, V.; Venditti, A.; Maurillo, L.; Kern, W.; Walter, R.B.; Wood, B.L.; Roumier, C.; Philippé, J.; et al. Technical Aspects of Flow Cytometry-based Measurable Residual Disease Quantification in Acute Myeloid Leukemia: Experience of the European LeukemiaNet MRD Working Party. Hemasphere 2021, 6, e676. [Google Scholar] [CrossRef]
  32. Sui, J.; Chen, Q.; Zhang, Y.; Sheng, Y.; Wu, J.; Li, J.; Weng, X.; Chen, B. Identifying leukemia-associated immunophenotype-based individualized minimal residual disease in acute myeloid leukemia and its prognostic significance. Am. J. Hematol. 2019, 94, 528–538. [Google Scholar] [CrossRef] [PubMed]
  33. Röhnert, M.A.; Kramer, M.; Schadt, J.; Ensel, P.; Thiede, C.; Krause, S.W.; Bücklein, V.; Hoffmann, J.; Jaramillo, S.; Schlenk, R.F.; et al. Reproducible measurable residual disease detection by multiparametric flow cytometry in acute myeloid leukemia. Leukemia 2022, 36, 2208–2217. [Google Scholar] [CrossRef] [PubMed]
  34. Lacombe, F.; Lechevalier, N.; Vial, J.P.; Béné, M.C. An R-Derived FlowSOM Process to Analyze Unsupervised Clustering of Normal and Malignant Human Bone Marrow Classical Flow Cytometry Data. Cytom. Part A 2019, 95, 1191–1197. [Google Scholar] [CrossRef] [PubMed]
  35. Vial, J.P.; Lechevalier, N.; Lacombe, F.; Dumas, P.-Y.; Bidet, A.; Leguay, T.; Vergez, F.; Pigneux, A.; Béné, M.C. Unsupervised Flow Cytometry Analysis Allows for an Accurate Identification of Minimal Residual Disease Assessment in Acute Myeloid Leukemia. Cancers 2021, 13, 629. [Google Scholar] [CrossRef]
  36. Piñero, P.; Morillas, M.; Gutierrez, N.; Barragán, E.; Such, E.; Breña, J.; García-Hernández, M.C.; Gil, C.; Botella, C.; González-Navajas, J.M.; et al. Identification of Leukemia-Associated Immunophenotypes by Databaseguided Flow Cytometry Provides a Highly Sensitive and Reproducible Strategy for the Study of Measurable Residual Disease in Acute Myeloblastic Leukemia. Cancers 2022, 14, 4010. [Google Scholar] [CrossRef]
  37. Zeijlemaker, W.; Grob, T.; Meijer, R.; Hanekamp, D.; Kelder, A.; Carbaat-Ham, J.C.; Oussoren-Brockhoff, Y.J.M.; Snel, A.N.; Veldhuizen, D.; Scholten, W.J.; et al. CD34+CD38− leukemic stem cell frequency to predict outcome in acute myeloid leukemia. Leukemia 2019, 33, 1102–1112. [Google Scholar] [CrossRef]
  38. Li, S.-Q.; Xu, L.-P.; Wang, Y.; Zhang, X.-H.; Chen, H.; Chen, Y.-H.; Wang, F.-R.; Han, W.; Sun, Y.-Q.; Yan, C.-H.; et al. An LSC-based MRD assay to complement the traditional MFC method for prediction of AML relapse: A prospective study. Blood 2022, 140, 516–520. [Google Scholar] [CrossRef]
  39. Soh, K.T.; Conway, A.; Liu, X.; Wallace, P.K. Development of a 27-color panel for the detection of measurable residual disease in patients diagnosed with acute myeloid leukemia. Cytom. Part A 2022, 101, 970–983. [Google Scholar] [CrossRef]
  40. Gabert, J.; Beillard, E.; van der Velden, V.H.J.; Bi, W.; Grimwade, D.; Pallisgaard, N.; Barbany, G.; Cazzaniga, G.; Cayuela, J.M.; Cavé, H.; et al. Standardization and quality control studies of ‘real-time’ quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia—A Europe Against Cancer Program. Leukemia 2003, 17, 2318–2357. [Google Scholar] [CrossRef]
  41. Sanz, M.A.; Fenaux, P.; Tallman, M.S.; Estey, E.H.; Löwenberg, B.; Naoe, T.; Lengfelder, E.; Döhner, H.; Burnett, A.K.; Chen, S.-J.; et al. Management of acute promyelocytic leukemia: Updated recommendations from an expert panel of the European LeukemiaNet. Blood 2019, 133, 1630–1643. [Google Scholar] [CrossRef]
  42. Jourdan, E.; Boissel, N.; Chevret, S.; Delabesse, E.; Renneville, A.; Cornillet, P.; Blanchet, O.; Cayuela, J.-M.; Recher, C.; Raffoux, E.; et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood 2013, 121, 2213–2223. [Google Scholar] [CrossRef]
  43. Corbacioglu, A.; Scholl, C.; Schlenk, R.F.; Eiwen, K.; Du, J.; Bullinger, L.; Fröhling, S.; Reimer, P.; Rummel, M.; Derigs, H.-G.; et al. Prognostic Impact of Minimal Residual Disease inCBFB-MYH11–Positive Acute Myeloid Leukemia. J. Clin. Oncol. 2010, 28, 3724–3729. [Google Scholar] [CrossRef]
  44. Beillard, E.; Pallisgaard, N.; van der Velden, V.H.J.; Bi, W.; Dee, R.; van der Schoot, E.; Delabesse, E.; Macintyre, E.; Gottardi, E.; Saglio, G.; et al. Evaluation of candidate control genes for diagnosis and residual disease detection in leukemic patients using ‘real-time’ quantitative reverse-transcriptase polymerase chain reaction (RQ-PCR)—A Europe against cancer program. Leukemia 2003, 17, 2474–2486. [Google Scholar] [CrossRef]
  45. Gorello, P.; Cazzaniga, G.; Alberti, F.; Dell’Oro, M.G.; Gottardi, E.; Specchia, G.; Roti, G.; Rosati, R.; Martelli, M.F.; Diverio, D.; et al. Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations. Leukemia 2006, 20, 1103–1108. [Google Scholar] [CrossRef]
  46. Ivey, A.; Hills, R.K.; Simpson, M.A.; Jovanovic, J.V.; Gilkes, A.; Grech, A.; Patel, Y.; Bhudia, N.; Farah, H.; Mason, J.; et al. Assessment of Minimal Residual Disease in Standard-Risk AML. N. Engl. J. Med. 2016, 374, 422–433. [Google Scholar] [CrossRef]
  47. Dillon, L.W.; Hayati, S.; Roloff, G.; Tunc, I.; Pirooznia, M.; Mitrofanova, A.; Hourigan, C.S. Targeted RNA-sequencing for the quantification of measurable residual disease in acute myeloid leukemia. Haematologica 2019, 104, 297–304. [Google Scholar] [CrossRef]
  48. Thol, F.; Gabdoulline, R.; Liebich, A.; Klement, P.; Schiller, J.; Kandziora, C.; Hambach, L.; Stadler, M.; Koenecke, C.; Flintrop, M.; et al. Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML. Blood 2018, 132, 1703–1713. [Google Scholar] [CrossRef]
  49. Schnittger, S.; Kern, W.; Tschulik, C.; Weiss, T.; Dicker, F.; Falini, B.; Haferlach, C.; Haferlach, T. Minimal residual disease levels assessed by NPM1 mutation–specific RQ-PCR provide important prognostic information in AML. Blood 2009, 114, 2220–2231. [Google Scholar] [CrossRef]
  50. Ravandi, F.; Walter, R.B.; Freeman, S.D. Evaluating measurable residual disease in acute myeloid leukemia. Blood Adv. 2018, 2, 1356–1366. [Google Scholar] [CrossRef]
  51. Galimberti, S.; Balducci, S.; Guerrini, F.; Del Re, M.; Cacciola, R. Digital Droplet PCR in Hematologic Malignancies: A New Useful Molecular Tool. Diagnostics 2022, 12, 1305. [Google Scholar] [CrossRef] [PubMed]
  52. Coccaro, N.; Tota, G.; Anelli, L.; Zagaria, A.; Specchia, G.; Albano, F. Digital PCR: A Reliable Tool for Analyzing and Monitoring Hematologic Malignancies. Int. J. Mol. Sci. 2020, 21, 3141. [Google Scholar] [CrossRef]
  53. Bacher, U.; Dicker, F.; Haferlach, C.; Alpermann, T.; Rose, D.; Kern, W.; Haferlach, T.; Schnittger, S. Quantification of rare NPM1 mutation subtypes by digital PCR. Br. J. Haematol. 2014, 167, 710–714. [Google Scholar] [CrossRef] [PubMed]
  54. Brambati, C.; Galbiati, S.; Xue, E.; Toffalori, C.; Crucitti, L.; Greco, R.; Sala, E.; Crippa, A.; Chiesa, L.; Soriani, N.; et al. Droplet digital polymerase chain reaction for DNMT3A and IDH1/2 mutations to improve early detection of acute myeloid leukemia relapse after allogeneic hematopoietic stem cell transplantation. Haematologica 2016, 101, e157–e161. [Google Scholar] [CrossRef]
  55. Crowgey, E.L.; Mahajan, N.; Wong, W.H.; Gopalakrishnapillai, A.; Barwe, S.P.; Kolb, E.A.; Druley, T.E. Error-corrected sequencing strategies enable comprehensive detection of leukemic mutations relevant for diagnosis and minimal residual disease monitoring. BMC Med. Genom. 2020, 13, 32. [Google Scholar] [CrossRef]
  56. Patkar, N.; Kakirde, C.; Shaikh, A.F.; Salve, R.; Bhanshe, P.; Chatterjee, G.; Rajpal, S.; Joshi, S.; Chaudhary, S.; Kodgule, R.; et al. Clinical impact of panel-based error-corrected next generation sequencing versus flow cytometry to detect measurable residual disease (MRD) in acute myeloid leukemia (AML). Leukemia 2021, 35, 1392–1404. [Google Scholar] [CrossRef] [PubMed]
  57. Vonk, C.M.; Al Hinai, A.S.A.; Hanekamp, D.; Valk, P.J.M. Molecular Minimal Residual Disease Detection in Acute Myeloid Leukemia. Cancers 2021, 13, 5431. [Google Scholar] [CrossRef]
  58. McGowan, P.F.; Hyter, S.D.; Cui, W.; Plummer, R.M.; Godwin, A.K.; Zhang, D. Comparison of flow cytometry and next-generation sequencing in minimal residual disease monitoring of acute myeloid leukemia: One institute’s practical clinical experience. Int. J. Lab. Hematol. 2022, 44, 118–126. [Google Scholar] [CrossRef]
  59. Hasserjian, R.P.; Steensma, D.P.; Graubert, T.A.; Ebert, B.L. Clonal hematopoiesis and measurable residual disease assessment in acute myeloid leukemia. Blood 2020, 135, 1729–1738. [Google Scholar] [CrossRef]
  60. Heuser, M.; Heida, B.; Büttner, K.; Wienecke, C.P.; Teich, K.; Funke, C.; Brandes, M.; Klement, P.; Liebich, A.; Wichmann, M.; et al. Posttransplantation MRD monitoring in patients with AML by next-generation sequencing using DTA and non-DTA mutations. Blood Adv. 2021, 5, 2294–2304. [Google Scholar] [CrossRef]
  61. Cappelli, L.V.; Meggendorfer, M.; Baer, C.; Nadarajah, N.; Hutter, S.; Jeromin, S.; Dicker, F.; Kern, W.; Haferlach, T.; Haferlach, C.; et al. Indeterminate and oncogenic potential: CHIP vs CHOP mutations in AML with NPM1 alteration. Leukemia 2022, 36, 394–402. [Google Scholar] [CrossRef] [PubMed]
  62. Höllein, A.; Meggendorfer, M.; Dicker, F.; Jeromin, S.; Nadarajah, N.; Kern, W.; Haferlach, C.; Haferlach, T. NPM1 mutated AML can relapse with wild-type NPM1: Persistent clonal hematopoiesis can drive relapse. Blood Adv. 2018, 2, 3118–3125. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Tools for diagnosis and monitoring of AML. A combination of immunophenotyping and molecular studies is required for AML diagnosis and MRD monitoring. MRD: Measurable residual disease; LAIP: Leukemia-associated immunophenotype; DfN: Different from normal; qPCR: quantitative PCR; ddPCR: Droplet digital PCR.
Figure 1. Tools for diagnosis and monitoring of AML. A combination of immunophenotyping and molecular studies is required for AML diagnosis and MRD monitoring. MRD: Measurable residual disease; LAIP: Leukemia-associated immunophenotype; DfN: Different from normal; qPCR: quantitative PCR; ddPCR: Droplet digital PCR.
Curroncol 30 00395 g001
Table 1. Flow cytometry diagnostic and MRD markers.
Table 1. Flow cytometry diagnostic and MRD markers.
Diagnostic Markers
Lineage assigning antigens
MyeloperoxidaseMyeloid lineage
CD11c, CD14, CD64, lysozymeMyeloid lineage with monocytic differentiation
CD19B-lineage. Requires also at least one (CD19 strong) or two (CD19 weak) from CD22, CD10 and CD79a
CD3 (surface or cytoplasmic)T-lineage
Myeloid differentiation-associated antigens
CD13, CD33, CD11b, CD15, CD64Myeloid
CD14, CD36, CD64, CD4, CD11cMonocytic
CD41, CD42b, CD61, CD36Megakaryocytic
CD235a, CD71 strong, CD105, CD36Erythroid
CD203c, CD123Basophil
CD123, CD4, HLA-DR strong, CD303, CD304Dendritic
CD117 strongMastocytic
MRD markers
Basic markers
CD34, CD117, HLA-DR, CD45, CD13, CD33 Myeloid precursor identification
CD7, CD56Lymphoid antigen aberrancies
Other useful markers
CD64, CD14, CD11b, CD4Monocytic
CD19, CD2, CD5Lymphoid antigen aberrancies
CD38, CD123, CD133Leukemia stem cell identification
Table 2. Classification of AML according to ICC and WHO 2022.
Table 2. Classification of AML according to ICC and WHO 2022.
ICC 2022WHO 2022
CategoryBlasts Required for DiagnosisCategoryBlasts Required for Diagnosis
AML with Recurrent Genetic AbnormalitiesAML with Defining Genetic Abnormalities
Acute promyelocytic leukemia (APL) with t(15;17)(q24.1;q21.2)/PML::RARA≥10%Acute promyelocytic leukemia (APL) with PML::RARA fusionNo threshold
APL with other RARA rearrangements ≥10%
AML with t(8;21)(q22;q22.1)/RUNX1::RUNX1T1≥10%AML with RUNX1::RUNX1T1 fusionNo threshold
AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22)/CBFB::MYH11≥10%AML with CBFB::MYH11 fusionNo threshold
AML with t(9;11)(p21.3;q23.3)/MLLT3::KMT2A≥10%AML with KMT2A rearrangementNo threshold
AML with other KMT2A rearrangements ≥10%
AML with t(6;9)(p22.3;q34.1)/DEK::NUP214≥10%AML with DEK::NUP214 fusionNo threshold
AML with inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2)/GATA2;
MECOM(EVI1)
≥10%AML with MECOM rearrangementNo threshold
AML with other MECOM rearrangements ≥10%
AML with other rare recurring translocations≥10%AML with NUP98 rearrangementNo threshold
AML with RBM15::MRTFA fusionNo threshold
AML with t(9;22)(q34.1;q11.2)/BCR::ABL1≥20%AML with BCR::ABL1 fusion≥20%
AML with mutated NPM1≥10%AML with NPM1 mutationNo threshold
AML with in-frame bZIP CEBPA mutations≥10%AML with CEBPA mutation≥20%
AML and MDS/AML with mutated TP5310–19% (MDS/AML) and
≥20% (AML)
AML and MDS/AML with myelodysplasia-related gene mutations
Defined by mutations in ASXL1, BCOR, EZH2, RUNX1, SF3B1, SRSF2, STAG2, U2AF1, or ZRSR2
10–19% (MDS/AML) and
≥20% (AML)
AML, myelodysplasia-related
Defined by one or more cytogenetics of molecular alterations:
-
Somatic mutations in ASXL1, BCOR, EZH2, SF3B1, SRSF2, STAG2, U2AF1, or ZRSR2
-
Cytogenetic abnormalities: complex karyotype (≥3 abnormalities); del(5q) or 5q loss; −7, del(7q) or 7q loss; del(11q); del(12p) or 12p loss; −13 or del(13q); del(17p) or 17p loss, i(17q), idic(X)(q13)
≥20%
AML with myelodysplasia-related cytogenetic abnormalities
Defined by a complex karyotype (≥3 unrelated clonal chromosomal abnormalities in the absence of other class-defining recurring genetic abnormalities), del(5q)/t(5q)/add(5q), −7/del(7q), +8, del(12p)/t(12p)/add(12p), i(17q),−17/add(17p) or del(17p), del(20q), and/or idic(X)(q13) clonal abnormalities
10–19% (MDS/AML) and
≥20% (AML)
AML with other defined genetic alterationsNo threshold
AML without recurrent genetic abnormalitiesAML without defining genetic abnormalities
AML not otherwise specified (NOS)10–19% (MDS/AML) and
≥20% (AML)
AML defined by differentiation≥20%
Table 3. Molecular AML markers and their clinical value.
Table 3. Molecular AML markers and their clinical value.
GeneDiagnosticPrognosticTherapeuticMRD Marker
FLT3
IDH1
IDH2
NPM1
CEBPA *
DDX41 *
TP53 *
ASXL1
BCOR
EZH2
RUNX1 *
SF3B1
SRSF2
STAG2
U2AF1
ZRSR2
ANKRD26 *
BCORL1
BRAF
CBL
CSF3R
DNMT3A
ETV6 *
GATA2 *
JAK2
KIT
KRAS
NRAS
NF1
PHF6
PPM1D
PTPN11
RAD21
SETBP1
TET2
WT1
Fusion GeneDiagnosticPrognosticTherapeuticMRD Marker
PML::RARA
CBFB::MYH11
RUNX1:RUNX1T1
KMT2Ar
BCR::ABL1
DEK::NUP214
MECOMr
NUP98r
KAT6A::CREBBP
RBM15::MRTFA
X::RARA
Grey colored cell: Marker with diagnostic, prognostic and/or therapeutic value in AML. Robust MRD marker. Less validated MRD marker. Potential MRD marker. * Excluded as MRD marker if suspicion of germline mutation (variant allele frequency higher than 30%). Excluded MRD marker.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guijarro, F.; Garrote, M.; Villamor, N.; Colomer, D.; Esteve, J.; López-Guerra, M. Novel Tools for Diagnosis and Monitoring of AML. Curr. Oncol. 2023, 30, 5201-5213. https://doi.org/10.3390/curroncol30060395

AMA Style

Guijarro F, Garrote M, Villamor N, Colomer D, Esteve J, López-Guerra M. Novel Tools for Diagnosis and Monitoring of AML. Current Oncology. 2023; 30(6):5201-5213. https://doi.org/10.3390/curroncol30060395

Chicago/Turabian Style

Guijarro, Francesca, Marta Garrote, Neus Villamor, Dolors Colomer, Jordi Esteve, and Mónica López-Guerra. 2023. "Novel Tools for Diagnosis and Monitoring of AML" Current Oncology 30, no. 6: 5201-5213. https://doi.org/10.3390/curroncol30060395

Article Metrics

Back to TopTop