You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 Lorena Diéguez -- 1744 2023-03-23 16:26:37 |
2 format correction Dean Liu -4 word(s) 1740 2023-03-27 13:02:37 |

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Teixeira, A.; Carreira, L.; Abalde-Cela, S.; Sampaio-Marques, B.; Areias, A.C.; Ludovico, P.; Diéguez, L. Techniques for Monitoring of MRD in AML. Encyclopedia. Available online: https://encyclopedia.pub/entry/42485 (accessed on 14 December 2025).
Teixeira A, Carreira L, Abalde-Cela S, Sampaio-Marques B, Areias AC, Ludovico P, et al. Techniques for Monitoring of MRD in AML. Encyclopedia. Available at: https://encyclopedia.pub/entry/42485. Accessed December 14, 2025.
Teixeira, Alexandra, Luís Carreira, Sara Abalde-Cela, Belém Sampaio-Marques, Anabela C. Areias, Paula Ludovico, Lorena Diéguez. "Techniques for Monitoring of MRD in AML" Encyclopedia, https://encyclopedia.pub/entry/42485 (accessed December 14, 2025).
Teixeira, A., Carreira, L., Abalde-Cela, S., Sampaio-Marques, B., Areias, A.C., Ludovico, P., & Diéguez, L. (2023, March 23). Techniques for Monitoring of MRD in AML. In Encyclopedia. https://encyclopedia.pub/entry/42485
Teixeira, Alexandra, et al. "Techniques for Monitoring of MRD in AML." Encyclopedia. Web. 23 March, 2023.
Techniques for Monitoring of MRD in AML
Edit

After diagnosis of acute myeloid leukemia (AML), patients undergo chemotherapy, achieving complete remission (CR) in the majority of cases. However, some residual leukemic cells (up to 1010 to 1012 cells) can remain undetectable, causing a high percentage of patients to relapse. This condition is conventionally referred to as minimal residual disease, although more recently this nomenclature has been replaced by measurable residual disease (MRD). The term “measurable” was proposed to indicate some contexts where Leukemic Stem Cell (LSC) levels are detectable by modern technologies that have a high sensitivity. MRD monitoring in AML is performed routinely in clinical practice since it is a strong indicator of relapse. Besides, MRD can also have implications in the planning and personalization of treatment, when assessed in conjunction with other well-established clinical, cytogenetic, and molecular data. Furthermore, it has been shown that when MRD detection is successfully performed at an early stage, it results in improved prognosis, disease management, and patient outcome. Some of the currently used techniques for MRD assessment in AML are multiparametric flow cytometry (MFC) and molecular-based techniques, such as reverse transcription polymerase chain reaction (RT-PCR) and next-generation sequencing (NGS), further described below.

acute myeloid leukemia (AML) measurable residual disease (MRD) microfluidics

1. Multiparametric Flow Cytometry (MFC)

MFC is one of the most common techniques used for the diagnosis, classification, and prognosis of AML disease. This technique identifies a signature of aberrant expression of antigens present on AML cells, through specific panels of fluorochrome-labeled monoclonal antibodies [1]. Recent advances, based on the development of multicolor panels (>8 colors), brought the levels of sensitivity of cytometric analysis closer to those obtained by molecular biology techniques [2][3]. Thus, MRD analysis based on MCF presents moderate sensitivity, with a LOD of 1:10−3 up to 1:10−4 (1 leukemic cell in 10,000 to 10,000 WBCs), which is dependent on the number of cells analyzed, the gating strategy, and the number of antibody markers used [4][5][6]. The main advantage of MCF over other techniques is the ability to characterize millions of cells in a short period of time, while distinguishing viable cells from debris and dead cells [7][8][9]. Furthermore, it has the advantage of being applicable to the vast majority of AML patients, unlike other techniques that focus on the genetic and/or molecular signature [6].
MRD condition can be monitored in AML patients by MFC, assessing the abnormal expression of immunophenotypic markers that provide the distinction of the leukemic cells, also called leukemia-associated immunophenotypes (LAIPs)  [10][11]. Conventionally, for the detection of LAIPs, several markers can be used, including CD34, CD117, CD45, CD33, CD13, CD56, CD7, HLA-DR (for MRD), among others [4]. Much work has been completed to improve this strategy and its application in MRD. In most of the cases, the improvements consist of the combination of several antibodies with different fluorophores (at least eight) to detect more than one LAIP at a time, improving the diagnostic performance [12][13].
Several other studies were conducted to understand the ability and efficiency of cytometric analysis to detect MRD. For example, a study with 451 BM samples from adult AML patients assessed the clinical utility of MRD detection, demonstrating that MFC is useful for outcome prediction and guidance of post-remission in AML [14].
The biggest drawback of this technique is the requirement of significant technical expertise and the intervention of an interpreting pathologist. The future use of artificial intelligence to reduce potential bias and/or human interpretive errors can bring accurate interpretations of the results. The low sensitivity and the high change of the aberrant immunophenotype during disease progression makes the assessment of MRD more difficult, leading to a high rate of false negatives and limiting the detection of the different aberrant markers.

2. Molecular-Based Techniques

Molecular techniques are used to assess the presence of MRD by analyzing well-known AML mutations [4][5]. The most common markers used to monitor MRD are NPM1 insertion, CFB-MYH11, AML1-ETO, and PML-RARA (chimeric fusion genes) [15]. NPM1 is one of the most prevalent mutations in AML (present in 25–35% of patients) and is reported as a definitive leukemia-founder mutation that has already been validated as an optimal and reliable MRD marker [16][17]. Recent studies are focused on introducing/validating new markers for MRD monitoring. For example, many studies are now focused on the relevance of IDH mutations [4]. IDH1 and IDH2 mutations have been shown to be prevalent in AML (occurring in ~20% of cases), but there are still conflicting data, whether they represent pre-leukemic mutations or dominant clonal mutations, hence their value in monitoring MRD is not yet well established [2][16][18][19]. Moreover, NPM1 is frequently one of the references for the studies involving IDH mutations [20][21].
Furthermore, the prognostic significance of combined mutations, such as NPM1, FLT3, DNMT3A, and IDH is still unclear. However, recent studies state that some combinations may have an impact on the risk of relapse and overall survival of AML patients compared with other specific combinations [22][23][24]. Thus, in AML disease, and even in the MRD condition, it is crucial to use reliable and sensitive molecular techniques to detect these mutations, even when present in small amounts, in order to have a better understanding of their prognostic value.

2.1. Reverse Transcription Polymerase Chain Reaction (RT-PCR)

In AML, the development of RT-PCR has been crucial to detect residual elements conventionally characterized by molecularly cloned chromosome translocations [25][26]. The use of RT-PCR enables the detection of the fusion genes RUNX1-RUNX1T1 and CBFB-MYH11 [27] as well as PML-RARA [28][29] and mutated-NPM1 [30][31], important for the sensitive detection and quantification of MRD in AML [2][32]. In fact, in the case of CBF fusion transcripts (RUNX1-RUNX1T1 and CBFB-MYH11), some studies in peripheral blood (PB) or bone marrow (BM) have demonstrated the prognostic value of detection and quantification of MRD at specific time points allowing the identification of patients with a high risk of relapse [27][33]. In addition, several studies have investigated the clinical implications of monitoring NPM1 and its association with relapse and survival rates [34][35]. In the study performed by Ivey et al., using BM and PB of the AML patients, the presence of NPM1-mutated transcripts after the second chemotherapy cycle was associated with a significantly higher relapse risk and poorer survival rates, independent of other known prognostic factors [34].
Given the high specificity and sensitivity for LSCs, the decreased risk of contamination and better evaluation of RNA quality, the technique of RT-PCR has been widely implemented for routine patient care [5]. However, the detection of MRD based on RT-PCR or PCR is currently limited to around 50% of patients, since not all patients carry the fusion transcript RUNX1-RUNX1T1 (characteristic of t[8;21]), CBFB-MYH11 [inv 16] or t[16;16]), or NPM1 mutations [36]. The optimized RT-PCR assays presents a limit of detection (LOD) of 10−4 to 10−6, and is therefore more sensitive or equal than other technologies used, such as MFC (range 1:10−3 up to 1:10−4 ) [2][37][38][39].
As is the case with any other technique, there are some limitations of RT-PCR-based MRD tests, namely their dependence on specific mutations, requiring individual standard reference curves based on serial dilutions of targets, intensive labour, expertise, cost, time-consuming, and computationally demanding work [3][40].

2.2. Next-Generation Sequencing (NGS)

Next-generation sequencing (NGS) refers to the deep, high-throughput, in-parallel DNA sequencing technologies that were developed in the decades after 1977, when the Sanger DNA sequencing method was developed. Unlike Sanger sequencing, NGS procedures provide a massive parallel and extremely high-throughput analysis (millions to billions of nucleotides) of multiple samples, bringing much faster results (due to multiplexing), and at a lower cost than individual testing [41]. Therefore, NGS is a tool that easily assesses genomic, transcriptomic, and epigenomic features [42]. More specifically, in the AML context, NGS has extreme relevance for diagnosis, due to the high clonal heterogeneity characteristic of this disease [43]. It provides information about the different AML mutations present in a patient, providing more information and allowing the design of personalized therapies, which will ultimately result in a better prognosis of remission and less cases of relapse [3]. It was already described that for patients in CR, the estimated percentage of the MRD measured by NGS was much greater than of aberrant blasts detected by MFC [44].
This technology has also revealed a set of mutations in rare mutant cells and gene sequences, as well as the discovery of genetic alterations that occur between diagnosis and relapse times [45]. In a study with a cohort of 482 patients, at least one mutation was detected in 430 patients (89.2%) showing the broad clinical application of targeted NGS [46]. Additionally, NGS showed a significant additive prognostic value compared to flow cytometry for the detection of MRD [46].
Recently, Alonso et al. tested a 19-gene AML-targeted NGS in a small cohort of 162 patients and showed that well-defined NGS panels are reliable in guiding clinical decisions by the current standards. Results had a 100% correlation with conventional molecular biology techniques, and all patients were successfully classified according to 2016 WHO classification systems (2016 WHO diagnostic categories, 2017 European LeukemiaNet, and Genomic classification) [47]. NGS was also explored in the context of allogeneic hematopoietic cell transplantation (alloHCT). For example, Thol et al. published a clinical study with a cohort of more than one hundred patients, demonstrating that MRD detection utilizing NGS before alloHCT is highly predictive of relapse and survival, and can therefore improve patient management [48]. It is worth noting that similar results were obtained from both BM and PB samples, demonstrating the usefulness of using less invasive body fluids [48]. Thus, through the molecular characterization of AML cells, NGS promotes a personalized and precise method for the assessment of MRD, since can reach a sensitivity of 10−4 to 10−5 [95, 132]. However, the widespread use of NGS has been limited due to its high cost, and the expertise required for the data analysis and interpretation [49][50]. Furthermore, other drawbacks of this technique are the non-standardization of the assay, the high variance of sensitivity across different platforms, and a long waiting time for the result analysis.
In summary, MRD has been shown to be an important and independent prognostic factor and predictor of relapse in AML, and also plays a crucial role in the design of personalized treatment strategies. However, when values of cells are lower than 10−5, it is generally more challenging to be detectable by the diagnostic procedures used in clinical settings. Thus, the development of novel techniques, such as microfluidics and surface-enhanced Raman scattering spectroscopy, has the potential to enable a more reliable and satisfactory detection of MRD and CR as well as the stratification of patients into different subtypes. Yet, the sensitivity of these techniques is still being optimized, and their implementation in clinical routine is pending. Despite the technological challenges, it remains crucial to develop new technologies for MRD detection and quantification that are more affordable, faster, and offer higher accuracy and sensitivity.

References

  1. Tomlinson, B.; Lazarus, H.M. Enhancing acute myeloid leukemia therapy—Monitoring response using residual disease testing as a guide to therapeutic decision-making. Expert Rev. Hematol. 2017, 10, 563–574.
  2. 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.
  3. Ravandi, F.; Walter, R.B.; Freeman, S.D. Evaluating measurable residual disease in acute myeloid leukemia. Blood Adv. 2018, 2, 1356–1366.
  4. 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.
  5. Voso, M.T.; Ottone, T.; Lavorgna, S.; Venditti, A.; Maurillo, L.; Lo-Coco, F.; Buccisano, F. MRD in AML: The Role of New Techniques. Front. Oncol. 2019, 9, 655.
  6. Short, N.J.; Ravandi, F. How close are we to incorporating measurable residual disease into clinical practice for acute myeloid leukemia? Haematologica 2019, 104, 1532–1541.
  7. Brooimans, R.A.; van der Velden, V.H.J.; Boeckx, N.; Slomp, J.; Preijers, F.; te Marvelde, J.G.; Van, N.M.; Heijs, A.; Huys, E.; van der Holt, B. Immunophenotypic measurable residual disease (MRD) in acute myeloid leukemia: Is multicentric MRD assessment feasible? Leuk. Res. 2019, 76, 39–47.
  8. Freeman, S.D.; Virgo, P.; Couzens, S.; Grimwade, D.; Russell, N.; Hills, R.K.; Burnett, A.K. Prognostic relevance of treatment response measured by flow cytometric residual disease detection in older patients with acute myeloid leukemia. J. Clin. Oncol. 2013, 31, 4123–4131.
  9. Buccisano, F.; Maurillo, L.; Spagnoli, A.; Del Principe, M.I.; Fraboni, D.; Panetta, P.; Ottone, T.; Consalvo, M.I.; Lavorgna, S.; Bulian, P.; et al. Cytogenetic and molecular diagnostic characterization combined to postconsolidation minimal residual disease assessment by flow cytometry improves risk stratification in adult acute myeloid leukemia. Blood 2010, 116, 2295–2303.
  10. Sui, J.-N.; Chen, Q.-S.; Zhang, Y.-X.; Sheng, Y.; Wu, J.; Li, J.-M.; Weng, X.-Q.; 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.
  11. 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.
  12. Rasheed, H.M.; Donia, H.M.; Nadwan, E.A.; Mourad, Z.I.; Farahat, N. Identifying leukemia-associated immunophenotypes in acute myeloid leukemia patients using multiparameter flow cytometry. Oman Med. J. 2021, 36, e323.
  13. Kern, W.; Voskova, D.; Schoch, C.; Hiddemann, W.; Schnittger, S.; Haferlach, T. Determination of relapse risk based on assessment of minimal residual disease during complete remission by multiparameter flow cytometry in unselected patients with acute myeloid leukemia. Blood 2004, 104, 3078–3085.
  14. Patkar, N.; Kakirde, C.; Bhanshe, P.; Joshi, S.; Chaudhary, S.; Badrinath, Y.; Ghoghale, S.; Deshpande, N.; Kadechkar, S.; Chatterjee, G.; et al. Utility of Immunophenotypic Measurable Residual Disease in Adult Acute Myeloid Leukemia-Real-World Context. Front. Oncol. 2019, 9, 450.
  15. Cruz, N.M.; Mencia-Trinchant, N.; Hassane, D.C.; Guzman, M.L. Minimal residual disease in acute myelogenous leukemia. Int. J. Lab. Hematol. 2017, 39, 53–60.
  16. Kövy, P.; Őrfi, Z.; Bors, A.; Kozma, A.; Gopcsa, L.; Dolgos, J.; Lovas, N.; Harasztdombi, J.; Lakatos, V.; Király, Á. Nucleophosmin1 and isocitrate dehydrogenase 1 and 2 as measurable residual disease markers in acute myeloid leukemia. PLoS ONE 2021, 16, e0253386.
  17. Hindley, A.; Catherwood, M.A.; McMullin, M.F.; Mills, K.I. Significance of NPM1 Gene Mutations in AML. Int. J. Mol. Sci. 2021, 22, 10040.
  18. Grimwade, D.; Ivey, A.; Huntly, B.J.P. Molecular landscape of acute myeloid leukemia in younger adults and its clinical relevance. Blood 2016, 127, 29–41.
  19. Debarri, H.; Lebon, D.; Roumier, C.; Cheok, M.; Marceau-Renaut, A.; Nibourel, O.; Geffroy, S.; Helevaut, N.; Rousselot, P.; Gruson, B.; et al. IDH1/2 but not DNMT3A mutations are suitable targets for minimal residual disease monitoring in acute myeloid leukemia patients: A study by the Acute Leukemia French Association. Oncotarget 2015, 6, 42345–42353.
  20. Jeziskova, I.; Razga, F.; Toskova, M.; Dvorakova, D.; Timilsina, S.; Mayer, J.; Racil, Z. Quantitative detection of IDH2 mutation for minimal residual disease monitoring in patients with acute myeloid leukemia and its comparison with mutations in NPM1 gene. Leuk. Lymphoma 2013, 54, 867–870.
  21. Petrova, L.; Vrbacky, F.; Lanska, M.; Zavrelova, A.; Zak, P.; Hrochova, K. IDH1 and IDH2 mutations in patients with acute myeloid leukemia: Suitable targets for minimal residual disease monitoring? Clin. Biochem. 2018, 61, 34–39.
  22. Abd El-Lateef, A.E.; Ismail, M.M.; Almohammadi, M.; Gawaly, A.M. Prognostic relevance of combined IDH1 and NPM1 mutations in the intermediate cytogenetic de novo acute myeloid leukemia. Cell. Mol. Biol. 2021, 67, 92–98.
  23. Paschka, P.; Schlenk, R.F.; Gaidzik, V.I.; Habdank, M.; Krönke, J.; Bullinger, L.; Späth, D.; Kayser, S.; Zucknick, M.; Götze, K.; et al. IDH1 and IDH2 Mutations Are Frequent Genetic Alterations in Acute Myeloid Leukemia and Confer Adverse Prognosis in Cytogenetically Normal Acute Myeloid Leukemia With NPM1 Mutation Without FLT3 Internal Tandem Duplication. J. Clin. Oncol. 2010, 28, 3636–3643.
  24. Dunlap, J.B.; Leonard, J.; Rosenberg, M.; Cook, R.; Press, R.; Fan, G.; Raess, P.W.; Druker, B.J.; Traer, E. The combination of NPM1, DNMT3A, and IDH1/2 mutations leads to inferior overall survival in AML. Am. J. Hematol. 2019, 94, 913–920.
  25. Salto-Tellez, M.; Shelat, S.G.; Benoit, B.; Rennert, H.; Carroll, M.; Leonard, D.G.B.; Nowell, P.; Bagg, A. Multiplex RT-PCR for the detection of leukemia-associated translocations: Validation and application to routine molecular diagnostic practice. J. Mol. Diagn. 2003, 5, 231–236.
  26. Wertheim, G.B.W. Molecular characterization and testing in acute myeloid leukemia. J. Hematop. 2015, 8, 177–189.
  27. 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 in CBFB-MYH11–Positive Acute Myeloid Leukemia. J. Clin. Oncol. 2010, 28, 3724–3729.
  28. Cicconi, L.; Lo-Coco, F. Current management of newly diagnosed acute promyelocytic leukemia. Ann. Oncol. 2016, 27, 1474–1481.
  29. Sanz, M.A.; Grimwade, D.; Tallman, M.S.; Lowenberg, B.; Fenaux, P.; Estey, E.H.; Naoe, T.; Lengfelder, E.; Büchner, T.; Döhner, H.; et al. Management of acute promyelocytic leukemia: Recommendations from an expert panel on behalf of the European LeukemiaNet. Blood 2009, 113, 1875–1891.
  30. DeAngelo, D.J.; Stein, E.M.; Ravandi, F. Evolving Therapies in Acute Myeloid Leukemia: Progress at Last? Am. Soc. Clin. Oncol. Educ. B 2016, 36, e302–e312.
  31. Schnittger, S.; Schoch, C.; Kern, W.; Mecucci, C.; Tschulik, C.; Martelli, M.F.; Haferlach, T.; Hiddemann, W.; Falini, B. Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype. Blood 2005, 106, 3733–3739.
  32. Krönke, J.; Schlenk, R.F.; Jensen, K.-O.; Tschürtz, F.; Corbacioglu, A.; Gaidzik, V.I.; Paschka, P.; Onken, S.; Eiwen, K.; Habdank, M.; et al. Monitoring of Minimal Residual Disease in NPM1-Mutated Acute Myeloid Leukemia: A Study From the German-Austrian Acute Myeloid Leukemia Study Group. J. Clin. Oncol. 2011, 29, 2709–2716.
  33. Jourdan, E.; Boissel, N.; Chevret, S.; Delabesse, E.; Renneville, A.; Cornillet, P.; Blanchet, O.; Cayuela, J.-M.; Recher, C.; Raffoux, E. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood J. Am. Soc. Hematol. 2013, 121, 2213–2223.
  34. Ivey, A.; Hills, R.K.; Simpson, M.A.; Jovanovic, J.V.; Gilkes, A.; Grech, A.; Patel, Y.; Bhudia, N.; Farah, H.; Mason, J. Assessment of minimal residual disease in standard-risk AML. N. Engl. J. Med. 2016, 374, 422–433.
  35. 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 J. Am. Soc. Hematol. 2009, 114, 2220–2231.
  36. Platzbecker, U.; Middeke, J.M.; Sockel, K.; Herbst, R.; Wolf, D.; Baldus, C.D.; Oelschlägel, U.; Mütherig, A.; Fransecky, L.; Noppeney, R.; et al. Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): An open-label, multicentre, phase 2 trial. Lancet Oncol. 2018, 19, 1668–1679.
  37. Del Principe, M.I.; Buccisano, F.; Maurillo, L.; Sconocchia, G.; Cefalo, M.; Consalvo, M.I.; Sarlo, C.; Conti, C.; De Santis, G.; De Bellis, E.; et al. Minimal Residual Disease in Acute Myeloid Leukemia of Adults: Determination, Prognostic Impact and Clinical Applications. Mediterr. J. Hematol. Infect. Dis. 2016, 8, e2016052.
  38. Leroy, H.; de Botton, S.; Grardel-Duflos, N.; Darre, S.; Leleu, X.; Roumier, C.; Morschhauser, F.; Lai, J.-L.; Bauters, F.; Fenaux, P.; et al. Prognostic value of real-time quantitative PCR (RQ-PCR) in AML with t(8;21). Leukemia 2005, 19, 367–372.
  39. Colmenares, R.; Álvarez, N.; Barrio, S.; Martínez-López, J.; Ayala, R. The Minimal Residual Disease Using Liquid Biopsies in Hematological Malignancies. Cancers 2022, 14, 1310.
  40. Nolan, T.; Hands, R.E.; Bustin, S.A. Quantification of mRNA using real-time RT-PCR. Nat. Protoc. 2006, 1, 1559.
  41. Kulski, J.K. Next-Generation Sequencing—An Overview of the History, Tools, and “Omic” Applications; IntechOpen: Rijeka, Croatia, 2016; p. 61964.
  42. Hwang, B.; Lee, J.H.; Bang, D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp. Mol. Med. 2018, 50, 96.
  43. Leisch, M.; Jansko, B.; Zaborsky, N.; Greil, R.; Pleyer, L. Next Generation Sequencing in AML-On the Way to Becoming a New Standard for Treatment Initiation and/or Modulation? Cancers 2019, 11, 252.
  44. Getta, B.M.; Devlin, S.M.; Levine, R.L.; Arcila, M.E.; Mohanty, A.S.; Zehir, A.; Tallman, M.S.; Giralt, S.A.; Roshal, M. Multicolor Flow Cytometry and Multigene Next-Generation Sequencing Are Complementary and Highly Predictive for Relapse in Acute Myeloid Leukemia after Allogeneic Transplantation. Biol. Blood Marrow Transplant. 2017, 23, 1064–1071.
  45. Vosberg, S.; Greif, P.A. Clonal evolution of acute myeloid leukemia from diagnosis to relapse. Genes. Chromosomes Cancer 2019, 58, 839–849.
  46. Jongen-Lavrencic, M.; Grob, T.; Hanekamp, D.; Kavelaars, F.G.; al Hinai, A.; Zeilemaker, A.; Erpelinck-Verschueren, C.A.J.; 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.
  47. Alonso, C.M.; Llop, M.; Sargas, C.; Pedrola, L.; Panadero, J.; Hervás, D.; Cervera, J.; Such, E.; Ibáñez, M.; Ayala, R.; et al. Clinical Utility of a Next-Generation Sequencing Panel for Acute Myeloid Leukemia Diagnostics. J. Mol. Diagnostics 2019, 21, 228–240.
  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.
  49. Aitken, M.J.L.; Ravandi, F.; Patel, K.P.; Short, N.J. Prognostic and therapeutic implications of measurable residual disease in acute myeloid leukemia. J. Hematol. Oncol. 2021, 14, 137.
  50. Döhner, H.; Weisdorf, D.J.; Bloomfield, C.D. Acute Myeloid Leukemia. N. Engl. J. Med. 2015, 373, 1136–1152.
More
Upload a video for this entry
Information
Subjects: Hematology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , Sara Abalde-Cela , Belém Sampaio-Marques , Anabela C. Areias , Paula Ludovico , Lorena Diéguez
View Times: 619
Revisions: 2 times (View History)
Update Date: 27 Mar 2023
Notice
You are not a member of the advisory board for this topic. If you want to update advisory board member profile, please contact office@encyclopedia.pub.
OK
Confirm
Only members of the Encyclopedia advisory board for this topic are allowed to note entries. Would you like to become an advisory board member of the Encyclopedia?
Yes
No
${ textCharacter }/${ maxCharacter }
Submit
Cancel
There is no comment~
${ textCharacter }/${ maxCharacter }
Submit
Cancel
${ selectedItem.replyTextCharacter }/${ selectedItem.replyMaxCharacter }
Submit
Cancel
Confirm
Are you sure to Delete?
Yes No
Academic Video Service