Next Article in Journal
Intelligent Identification of Early Esophageal Cancer by Band-Selective Hyperspectral Imaging
Previous Article in Journal
A Subset of Secreted Proteins in Ascites Can Predict Platinum-Free Interval in Ovarian Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MRD Monitoring by Multiparametric Flow Cytometry in AML: Is It Time to Incorporate Immune Parameters?

by
Ilias Pessach
1,
Theodoros Spyropoulos
2,
Eleftheria Lamprianidou
2 and
Ioannis Kotsianidis
2,*
1
Department of Hematology, Athens Medical Center, 11634 Athens, Greece
2
Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 69100 Alexandroupolis, Greece
*
Author to whom correspondence should be addressed.
Submission received: 9 August 2022 / Revised: 27 August 2022 / Accepted: 29 August 2022 / Published: 1 September 2022

Abstract

:

Simple Summary

Measurable residual disease (MRD) is emerging as an important prognostic and predictive biomarker in acute myeloid leukemia (AML). However, its use is currently hampered by the disparity and lack of harmonization between the available MRD methodologies. In addition, the current assessment of MRD in AML focuses only on the quantification of the residual leukemic burden, without addressing the parallel alterations of the antineoplastic immune response that can critically affect the course and outcome of AML, often despite MRD persistence. Incorporating parameters of immune competence provides more consistency with the biological concept of MRD and may lead to higher accuracy. Multiparameter flow cytometry (MFC) is a highly efficacious and sensitive technology for the thorough and synchronous investigation of the kinetics of both antitumor immunity and the leukemic clone. MFC-based MRD provides the platform for the development of a composite leukemia- and immune-based biomarker which can outcompete the current MRD assessment.

Abstract

Acute myeloid leukemia (AML) is a heterogeneous group of clonal myeloid disorders characterized by intrinsic molecular variability. Pretreatment cytogenetic and mutational profiles only partially inform prognosis in AML, whereas relapse is driven by residual leukemic clones and mere morphological evaluation is insensitive for relapse prediction. Measurable residual disease (MRD), an independent post-diagnostic prognosticator, has recently been introduced by the European Leukemia Net as a new outcome definition. However, MRD techniques are not yet standardized, thus precluding its use as a surrogate endpoint for survival in clinical trials and MRD-guided strategies in real-life clinical practice. AML resistance and relapse involve a complex interplay between clonal and immune cells, which facilitates the evasion of the leukemic clone and which is not taken into account when merely quantifying the residual leukemia. Multiparameter flow cytometry (MFC) offers the possibility of capturing an overall picture of the above interactions at the single cell level and can simultaneously assess the competence of anticancer immune response and the levels of residual clonal cells. In this review, we focus on the current status of MFC-based MRD in diverse AML treatment settings and introduce a novel perspective of combined immune and leukemia cell profiling for MRD assessment in AML.

1. Introduction

What is actually “remission” in AML? Acute myeloid leukemia (AML) is a group of heterogeneous clonal myeloid disorders encompassing a wide range of molecular alterations, as reflected by the plethora of disease subtypes in the current WHO classification [1]. AML particularly impacts older adults conveying a poor prognosis and only 35–40% of patients under 60 years of age and 5–15% of those older than 60 years of age will attain long term remissions [1]. Intensive chemotherapy has been the standard tool to fight AML for many decades, and in several cases the cytotoxic therapy is followed by allogeneic stem cell transplantation (alloSCT) [2]. A wide range of pretreatment cytogenetic and mutational profiles informs prognosis in AML and allows for categorization of patients with adverse, intermediate, or favorable-risk disease as outlined by the European Leukemia Net (ELN) criteria [3]. However, the current prognostic assessment of AML cannot precisely predict the curative potential of chemotherapy alone or accurately estimate the benefit of alloSCT strategies. In addition, the advent of targeted therapies has, in several cases, disconnected the quality of response from survival benefit, thus adding another layer of complexity to the prognostic assessment of AML [4,5]. According to the latest ELN criteria [6] an AML patient is considered to be in complete remission (CR) if all of the above criteria are fulfilled: bone marrow blasts <5%; absence of circulating blasts or blasts with Auer rods; absence of extramedullary disease; ANC ≥1.0 × 109/L (1000/μL); and platelet count ≥100 × 109/L (100,000/μL).
Complete remission (CR) achievement is a necessity for the cure and prolonged survival of AML patients. However, as morphological remission has long been known to be insensitive for relapse prediction and approximately 50% patients who achieve CR eventually relapse, the notion of “measurable residual disease” negativity (MRD-negative CR) has recently been introduced by the ELN as a new outcome definition [7,8]. MRD indicates the presence of leukemia cells at frequencies below that of routine measurement, either by morphology or cytogenetics, with sensitivity down to one in 10−4 to one in 10−6 of total leukocytes compared to one in 20 in standard morphology [9]. The added prognostic value of MRD in patients in morphologic CR is well established [8,9,10,11]; however, the lack of standardization and comparability among the various methodologies and the reporting of results currently precludes, in most circumstances, leveraging MRD-guided strategies (Table 1).

2. Multiparametric Flow Cytometry-MRD (MFC-MRD) Testing in AML

The investigation and use of cell surface antigens to differentiate normal hematopoietic cells from leukemic blasts started decades ago. Baker et al., in 1974, introduced a rabbit-derived antiserum that was non-reactive with normal cells but highly reactive with leukemic blasts from patients with ALL and AML, presenting evidence that antigenic differences existed between leukemic and non-leukemic cells [16]. Greaves et al. first proposed that the use of immunofluorescent reagents and antibodies directed against leukemia-associated antigens could detect residual leukemic cells, helping to monitor patients for early signs of relapse [17].
The main advantages of MFC-MRD in AML, namely the high applicability (>90%), short turnaround time, relatively high sensitivity, and the possibility to discriminate between living and dead cells [7,8,12,18,19], make MFC-MRD the ideal method to target real-time therapeutic clinical decision making [9,13,14,15]. The sensitivity of the MFC-based MRD assessment is 10−3 to 10−5 depending on the method, the number of cells analyzed, the design of the panel, the instrument settings, the number and type of antibodies used, and a clinically validated cut-off point for MRD positivity [7,8,11]. The predictive value of MFC-MRD assessment in patients with AML has been studied in several settings, but most data are currently derived from studies with intensive cytotoxic regimens and in settings with allogeneic stem cell transplantation, whereas data on low-intensity therapies are still limited [11] (Table 2).

2.1. MFC-MRD after Intensive Chemotherapy

The pediatric protocol AML02 investigated the use of targeted chemotherapy and alloSCT, and a better clinical outcome was reported compared to previous studies. In this study, risk categories were based on the genetic/cytogenetic profile of the patients and MRD findings. FCM-MRD was applied after the first cycle of chemotherapy and treatment was intensified by gemtuzumabozogamicin in patients with high levels of MRD [34]. In the international prospective pediatric study conducted by Langebrake et al., 2006 [21], it was demonstrated that the detection of residual blast cells by flow cytometry at early follow-ups (until day 84) was a significant predictor of treatment outcome, defined as 3-year EFS. The authors utilized MFC-MRD at four timepoints, namely 15 days from start of therapy, before the second induction and before the first and second consolidation course. Already in the early course of therapy before the start of the second induction, at day 28 from diagnosis, MFC-MRD exhibited high prediction accuracy. Coustan-Smith et al. introduced the analysis of residual blasts at the end of remission induction therapy using a specific panel of markers/combinations based on the initial immunophenotype. They established and validated a method to monitor residual disease in children with AML, and introduced immunophenotypes suitable for the detection of residual disease that can be identified by four-color flow cytometry, which allows for sensitivity of detection to be as much as 100 times greater compared to morphological examination [25]. Flow cytometric evidence of leukemia after the initiation of therapy seems to be an independent prognostic factor associated with poor outcome in pediatric studies [20].
Regarding adult patients, in the study by Liu et al. [22], MFC-MRD status after two consolidation cycles had greater accuracy in predicting relapse compared to MRD status after induction. This result was consistent for all different sub-groups (young vs. elderly patients, ELN cytogenetic low- or intermediate-risk patients). By contrast, Minetto et al., 2021 [37], reported that an earlier MRD evaluation timepoint may provide the most significant information on outcome in cases of induction regimens with the combination of fludarabine plus high dose cytarabine. MRD monitoring indicated that patients who had detectable leukemia after induction but achieved MRD negativity after the second consolidation had the same prognosis as those with a negative MRD at both timepoints. Similarly, in the HOVON/SAKK (Dutch–Belgian Haemato-Oncology Cooperative Group and the Swiss Group for Clinical Cancer Research) AML42a study, a prospective analysis of MRD using Leukemia-Associated Immunophenotypes (LAIPs), MRD positivity after cycle 2 was associated with a higher risk of relapse, with 4-year relapse-free survival (RFS) of 23% and relapse incidence of 72% compared to 52% and 42%, respectively, for MRD-negative patients [26].
The QUAZAR AML-001 Maintenance Trial is the first large prospective, double-blind, placebo-controlled randomized trial with long-term longitudinal assessment of MRD in patients with AML in remission. In this trial, administration of the oral formulation of azacytidine (CC-486) as maintenance was associated with significantly longer overall and relapse-free survival than placebo among older patients with AML who were in remission after chemotherapy. In both treatment arms, MRD-positive status (≥0.1%) after induction ± consolidation was associated with significantly shorter OS and RFS compared to MRD-negative status [38].

2.2. MFC-MRD after Lower Intensity Treatment

The prognostic value of MRD assessment after intensive chemotherapy and allogeneic hematopoietic stem cell transplantation (SCT) has been widely studied. Lower intensity therapy (LIT) has been introduced for the treatment of older or so-called “unfit/frail” patients and, apart from the improved tolerability, it has resulted in longer overall survival compared to conventional care regimens [39]
The MFC-MRD-based prediction of prognosis in older patients with AML who are not eligible to receive intensive chemotherapy was investigated in many clinical trials [30]. Maiti et al. [31] analyzed the prognostic value of achieving negative MRD in this group of patients receiving first-line therapy, i.e., DEC10-VEN regimens. The achievement of MRD-negative status at 1 and 2 months after starting therapy was associated with better OS in older patients with AML with intermediate- and adverse-risk cytogenetics. MRD-negative status at 1, 2, and 4 months after starting therapy was associated with significantly better survival in older/unfit patients with AML.
The combination of venetoclax plus hypomethylating agents seems to be an effective treatment regimen leading to improvements in complete remission rates and overall survival. In a single center phase II study in patients with newly diagnosed (ND) AML older than 60 years of age, secondary AML (sAML), and relapsed or refractory (R/R) AML, the administration of azacytidine with oral venetoclax was highly efficacious and safe [40,41]. The value of MRD assessment was again evident as patients with composite complete remission with negative MFC-MRD based on the 0.1% cut-off had comparable 2-year OS regardless of treatment type (73.6% in the azacitidine–venetoclax group and 63.6% in the placebo group). In addition, for MRD-negative patients in the venetoclax arm, the median EFS and OS were not reached in patients with CR and MRD < 0.1%, whereas the ones with MRD > 0.1% had median EFS and OS at 10.6 and 18.7 months, respectively [32].
The PETHEMA-FLUGAZA phase III trial in elderly AML patients reported that MFC-MRD status was the only independent predictor of relapse-free survival in 72 patients who achieved CR either by the combination of low-dose cytarabine plus fludarabine or azacytidine at standard doses. However, even patients with MRD negativity exhibited substantial genetic abnormalities in CD34+ progenitors, indicative of the inability of the current MRD threshold to detect the existence of very low levels of clonal cells that can drive relapse [42].

2.3. MFC-MRD Prior to and after AlloSCT

In a meta-analysis of large-scale AML studies reported by Short et al., 2020, patients with MRD negativity had improved rates of overall survival (OS, 68% vs. 34%) and disease-free survival (DFS, 64%vs. 25%) at 5 years versus patients with MRD positivity [29]. Patients with MRD positivity may benefit from pre-transplantation strategies and MRD assessment may contribute to choosing the appropriate conditioning regimen for alloSCT [33,34]. Hourigan et al. demonstrated that reduced-intensity conditioning resulted in worse outcomes when compared to full conditioning in patients with MRD positivity [43]. Various studies showed that both pediatric and adult populations with AML who retain MRD positivity after induction treatment display a worse outcome following alloSCT compared to patients with MRD negativity. In a meta-analysis performed by Buckley et al., 2017, MRD positivity before alloSCT was associated with decreased leukemia-free survival (LFS, hazard ratio [HR] = 2.76), OS (HR = 2.36), and cumulative incidence of relapse (CIR, HR = 3.65) [44].
In the GIMEMA adult AML1310 trial [24], patients with intermediate-risk AML received either autologous-SCT or alloSCT depending on the level of FCM-MRD (threshold of 0.035%). Overall and disease-free survival (DFS) at 24 months was assessed, with values of 78.6% and 61.4% in MRD-positive and 69.8% and 66.6% in MRD-negative patients, respectively.
The National Cancer Research Institute’s AML17 trial is currently the largest study to investigate the key elements impacting MRD in AML. A total of 1874 adults < 60 years of age with AML were enrolled and treated with standard daunorubicin plus cytarabine based induction treatment followed by risk-adapted chemotherapy consolidation, with or without alloSCT. Patients with MRD positivity (defined as ≥0.1% by the MFC assay) after cycle 1 had similar 5-year overall survival as patients who only achieved a partial response (51% vs. 46%, respectively), emphasizing the poor outcomes associated with persistent MRD [11]. Other studies have also shown that MRD persistence prior to alloSCT has a negative predictive value, while MRD negativity in the same setting leads to favorable outcomes (3-year overall survival estimates of >70% and a relapse risk of 20–25%) after myeloablative alloSCT [35,45]. Interestingly, post-alloSCT MRD status appeared to be more informative than pre-alloSCT status in the study by Zhou et al. [36]. The 3-year overall survival rate for patients with pre-alloSCT MRD that persisted after the transplant was 19%, whereas in patients with pre-alloSCT MRD that cleared with transplantation it was 29%. Monitoring for residual disease post-alloSCT can be predictive of relapse and monthly monitoring during the first 6 months has been proposed for the early detection of relapse [27].
Of note, the combination of MFC-MRD with mutational analysis using NGS or digital PCR increased the sensitivity for the diagnosis significantly [28]. The first study to show that a targeted multigene NGS panel can be used to detect residual mutations in AML patients immediately before allogeneic transplantation was conducted by Getta et al. [23], who compared MFC-MRD with a multigene NGS assay for the first time and showed that the presence of residual mutations and atypical blasts is associated with post-transplantation relapse and survival. The burden of residual disease, as measured by residual leukemia alleles, was significantly higher than the percentage of aberrant blasts assessed by MFC, indicating the presence of residual leukemia alleles in non-blast compartments at the time of remission.

3. Future Perspective: A Holistic Approach for MFC-MRD in AML

As MRD is becoming an invaluable outcome predictor for AML, the need for standardization and improvement of sensitivity and specificity of the current methods is imperative. Crucial limitations still exist, as reflected by the fact that the false-negative rates of MFC-MRD in AML vary from 13–30% and that up to 70% of patients with low/negative MRD, based on the 0.1% cut-off, will finally relapse [46,47,48].
Beyond the technological and methodological limitations of MRD evaluation, there are other issues pertaining to the biological concept of MRD. Merely quantitative measurement of the residual leukemic cells, although established in several clinical settings [3,49], has obvious drawbacks. The individual properties of the remaining leukemic clone(s), namely the refractoriness and leukemogenic potential, are not fully addressed with the current panels of MFC-MRD, whereas persistent or donor-derived clonal hematopoiesis in the case of alloSCT is often difficult to distinguish from true residual AML [50]. Surface phenotypic characterization of leukemic subpopulations correlates poorly with the functional status of the latter and cannot dissect the wide inter- and intratumor heterogeneity of AML. Interrogation of intracellular signaling pathways, and molecular studies in xenograft models [51] revealed a decoupling of the surface phenotype of leukemic progenitors from their leukemia-regenerating capacity, indicating that MRD based solely on conventional surface phenotyping cannot reliably estimate the leukemogenic potential of the residual clonal cells.
An even more important issue is the fact that the mere measurement of residual leukemic burden does not take into account tumor–immune system interactions. Oncogenesis is not merely a cell-intrinsic process but involves complex interactions of the malignant clone with neighboring non-clonal cells [52]. The patient’s immune system can suppress but also promote cancerous growth and appears to sculpt tumor immunogenicity, as theorized by the cancer immunoediting model [53]. On the other hand, malignant neoplasms subvert the anti-tumor immune response by employing a myriad of mechanisms involving cellular and soluble immune elements both systemically and locally [54,55]. The ultimate consequence of this process is the development of resistant survivor clones that drive disease relapse after MRD “negativity”, as has been demonstrated in preclinical models [56,57]. The prognostic value of tumor-infiltrating T cell subsets is well established in solid tumors [58] but has not yet been incorporated in the risk assessment of AML, despite existing evidence for the prognostic relevance of the immune biosignature of AML [59,60].
The pivotal role of crosstalk between AML and immune cells in disease relapse after an initial remission is long known, but three recent studies provide intriguing data that arouse further concerns about the validity of conventional MFC-MRD. By analyzing primary AML samples, Christofer et al. demonstrated that relapse after allogeneic hematopoietic stem cell transplantation (alloSCT) is accompanied by a theoretically reversible epigenetic dysregulation of immune pathways, which may be important in immune evasion of the leukemic clone [61]. Almost identical findings were reported by Toffalori et al. who identified a predictive transcriptional signature in AML blasts of transplanted patients, characterized by upregulation of inhibitory ligands and subversion of antigen presentation [62]. Evidently, both biosignatures cannot be traced with current MRD methods and this may, in part, account for the currently unsatisfactory accuracy of MRD testing [63]. In addition to the above findings in leukemic blasts, a third study revealed the prognostic significance of cellular and soluble immune system elements by investigating the effect of immune reconstitution after alloSCT on the outcome of patients. Mass cytometry analysis of 89 immune cell subsets and serum profiling of soluble mediators were performed longitudinally after alloSCT and prognostically relevant immune signatures were constructed [64].
Though both targeted and conventional therapies can induce immune cell priming by several mechanisms, including immunogenic cell death and eradication of the immunosuppressive tumor milieu, the increasing use of low-intensity therapies in younger patients poses another challenge. In a phase II study combining azacytidine and nivolumab in relapsed/refractory (R/R) AML, the pretherapeutic size of the bone marrow T cell pool predicted response to treatment, whereas two CD4+ and CD8+ effector subsets expanded further post-treatment [65]. In addition, recent results from our group suggest that modulation of the IL−6/STAT3 signaling axis in conventional CD4+ T cells of high risk MDS patients is strongly associated with response and outcome to azacytidine, potentially representing immune-mediated antileukemic activity of the latter [66]. Similarly, venetoclax, the standard partner of azacytidine in the treatment of elderly and/or frail AML patients, has recently been shown to induce T cell antileukemic response by increasing the intracellular levels of reactive oxygen species in conventional and CD3+CD4CD8 double-negative T cells [67]. Collectively, these data suggest the pleiotropic action of newer agents by engaging a multitude of immune-mediated mechanisms and, at least partially, explain the frequently observed delayed responses and the dissociation between clinical response and overall survival [4,5].
Two studies of multiple myeloma reported that the concomitant measurement of immune cell subsets by flow cytometry considerably improves the prognostic power of MRD in diverse disease settings [68,69]. However, the complex tumor–immune cell interactions, the interindividual variability of immune systems, and the dynamic nature of immune reconstitution after AML treatment require more in-depth analysis with functional phenotyping and molecular characterization of immune components. On the other hand, a future application of a combined myeloid and immune-based MRD should be simple, standardized, and comprehensible in order to be used as a prognostic marker and/or endpoint in clinical trials. Designing an informative panel of immune markers for MFC-MRD in AML poses an obviously huge challenge considering the vast complexity of the immune system. However, based on the aforementioned studies, the immune panel can include markers of T helper and cytotoxic cell differentiation, i.e., FOXP3 and the intracellular cytokines interferon (IFN)-γ, IL-4, and IL-17 and TGFβ, IFNα/β, γ and λ receptors, immune inhibitory receptors, and their ligands (i.e., PD1, PDL1, TIM-3, CTLA-4) and cytolytic enzymes (perforin, granzymes A and B). Functional phenotyping can also be used in both T and blast cells in order to address ROS levels and the STAT signaling biosignatures (Figure 1).

4. Conclusions

Despite its unequivocal value in treatment guidance, MRD is not yet used routinely and has not been incorporated in the treatment algorithms relating to AML. Establishing MRD negativity as a surrogate endpoint for survival will accelerate drug selection and approval, whereas in everyday practice this will spare patients unnecessary and potentially harmful treatment approaches. However, the current methods of MRD assessment are solely focused on measuring the size of the residual leukemic clones without integrating the intrinsic genetic variability of the clonal cells and/or any metric of immune competence, thus overlooking the parallel alterations of the immune milieu (Figure 1). This fact may account for the still imperfect predictive accuracy of MRD as the status of antitumor immunity can critically affect disease progression, independent of the levels of residual leukemia. For instance, the spontaneous achievement of MRD negativity in post-chemotherapy MRD+ AML patients bearing the NPM1 mutation [70] potentially mirrors the gradual eradication of the residual leukemic burden through the restoration of an effective antileukemic immune response after chemotherapy. The simultaneous assessment of the alterations of the leukemic clone and the immune system has the potential to overcome the limitations of current MRD methods. A snapshot of the antitumor immune response at relevant post-treatment timepoints provides the possibility of quantifying the competence of antitumor immunity and redefining MRD status, independent of the persistence of measurable leukemic burden.
Flow cytometry remains the key tool for the comprehensive characterization of both myeloid and immune cells and its versatility is ideal for the synchronous study of these two pivotal players in the pathobiology of AML relapse. As flow cytometric technologies are rapidly advancing, and the combined analysis of immunophenotypic, spatial, and morphological traits is becoming a reality, the parallel study [71] of the kinetics of leukemic and immune components in AML could identify a serviceable and easily applicable MRD marker which would outcompete the current flow-based MRD method.

Author Contributions

I.P., T.S., E.L. and I.K. collected the literature and data, as well as designed and wrote the article. All authors have read and agreed to the published version of the manuscript.

Funding

The study was funded by the “InTechThrace: Integrated Technologies in biomedical research: multilevel biomarker analysis in Thrace” project (MIS Code 5047285) under the Operational Program “Competitiveness, Entrepreneurship & Innovation” (EPAnEK), co-funded by the European Regional Development Fund (ERDF) and national resources (Partnership Agreement 2014–2020). No publication fees were applied by the journal.

Acknowledgments

The study was conducted by the “InTechThrace: Integrated Technologies in biomedical research: multilevel biomarker analysis in Thrace” project (MIS Code 5047285) under the Operational Program “Competitiveness, Entrepreneurship & Innovation” (EPAnEK), co-funded by the European Regional Development Fund (ERDF) and national resources (Partnership Agreement 2014–2020).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Döhner, H.; Weisdorf, D.J.; Bloomfield, C.D. Acute Myeloid Leukemia. N. Engl. J. Med. 2015, 373, 1136–1152. [Google Scholar] [CrossRef] [PubMed]
  2. Ngai, L.L.; Kelder, A.; Janssen, J.J.W.M.; Ossenkoppele, G.J.; Cloos, J. MRD Tailored Therapy in AML: What We Have Learned So Far. Front. Oncol. 2021, 10, 603636. [Google Scholar] [CrossRef] [PubMed]
  3. Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Büchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef]
  4. Nazha, A.; Sekeres, M.A.; Garcia-Manero, G.; Barnard, J.; Ali, N.H.A.; Roboz, G.J.; Steensma, D.P.; DeZern, A.E.; Zimmerman, C.; Jabbour, E.J.; et al. Outcomes of patients with myelodysplastic syndromes who achieve stable disease after treatment with hypomethylating agents. Leuk. Res. 2016, 41, 43–47. [Google Scholar] [CrossRef]
  5. Papageorgiou, S.G.; Kontos, C.K.; Kotsianidis, I.; Vasilatou, D.; Symeonidis, A.; Galanopoulos, A.; Bouchla, A.; Hatzimichael, E.; Repousis, P.; Zikos, P. The outcome of patients with high-risk MDS achieving stable disease after treatment with 5-azacytidine: A retrospective analysis of the Hellenic (Greek) MDS study group. Hematol. Oncol. 2018, 36, 693–700. [Google Scholar] [CrossRef] [PubMed]
  6. Dohner, 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 ELN Recommendations from an International Expert Panel. Blood 2022, 129, 424–447. [Google Scholar] [CrossRef] [PubMed]
  7. Dix, C.; Lo, T.-H.; Clark, G.; Abadir, E. Measurable Residual Disease in Acute Myeloid Leukemia Using Flow Cytometry: A Review of Where We Are and Where We Are Going. J. Clin. Med. 2020, 9, 1714. [Google Scholar] [CrossRef]
  8. 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]
  9. 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]
  10. Paietta, E. Consensus on MRD in AML? Blood 2018, 131, 1265–1266. [Google Scholar] [CrossRef]
  11. 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. [Google Scholar] [CrossRef] [PubMed]
  12. 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.; et al. Immunophenotypic measurable residual disease (MRD) in acute myeloid leukemia: Is multicentric MRD assessment feasible? Leuk. Res. 2019, 76, 39–47. [Google Scholar] [CrossRef] [PubMed]
  13. Maurer-Granofszky, M.; Schumich, A.; Buldini, B.; Gaipa, G.; Kappelmayer, J.; Mejstrikova, E.; Karawajew, L.; Rossi, J.; Suzan, A.Ç.; Agriello, E.; et al. An Extensive Quality Control and Quality Assurance (QC/QA) Program Significantly Improves Inter-Laboratory Concordance Rates of Flow-Cytometric Minimal Residual Disease Assessment in Acute Lymphoblastic Leukemia: An I-BFM-FLOW-Network Report. Cancers 2021, 13, 6148. [Google Scholar] [CrossRef]
  14. Wood, B.L. Principles of minimal residual disease detection for hematopoietic neoplasms by flow cytometry. Cytom. Part B Clin. Cytom. 2016, 90, 47–53. [Google Scholar] [CrossRef]
  15. Wood, B.L. Acute Myeloid Leukemia Minimal Residual Disease Detection: The Difference from Normal Approach. Curr. Protoc. Cytom. 2020, 93, e73. [Google Scholar] [CrossRef] [PubMed]
  16. Baker, M.A.; Ramachandar, K.; Taub, R.N. Specificity of Heteroantisera to Human Acute Leukemia-Associated Antigens. J. Clin. Investig. 1974, 54, 1273–1278. [Google Scholar] [CrossRef]
  17. Greaves, M.F.; Brown, G.; Rapson, N.T.; Lister, T.A. Antisera to acute lymphoblastic leukemia cells. Clin. Immunol. Immunopathol. 1975, 4, 67–84. [Google Scholar] [CrossRef]
  18. Al Mawali, A.; Gillis, D.; Hissaria, P.; Lewis, I. Incidence, Sensitivity, and Specificity of Leukemia-Associated Phenotypes in Acute Myeloid Leukemia Using Specific Five-Color Multiparameter Flow Cytometry. Am. J. Clin. Pathol. 2008, 129, 934–945. [Google Scholar] [CrossRef]
  19. 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]
  20. Sievers, E.L.; Lange, B.J.; Alonzo, T.A.; Gerbing, R.B.; Bernstein, I.D.; Smith, F.O.; Arceci, R.J.; Woods, W.G.; Loken, M.R. Immunophenotypic evidence of leukemia after induction therapy predicts relapse: Results from a prospective Children’s Cancer Group study of 252 patients with acute myeloid leukemia. Blood 2003, 101, 3398–3406. [Google Scholar] [CrossRef] [Green Version]
  21. MRD-AML-BFM Study Group; Langebrake, C.; Creutzig, U.; Dworzak, M.; Hrusak, O.; Mejstrikova, E.; Griesinger, F.; Zimmermann, M.; Reinhardt, D. Residual disease monitoring in childhood acute myeloid leukemia by multiparameter flow cytometry: The MRD-AML-BFM Study Group. J. Clin. Oncol. 2006, 24, 3686–3692. [Google Scholar] [CrossRef]
  22. Liu, F.-J.; Cheng, W.-Y.; Lin, X.-J.; Wang, S.-Y.; Jiang, T.-Y.; Ma, T.-T.; Zhu, Y.-M.; Shen, Y. Measurable Residual Disease Detected by Multiparameter Flow Cytometry and Sequencing Improves Prediction of Relapse and Survival in Acute Myeloid Leukemia. Front. Oncol. 2021, 11, 677833. [Google Scholar] [CrossRef] [PubMed]
  23. Getta, B.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 Transpl. 2017, 23, 1064–1071. [Google Scholar] [CrossRef] [PubMed]
  24. Venditti, A.; Piciocchi, A.; Candoni, A.; Melillo, L.; Calafiore, V.; Cairoli, R.; de Fabritiis, P.; Storti, G.; Salutari, P.; Lanza, F.; et al. GIMEMA AML1310 trial of risk-adapted, MRD-directed therapy for young adults with newly diagnosed acute myeloid leukemia. Blood 2019, 134, 935–945. [Google Scholar] [CrossRef] [PubMed]
  25. Coustan-Smith, E.; Song, G.; Shurtleff, S.; Yeoh, A.E.-J.; Chng, W.J.; Chen, S.P.; Rubnitz, J.E.; Pui, C.-H.; Downing, J.R.; Campana, D. Universal monitoring of minimal residual disease in acute myeloid leukemia. JCI Insight 2018, 3, 98561. [Google Scholar] [CrossRef]
  26. Terwijn, M.; van Putten, W.L.J.; Kelder, A.; van der Velden, V.H.J.; Brooimans, R.A.; Pabst, T.; Maertens, J.; Boeckx, N.; de Greef, G.E.; Valk, P.J.M.; 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. [Google Scholar] [CrossRef]
  27. Jacobsohn, D.A.; Loken, M.R.; Fei, M.; Adams, A.; Brodersen, L.E.; Logan, B.R.; Ahn, K.W.; Shaw, B.E.; Kletzel, M.; Olszewski, M.; et al. Outcomes of Measurable Residual Disease in Pediatric Acute Myeloid Leukemia before and after Hematopoietic Stem Cell Transplant: Validation of Difference from Normal Flow Cytometry with Chimerism Studies and Wilms Tumor 1 Gene Expression. Biol. Blood Marrow Transplant. 2018, 24, 2040–2046. [Google Scholar] [CrossRef]
  28. Daga, S.; Rosenberger, A.; Kashofer, K.; Heitzer, E.; Quehenberger, F.; Halbwedl, I.; Graf, R.; Krisper, N.; Prietl, B.; Höfler, G.; et al. Sensitive and broadly applicable residual disease detection in acute myeloid leukemia using flow cytometry-based leukemic cell enrichment followed by mutational profiling. Am. J. Hematol. 2020, 95, 1148–1157. [Google Scholar] [CrossRef]
  29. Short, N.J.; Zhou, S.; Fu, C.; Berry, D.A.; Walter, R.B.; Freeman, S.D.; Hourigan, C.S.; Huang, X.; Nogueras Gonzalez, G.; Hwang, H.; et al. Association of Measurable Residual Disease with Survival Outcomes in Patients with Acute Myeloid Leukemia: A Systematic Review and Meta-analysis. JAMA Oncol. 2020, 6, 1890–1899. [Google Scholar] [CrossRef]
  30. Wei, A.H.; Döhner, H.; Pocock, C.; Montesinos, P.; Afanasyev, B.; Dombret, H.; Ravandi, F.; Sayar, H.; Jang, J.-H.; Porkka, K.; et al. Oral Azacitidine Maintenance Therapy for Acute Myeloid Leukemia in First Remission. N. Engl. J. Med. 2020, 383, 2526–2537. [Google Scholar] [CrossRef]
  31. Maiti, A.; DiNardo, C.D.; Wang, S.A.; Jorgensen, J.; Kadia, T.M.; Daver, N.G.; Short, N.J.; Yilmaz, M.; Pemmaraju, N.; Borthakur, G.; et al. Prognostic value of measurable residual disease after venetoclax and decitabine in acute myeloid leukemia. Blood Adv. 2021, 5, 1876–1883. [Google Scholar] [CrossRef] [PubMed]
  32. 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-Naive Acute Myeloid Leukemia With Venetoclax and Azacitidine. J. Clin. Oncol. 2022, 40, 855–865. [Google Scholar] [CrossRef] [PubMed]
  33. Araki, D.; Wood, B.L.; Othus, M.; Radich, J.P.; Halpern, A.B.; Zhou, Y.; Mielcarek, M.; Estey, E.H.; Appelbaum, F.R.; Walter, R.B. Allogeneic Hematopoietic Cell Transplantation for Acute Myeloid Leukemia: Time to Move Toward a Minimal Residual Disease-Based Definition of Complete Remission? J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 329–336. [Google Scholar] [CrossRef] [PubMed]
  34. Rubnitz, J.E.; Inaba, H.; Dahl, G.; Ribeiro, R.C.; Bowman, W.P.; Taub, J.; Pounds, S.; Razzouk, B.I.; Lacayo, N.J.; Cao, X.; et al. Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: Results of the AML02 multicentre trial. Lancet Oncol. 2010, 11, 543–552. [Google Scholar] [CrossRef]
  35. Walter, R.B.; Gooley, T.A.; Wood, B.L.; Milano, F.; Fang, M.; Sorror, M.L.; Estey, E.H.; Salter, A.I.; Lansverk, E.; Chien, J.W.; et al. Impact of Pretransplantation Minimal Residual Disease, As Detected by Multiparametric Flow Cytometry, on Outcome of Myeloablative Hematopoietic Cell Transplantation for Acute Myeloid Leukemia. J. Clin. Oncol. 2011, 29, 1190–1197. [Google Scholar] [CrossRef]
  36. Zhou, Y.; Othus, M.; Araki, D.; Wood, B.L.; Radich, J.P.; Halpern, A.B.; Mielcarek, M.; Estey, E.H.; Appelbaum, F.R.; Walter, R.B. Pre- and post-transplant quantification of measurable (‘minimal’) residual disease via multiparameter flow cytometry in adult acute myeloid leukemia. Leukemia 2016, 30, 1456–1464. [Google Scholar] [CrossRef]
  37. Guolo, F.; Di Grazia, C.; Minetto, P.; Raiola, A.M.; Clavio, M.; Miglino, M.; Tedone, E.; Contini, P.; Mangerini, R.; Kunkl, A.; et al. Pre-transplant minimal residual disease assessment and transplant-related factors predict the outcome of acute myeloid leukemia patients undergoing allogeneic stem cell transplantation. Eur. J. Haematol. 2021, 107, 573–582. [Google Scholar] [CrossRef]
  38. Roboz, G.J.; Ravandi, F.; Wei, A.H.; Dombret, H.; Döhner, H.; Thol, F.; Voso, M.T.; Schuh, A.C.; Porkka, K.; La Torre, I.; et al. CC-486 Prolongs Survival for Patients with Acute Myeloid Leukemia (AML) in Remission after Intensive Chemotherapy (IC) Independent of the Presence of Measurable Residual Disease (MRD) at Study Entry: Results from the QUAZAR AML-001 Maintenance Trial. Blood 2020, 136, 32–33. [Google Scholar] [CrossRef]
  39. de Leeuw, D.C.; Ossenkoppele, G.J.; Janssen, J. Older Patients with Acute Myeloid Leukemia Deserve Individualized Treatment. Curr. Oncol. Rep. 2022. [Google Scholar] [CrossRef]
  40. DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef]
  41. DiNardo, C.D.; Pratz, K.; Pullarkat, V.; Jonas, B.A.; Arellano, M.; Becker, P.S.; Frankfurt, O.; Konopleva, M.; Wei, A.H.; Kantarjian, H.M.; et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 2019, 133, 7–17. [Google Scholar] [CrossRef] [PubMed]
  42. Simoes, C.; Paiva, B.; Martinez-Cuadron, D.; Bergua, J.M.; Vives, S.; Algarra, L.; Tormo, M.; Martinez, P.; Serrano, J.; Herrera, P.; et al. Measurable residual disease in elderly acute myeloid leukemia: Results from the PETHEMA-FLUGAZA phase 3 clinical trial. Blood Adv. 2021, 5, 760–770. [Google Scholar] [CrossRef] [PubMed]
  43. 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] [PubMed]
  44. 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] [PubMed]
  45. Walter, R.B.; Buckley, S.A.; Pagel, J.M.; Wood, B.L.; Storer, B.E.; Sandmaier, B.M.; Fang, M.; Gyurkocza, B.; Delaney, C.; Radich, J.P.; et al. Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission. Blood 2013, 122, 1813–1821. [Google Scholar] [CrossRef]
  46. 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. [Google Scholar] [CrossRef]
  47. Hanekamp, D.; Snel, A.N.; Kelder, A.; Scholten, W.J.; Khan, N.; Metzner, M.; Irno-Consalvo, M.; Sugita, M.; de Jong, A.; Oude Alink, S.; et al. Applicability and reproducibility of acute myeloid leukaemia stem cell assessment in a multi-centre setting. Br. J. Haematol. 2020, 190, 891–900. [Google Scholar] [CrossRef]
  48. Terwijn, M.; Zeijlemaker, W.; Kelder, A.; Rutten, A.P.; Snel, A.N.; Scholten, W.J.; Pabst, T.; Verhoef, G.; Lowenberg, B.; Zweegman, S.; et al. Leukemic stem cell frequency: A strong biomarker for clinical outcome in acute myeloid leukemia. PLoS ONE 2014, 9, e107587. [Google Scholar] [CrossRef]
  49. Dominy, K.; Mokretar, K.; Reid, A.G.; Khorashad, J.S. Molecular Monitoring of Chronic Myeloid Leukemia. Methods Mol. Biol. 2020, 2065, 153–173. [Google Scholar] [CrossRef]
  50. Rp, H.; Dp, S.; Ta, G.; Bl, E. Clonal hematopoiesis and measurable residual disease assessment in acute myeloid leukemia. Blood 2020, 135, 1729–1738. [Google Scholar] [CrossRef]
  51. Boyd, A.L.; Aslostovar, L.; Reid, J.; Ye, W.; Tanasijevic, B.; Porras, D.P.; Shapovalova, Z.; Almakadi, M.; Foley, R.; Leber, B.; et al. Identification of Chemotherapy-Induced Leukemic-Regenerating Cells Reveals a Transient Vulnerability of Human AML Recurrence. Cancer Cell 2018, 34, 483–498.e485. [Google Scholar] [CrossRef] [PubMed]
  52. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer–immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef] [PubMed]
  53. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [PubMed]
  54. Blankenstein, T.; Coulie, P.G.; Gilboa, E.; Jaffee, E.M. The determinants of tumour immunogenicity. Nat. Rev. Cancer 2012, 12, 307–313. [Google Scholar] [CrossRef]
  55. Gotwals, P.; Cameron, S.; Cipolletta, D.; Cremasco, V.; Crystal, A.; Hewes, B.; Mueller, B.; Quaratino, S.; Sabatos-Peyton, C.; Petruzzelli, L.; et al. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat. Rev. Cancer 2017, 17, 286–301. [Google Scholar] [CrossRef]
  56. Kottke, T.; Boisgerault, N.; Diaz, R.M.; Donnelly, O.; Rommelfanger-Konkol, D.; Pulido, J.; Thompson, J.; Mukhopadhyay, D.; Kaspar, R.; Coffey, M.; et al. Detecting and targeting tumor relapse by its resistance to innate effectors at early recurrence. Nat. Med. 2013, 19, 1625–1631. [Google Scholar] [CrossRef]
  57. Boisgerault, N.; Kottke, T.; Pulido, J.; Thompson, J.; Diaz, R.M.; Rommelfanger-Konkol, D.; Embry, A.; Saenz, D.; Poeschla, E.; Pandha, H.; et al. Functional Cloning of Recurrence-specific Antigens Identifies Molecular Targets to Treat Tumor Relapse. Mol. Ther. 2013, 21, 1507. [Google Scholar] [CrossRef]
  58. Bruni, D.; Angell, H.K.; Galon, J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat. Rev. Cancer 2020, 20, 662–680. [Google Scholar] [CrossRef]
  59. Dufva, O.; Pölönen, P.; Brück, O.; Keränen, M.A.I.; Klievink, J.; Mehtonen, J.; Huuhtanen, J.; Kumar, A.; Malani, D.; Siitonen, S.; et al. Immunogenomic landscape of hematological malignancies. Cancer Cell 2020, 38, 380–399.E13. [Google Scholar] [CrossRef]
  60. Brück, O.; Dufva, O.; Hohtari, H.; Blom, S.; Turkki, R.; Ilander, M.; Kovanen, P.; Pallaud, C.; Ramos, P.M.; Lähteenmäki, H.; et al. Immune profiles in acute myeloid leukemia bone marrow associate with patient age, T-cell receptor clonality, and survival. Blood Adv. 2020, 4, 274–286. [Google Scholar] [CrossRef]
  61. Christopher, M.J.; Petti, A.A.; Rettig, M.P.; Miller, C.A.; Chendamarai, E.; Duncavage, E.J.; Klco, J.M.; Helton, N.M.; O’Laughlin, M.; Fronick, C.C.; et al. Immune escape of relapsed AML cells after allogeneic transplantation. N. Engl. J. Med. 2018, 379, 2330–2341. [Google Scholar] [CrossRef] [PubMed]
  62. Toffalori, C.; Zito, L.; Gambacorta, V.; Riba, M.; Oliveira, G.; Bucci, G.; Barcella, M.; Spinelli, O.; Greco, R.; Crucitti, L.; et al. Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation. Nat. Med. 2019, 25, 603–611. [Google Scholar] [CrossRef] [PubMed]
  63. Othus, M.; Gale, R.P.; Hourigan, C.S.; Walter, R.B. Statistics and measurable residual disease (MRD) testing: Uses and abuses in hematopoietic cell transplantation. Bone Marrow Transpl. 2020, 55, 843–850. [Google Scholar] [CrossRef]
  64. Lakshmikanth, T.; Olin, A.; Chen, Y.; Mikes, J.; Fredlund, E.; Remberger, M.; Omazic, B.; Brodin, P. Mass Cytometry and Topological Data Analysis Reveal Immune Parameters Associated with Complications after Allogeneic Stem Cell Transplantation. Cell Rep. 2017, 20, 2238–2250. [Google Scholar] [CrossRef] [PubMed]
  65. Daver, N.; Garcia-Manero, G.; Basu, S.; Boddu, P.C.; Alfayez, M.; Cortes, J.E.; Konopleva, M.; Ravandi-Kashani, F.; Jabbour, E.; Kadia, T.; et al. Efficacy, safety, and biomarkers of response to azacitidine and nivolumab in relapsed/refractory acute myeloid leukemia: A nonrandomized, open-label, phase II study. Cancer Discov. 2019, 9, 370–383. [Google Scholar] [CrossRef]
  66. Lamprianidou, E.; Kordella, C.; Kazachenka, A.; Zoulia, E.; Bernard, E.; Filia, A.; Laidou, S.; Garantziotis, P.; Vassilakopoulos, T.P.; Papageorgiou, S.G.; et al. Modulation of IL-6/STAT3 signaling axis in CD4+FOXP3− T cells represents a potential antitumor mechanism of azacitidine. Blood Adv. 2021, 5, 129. [Google Scholar] [CrossRef]
  67. Lee, J.B.; Khan, D.H.; Hurren, R.; Xu, M.; Na, Y.; Kang, H.; Mirali, S.; Wang, X.; Gronda, M.; Jitkova, Y.; et al. Venetoclax enhances T cell-mediated antileukemic activity by increasing ROS production. Blood 2021, 138, 234–245. [Google Scholar] [CrossRef]
  68. Ho, C.M.; McCarthy, P.L.; Wallace, P.K.; Zhang, Y.; Fora, A.; Mellors, P.; Tario, J.D.; McCarthy, B.L.S.; Chen, G.L.; Holstein, S.A.; et al. Immune signatures associated with improved progression-free and overall survival for myeloma patients treated with AHSCT. Blood Adv. 2017, 1, 1056–1066. [Google Scholar] [CrossRef]
  69. Paiva, B.; Cedena, M.-T.; Puig, N.; Arana, P.; Vidriales, M.-B.; Cordon, L.; Flores-Montero, J.; Gutierrez, N.C.; Martín-Ramos, M.-L.; Martinez-Lopez, J.; et al. Minimal residual disease monitoring and immune profiling in multiple myeloma in elderly patients. Blood 2016, 127, 3165–3174. [Google Scholar] [CrossRef]
  70. Tiong, I.S.; Dillon, R.; Ivey, A.; Kuzich, J.A.; Thiagarajah, N.; Sharplin, K.M.; Kok, C.H.; Tedjaseputra, A.; Rowland, J.P.; Grove, C.S.; et al. Clinical impact of NPM1-mutant molecular persistence after chemotherapy for acute myeloid leukemia. Blood Adv. 2021, 5, 5107–5111. [Google Scholar] [CrossRef]
  71. Schraivogel, D.; Kuhn, T.M.; Rauscher, B.; Rodríguez-Martínez, M.; Paulsen, M.; Owsley, K.; Middlebrook, A.; Tischer, C.; Ramasz, B.; Ordoñez-Rueda, D.; et al. High-speed fluorescence image-enabled cell sorting. Science 2022, 375, 315–320. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Quantifiable immune factors and other factors affecting leukemic relapse that can be used to increase MFC-MRD accuracy. (Created with BioRender.com, (Accessed on 8 August 2022)). Multiparametric flow cytometry can accurately assess post-treatment qualitative and quantitative alterations in immune and leukemic cell and bone marrow cytokines. Treatment with the hypomethylating agent azacytidine can modulate the signal transducer and activator of transcription (STAT) architecture in both CD4+ and CD34+ cells, while the bcl-2 inhibitor venetoclax can increase reactive oxygen species generation and boost the antileukemic activity of CD8+ and CD4/CD8 double-negative T cells. Conventional chemotherapy, on the other hand, leads to the emergence of a phenotypically and molecularly distinct subpopulation of leukemia-regenerating cells which cannot be traced with the current panels of MFC-MRD. Finally, relapse after allogeneic transplantation appears to be driven by epigenetic alterations in leukemic stem cells resulting in deregulation of the immune pathways involved in antigenic presentation and T cell co-stimulation.
Figure 1. Quantifiable immune factors and other factors affecting leukemic relapse that can be used to increase MFC-MRD accuracy. (Created with BioRender.com, (Accessed on 8 August 2022)). Multiparametric flow cytometry can accurately assess post-treatment qualitative and quantitative alterations in immune and leukemic cell and bone marrow cytokines. Treatment with the hypomethylating agent azacytidine can modulate the signal transducer and activator of transcription (STAT) architecture in both CD4+ and CD34+ cells, while the bcl-2 inhibitor venetoclax can increase reactive oxygen species generation and boost the antileukemic activity of CD8+ and CD4/CD8 double-negative T cells. Conventional chemotherapy, on the other hand, leads to the emergence of a phenotypically and molecularly distinct subpopulation of leukemia-regenerating cells which cannot be traced with the current panels of MFC-MRD. Finally, relapse after allogeneic transplantation appears to be driven by epigenetic alterations in leukemic stem cells resulting in deregulation of the immune pathways involved in antigenic presentation and T cell co-stimulation.
Cancers 14 04294 g001
Table 1. Advantages and disadvantages of methods used to detect MRD in AML.
Table 1. Advantages and disadvantages of methods used to detect MRD in AML.
MethodSensitivityAdvantagesDisadvantagesReferences
Flow cytometry
FC-LAIP
(Leukemia-Associated Immunophenotypes)
10−3 to 10−5Sensitivity
Applicability to >90% of patients
Rapid turnaround time
Available technique through laboratories
Can distinguish
between live and dead cells
Experienced staff needed for proper interpretation
Need for standardization
Stability of the leukemic phenotype missing
Diagnostic pretreatment sample needed
Extended antibody panel needed
Sensitivity depends on the antibody used
Brooimans 2019 [12]
Maurer-Granofszky 2021 [13]
[Wood 2016] [14]
Flow cytometry
FC-DfN (Different from Normal)
10−3 to 10−5Sensitivity
Applicability to >90% of patients
Diagnostic sample not required
Phenotypic shifts do not affect the results
Rapid
turn-around time
Can distinguish
between live and dead cells
Need for standardization
Experienced staff needed to operate the process, subjectivity in the definition of population
Sensitivity depends on the antibody used
Schuurhuis 2018 [9]
Maurer-Granofszky 2021 [13]
Wood 2020 [15]
−19NGS10−3 to 10−5Limited applicability
Easy to be conducted
High sensitivity, theoretically to 10−6, depending on the NGS platform
Need for standardization
Mutations can be identified in healthy populations (not necessarily linked with disease)
Sample contamination
Sensitivity is affected by error rate
Clonal evolution (if based on allelic ratios)
Ngai 2021 [2]
Dix 2020 [7]
RT-qPCR10−3 to 10−5High sensitivity (≥MFC)
Quality assurance integration
Applicability
Standardization
Time-consuming
Need for expertise
Threshold limit settings required
Expensive
Sensitivity is affected as well by the expression level of the target per cell
Molecular targets applicable to only ~50% of all AML patients (less in elderly)
Ngai 2021 [2]
Wood 2016 [14]
Table 2. Studies with Multiparametric Flow Cytometry (MFC)-based MRD in AML.
Table 2. Studies with Multiparametric Flow Cytometry (MFC)-based MRD in AML.
ReferenceNo. of PatientsAge (Years)
Median (Range)
MethodCut-Off
Level
Timepoint of MRD AssessmentOutcome
Intensive Chemotherapy
Sievers et al.,
2003 [20]
252 Pediatric0–2 or 10–21 MFC-MRD≥0.5% blastsBefore and after intensification therapy Before
MRDpos:
Relative risk of relapse
4.2 (95% CI = 2.6–6.8, p < 0.0001)
After
MRDpos:
5.3 (95% CI = 3.2–8.6, p < 0.0001)
All cohorts:
Median time to relapse:
MRDpos vs. MRD neg 168.5 days versus 293 days, p = 0.008)
Relative risk of death
MRDpos:3.4 (95% CI = 2.2–5.4, p < 0.0001)
Langebrake et al.,
2006 [21]
(2 Parts)
150 Part1
7.59 (0.2–17.7)
Part 2
9.98
(0.06–20)
LAIP
MFC-MRD
<0.1%-BPM1
First at 15 days from the start of treatment
-BPM2
Second at 29 days from the start of treatment
3-year EFS
BPM1:
ΜRDneg: 48% ± 9%
ΜRDpos: 71% ± 6%
p = 0.029
BPM2:
ΜRDneg: 50% ± 7%
ΜRDpos: 70% ± 6%
p = 0.033
Fu-Jia Liu et al., 2021 [22]49245
(15–74)
MFC-MRD
10-color FC
0.1%After induction therapyMRDneg:
<60, 276 (83.9%)
≥60, 53 (16.1%)
MRDpos:
<60, 136 (83.4%)
≥60, 27 (16.6%)
p = 1.000
Getta et al., 2017 [23]104 58
(21–78)
DfN MFC-MRD
alone or combined with NGS
10-color MFC assay
0.1%Pre-alloSCTMFC–MRDneg:
18-month relapse 9%
OS 73%
MFC–MRDpos:
18-month relapse: 37%
OS 48%
Vendittiet al., 2019 [24]500 49
(18–60.9)
LAIP MFC-MRD
combined with qPCR
8-color MFC assay
0.035%After
consolidation
Both neg: 2-year OS
89% and DFS 69%
MFCpos/PCR neg or
MFCneg/PCR pos:
2-year OS 88–89%
DFS 65–76%
Both pos: 2-year OS
55%, DFS 22%
Coustan-Smith et al., 2018 [25]370<1–63Novel
leukemia-specific
markers
1 in
105
At diagnosisClinical outcomes not determined
Terwijn et al., 2013 [26]51748
(18–60)
LAIPMFC-MRD0.1%After induction
therapy
MRDneg:
RFS > 47 months
4-year RFS 52%
MRDpos:
median RFS 8.6 months
4-year RFS 23%
Jacobsohn et al., 2018 [27]144Patients < 21 years of ageDfN MFC-MRD 0.02%preHCTMRDneg:
2-year relapse risk: 32%
2-year DFS: 55%
2-year OS: 63%
MRDpos:
2-year relapse risk: 70%
2-year DFS: 10%
2-year OS: 20%
Daga et al., 2020 [28]39 out of 41 patientsAdults > 60 yearsCombination of MFC-MRD
followed by (NGS) or digital PCR
0.1%After induction
therapy
MRDneg: 18 (48.2%)
% relapse 27.8%
MRDpos: 21 (53.8%)
% relapse 71.4%
p = 0.007
Median RFS 283 vs. not reached, p = 0.003
5-year CIR: 90.5% vs. 28%, p < 0.001
OS: not significant
p = 0.085
Median follow-up time: 559 days
Short et al., 2020 [29]
Meta-analysis of 81 studies
151Adult-PediatricMFC-MRD, qPCR NGS, or
cytogenetics/FISH
variousInduction or during/after
consolidation
25 (40) MFC-MRD detection studies with OS analysis
29 (43) MFC-MRD detection studies with DFS analysis
Overall analysis through all methods:
MRDneg
5-y DFS: 64%
5-y OS: 68%
MRDpos
5-y DFS: 25%
5-y OS: 34%
Wei et al., 2020 [30]47286
(55–86)
LAIP
MFC-MRD
0.1%First remission after IC2-year survival
MRDneg CC-486: 58.6%
MRDpos CC-486: 39.5%
MRDneg placebo: 51.7%
MRDpos placebo: 22.0%
2-year survival differences (95% CI)
MRDneg: 6.9 (5.8 to 19.5)
MRDpos: 17.5 (5.3 to 29.8)
Low-Intensity Chemotherapy
Mait et al., 2021 [31]97
Venetoclax plus Decitabine
72
(68–78)
MFC-MRD0.1%1, 2, 4 months of therapyMRDneg at 2 months:
Median RFS, not reached vs. 5.2 months
hazard ratio [HR] = 0.31; 95% CI, 0.12–0.78; p = 0.004
Median EFS, not reached vs. 5.8 months; HR, 0.25; 95% CI, 0.12–0.55; p = 0.001)
MRDneg CR
(median OS, 25.1 vs. 7.1 months; HR= 0.23; 95% CI, 0.110.51; p = 0.001)
MRD neg at 1 month
Median OS, 25.1 vs. 3.4 months; HR, 0.15; 95% CI, 0.03–0.64; p = 0.0001
Pratz et al., 2022 [32]164
Azacitidine–Venetoclax (N = 286)
Azacitidine–Placebo
(N = 145)
76
(49–91)
76
(60–90)
MFC-MRD0.1%After cycle 1 and every 3 cyclesMRDneg
Median EFS: not reached
Median OS: not reached
MRDpos
Median EFS: 10.6
Median OS: 18.7
Allo-HCT
Araki et al., 2016 [33]35950
(18.2–75.3)
DfN
MFC-MRD
10 colors
0.1%Pre-alloSCTMRDneg:
3-year OS >70%
Relapse risk 20–25%
MRDpos:
3-year OS 25%
Relapse risk 70%
Rubnitz et al., 2010 [34]2029.1
(2–21.4)
MFC-MRD>0.1%After induction I and IIAfter induction I:
MRDneg
3-year CIR: 16.9% ± 3.4%
3-year EFS: 73.6% ± 5%
MRDpos
3-year CIR: 38.6% ± 5.8%
3-year EFS: 43.1% ± 6.9%
After induction II:
MRD-neg
3-year CIR: 16.7% ± 3.1%
3-year EFS: 71.2% ± 4.7%
MRDpos
3-year CIR: 56.3% ± 8.4%
3-year EFS: 35.8% ± 8.6%
Walter et al., 2011 [35]9945.3
(0.6–69.5)
DfN
MFC-MRD
10-colors
0.1%Before HCTOS
MRDneg (n = 75) HR:1
MRDpos (n = 24) HR: 4.05 95% CI = 1.90 to 8.62
p < 0.001
Relapse
MRDneg HR:1
MRDpos HR: 8.49
95% CI: 3.67 to 19.65
p < 0.001
2-year OS
MRDneg: 76.6% (64.4% to 85.1%)
MRDpos: 30.2% (13.1% to 49.3%)
2-year DFS
MRDneg: 74.8% (62.8% to 83.4%)
MRDpos: 9.0% (1.6% to 24.9%)
Zhou Y et al., 2016 [36]279>18 yearsDfN
MFC-MRD
10 colors
Not usedPre-alloSCT and
post-alloSCT (day 28)
MRDpos pre-alloSCT and
MRDneg post:
3-year OS: 29%
3-year RFS: 18%
MRDpos at both timepoints:
3-year OS: 19%
3-year RFS: 14%
MRDneg at both timepoints:
3-year OS: 76%
3-year RFS: 71
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pessach, I.; Spyropoulos, T.; Lamprianidou, E.; Kotsianidis, I. MRD Monitoring by Multiparametric Flow Cytometry in AML: Is It Time to Incorporate Immune Parameters? Cancers 2022, 14, 4294. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14174294

AMA Style

Pessach I, Spyropoulos T, Lamprianidou E, Kotsianidis I. MRD Monitoring by Multiparametric Flow Cytometry in AML: Is It Time to Incorporate Immune Parameters? Cancers. 2022; 14(17):4294. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14174294

Chicago/Turabian Style

Pessach, Ilias, Theodoros Spyropoulos, Eleftheria Lamprianidou, and Ioannis Kotsianidis. 2022. "MRD Monitoring by Multiparametric Flow Cytometry in AML: Is It Time to Incorporate Immune Parameters?" Cancers 14, no. 17: 4294. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14174294

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop