Next Article in Journal
The Tumor Microenvironment in Tumorigenesis and Therapy Resistance Revisited
Next Article in Special Issue
Optimal Use of Novel Immunotherapeutics in B-Cell Precursor ALL
Previous Article in Journal
Predictive Biomarkers for Immune-Related Endocrinopathies following Immune Checkpoint Inhibitors Treatment
Previous Article in Special Issue
Modern Management Options for Ph+ ALL
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Optimizing Molecular Minimal Residual Disease Analysis in Adult Acute Lymphoblastic Leukemia

1
Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
2
GIMEMA Foundation, 00182 Rome, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(2), 374; https://doi.org/10.3390/cancers15020374
Submission received: 12 December 2022 / Revised: 2 January 2023 / Accepted: 3 January 2023 / Published: 6 January 2023
(This article belongs to the Special Issue Therapeutic Progress in Adult Acute Lymphoblastic Leukemia)

Abstract

:

Simple Summary

Minimal/measurable residual disease (MRD) monitoring is a powerful and independent predictor of outcomes in both children and adult acute lymphoblastic leukemia (ALL). MRD monitoring enables patients’ stratification into different risk-adapted treatment arms; it guides treatment decisions in clinical practice, including stem cell transplantation, and represents an early marker of impending relapse. Real-time quantitative PCR is the most widely used molecular method for MRD assessment, but there are some limitations that new approaches may overcome. In this review, we discuss the most recent technological advances in MRD monitoring that are allowing to increase the number of evaluable patients and the levels of quantification and also have the potential to study different disease compartments.

Abstract

Minimal/measurable residual disease (MRD) evaluation has resulted in a fundamental instrument to guide patient management in acute lymphoblastic leukemia (ALL). From a methodological standpoint, MRD is defined as any approach aimed at detecting and possibly quantifying residual neoplastic cells beyond the sensitivity level of cytomorphology. The molecular methods to study MRD in ALL are polymerase chain reaction (PCR) amplification-based approaches and are the most standardized techniques. However, there are some limitations, and emerging technologies, such as digital droplet PCR (ddPCR) and next-generation sequencing (NGS), seem to have advantages that could improve MRD analysis in ALL patients. Furthermore, other blood components, namely cell-free DNA (cfDNA), appear promising and are also being investigated for their potential role in monitoring tumor burden and response to treatment in hematologic malignancies. Based on the review of the literature and on our own data, we hereby discuss how emerging molecular technologies are helping to refine the molecular monitoring of MRD in ALL and may help to overcome some of the limitations of standard approaches, providing a benefit for the care of patients.

1. Introduction

Acute lymphoblastic leukemia (ALL) is a heterogeneous malignancy in terms of clinical manifestations, biological background, clinical course, and prognosis. It affects the early stages of hematopoiesis and more frequently the B cell lineage (75–80% in adults and 85–90% in pediatric patients) than the T cell lineage (20–25% in adults and 10–15% in pediatric patients). The acquisition of a series of genetic aberrations leads to impaired maturation, with an arrest in the differentiation process and an abnormal proliferation, generating a progeny of leukemic lymphoid blasts [1].
ALL is the most frequent neoplasm in childhood and also affects adults of all ages: between 2 and 5 years and above the age of 50 [2,3], with 60% of cases discovered in individuals below 20 years of age [4].
Despite the progressively improved 5-year survival rate [5,6,7,8,9,10], about 20% of children with ALL still relapse [11,12], and in adults, the risk of relapse affects up to 50% of patients [13,14,15].
In the modern era, the management of Philadelphia chromosome-negative (Ph-) B-ALL and T-ALL is based on multidrug chemotherapy regimens in order to obtain complete hematologic remission (CR) and a status of minimal residual disease (MRD) negativity with or without an allogeneic stem cell transplantation (SCT), plus an effective central nervous system prophylaxis.
The management of Ph+ ALL has also markedly changed over the years thanks to the advent of tyrosine kinase inhibitors (TKIs), with treatment programs that involve the use of a TKI (plus glucocorticoids and central nervous system prophylaxis), with or without deintensified chemotherapy in the induction phase, followed by monoclonal antibodies or multiagent chemotherapy with or without allogeneic transplantation [16].
Over the years, cooperative study groups, both for pediatric and adult ALL patients, have defined in their treatment protocols specific informative time points.
The concept of MRD refers to the proportion of remaining cancer cells among otherwise normal bone marrow or, more rarely, among circulating blood cells after any given treatment. In ALL, MRD monitoring has proven to be an independent prognostic factor and an important instrument for therapeutic decisions.
Several studies [17,18,19,20] have evaluated MRD levels by using either flow cytometry [17,18] or a molecular approach [19,20] at multiple time points during treatment, showing that MRD measurements in the first 3 months of treatment are the most informative for risk group assignment in childhood ALL, and MRD monitoring in the post-remission phase is more widely used in adult ALL.
Thus, MRD cutoff values were established, enabling patients’ stratification into different risk-adapted treatment arms. MRD is also utilized to monitor disease burden in the setting of SCT and represents an early marker of impending relapse at any time point during the course of the disease.
MRD assays are required to be highly sensitive (≥10−4) and to have broad applicability, accuracy, and reliability [21,22]. Furthermore, for molecular MRD analysis, its genetic target should be representative of the disease, expressed by all leukemic clones, stable over time (i.e., present at both diagnosis and relapse) to reflect the kinetics of the malignant clone during treatment, and sensitive [18]. Currently, the gold standard for molecular MRD analysis is polymerase chain reaction (PCR) amplification-based methods that use leukemia-specific (fusion gene transcripts) or patient-specific (immunoglobulin/T cell receptor (IG/TR) gene rearrangements) molecular markers [23,24,25,26,27,28].
These methods have been extensively standardized within the EuroMRD Consortium (www.euromrd.org, accessed on 28 December 2022), which established guidelines for the analysis and interpretation of real-time quantitative PCR (RQ-PCR) data in order to favor a homogeneous application of MRD studies within different treatment protocols for both pediatric and adult ALL patients [24,29].
In the present review, we will focus on how emerging molecular technologies may refine the molecular monitoring of MRD in ALL and overcome some of the limitations of standard approaches, such as: (1) difficulty in identifying a molecular marker and/or a sensitive patient-specific primer at the onset of disease in about 5–10%, (2) clonal evolution of IG/TR rearrangement patterns during the course of the disease and at relapse, (3) the need for diagnostic DNA for each MRD experiment, and finally, (4) RQ-PCR is not able to define precisely the amount of residual disease in those cases in which the disease burden is very low.

2. IG/TR Gene Rearrangements and Fusion Gene Transcripts

2.1. IG/TR Gene Rearrangements

Clonal rearrangements of IG/TR genes can be detected in >95% of ALL patients and are the most frequently used markers for MRD analysis [30]. IG/TR gene rearrangements are physiological events and are not directly linked to the pathogenesis of leukemia. In the case of the neoplastic transformation of a single lymphoid cell, all leukemic cells are supposed to contain the same rearranged clonal IG and/or TR gene(s), which can be exploited to detect a low number of ALL cells among a large number of normal lymphoid cells expressing gene rearrangements with different sequences.
To identify these molecular markers at diagnosis, genomic DNA derived from leukemic cells undergoes PCR amplification, and clonal PCR fragments [31,32] are sequenced by standard Sanger sequencing to define the junctional regions and to obtain complementary allele-specific oligonucleotide (ASO) primers for MRD monitoring, mostly performed by RQ-PCR. Amplification conditions and sensitivity testing for each ASO primer are established by a standard curve built on serial dilutions of diagnostic DNA in a DNA pool from healthy donors. The sensitive range reached by this method is about one leukemic cell in 100,000 (10−5) normal lymphoid cells. The standard curve built on the diagnostic material is used to quantify MRD in bone marrow (BM) or peripheral blood (PB) samples collected during and after treatment [33].
Notably, for biological (due to cell maturity stage) and technical reasons, it is not possible to monitor MRD by RQ-PCR in all patients, with a rate of failure of about 5–10%. Moreover, IG/TR gene rearrangements may change during therapy, potentially leading to false negative MRD results [34]. These changes may be due either to a clonal evolution or to ongoing secondary rearrangement processes [35].
Furthermore, the use of RQ-PCR can be limited by the availability of sufficient diagnostic material because the method is based on a comparison with a standard curve based on neoplastic DNA collected at the onset of the disease; this can limit the possibility of monitoring patients over a prolonged time period. Furthermore, a non-negligible fraction of patients with very low MRD levels is classified as positive not-quantifiable (PNQ) since the MRD resulted positive but is detected outside the quantitative range when compared with the standard curve of the RQ-PCR assay.
A better discrimination of these cases, which currently represents a challenge in clinical practice, is needed, particularly when MRD data guide therapeutic decisions.

2.2. Fusion Gene Transcripts

Chromosomal translocations are detected in about 40% of ALL patients. These genetic aberrations are ideal targets for MRD evaluation because they are leukemic cell-specific and extremely stable during the course of the disease [36,37]. The most frequent lesion in adults (25–30% of cases, with a further increase in the elderly population of up to 50% [38], and only 2–5% in pediatric age) is represented by the translocation t(9; 22) (q34; p11), i.e., the Ph chromosome, which determines the BCR-ABL1 rearrangement. The cryptic translocation t(12; 21) (p13; q22), responsible for the chimeric transcript ETV6/RUNX1, is very rare in adults (1% of cases), while it is observed in approximately 25–30% of childhood ALL, which is associated with a good prognosis.
Rearrangements of the KMT2A gene (alias MLL), located on chromosome 11q23, affect acute leukemias biologically unique for gene expression profiling. A number of genetic partners are currently known for the KMT2A gene. Among these, the AFF1 gene, involved in the balanced translocation t(4; 11) (q21; q23), is present in 2–5% of cases regardless of age. Children less than 1 year (infants) carry a rearrangement involving the KMT2A gene in about 80% of cases.
In T-ALL, genetic lesions generally involve the long arm of chromosomes 14q11 (where the TRA and TRD genes are located) and 7q34 (where the TRB gene), resulting in the juxtaposition of the TR loci with different transcription factors.
The alteration that most frequently characterizes the TAL1 gene is the deletion that juxtaposes the SIL gene (SIL/TAL1) and is detected in about 20% of cases [39]. Furthermore, other alterations involve ABL1; the more frequent rearrangements identified are NUP214, EML1, and ETV6.
Given the prognostic value of these chromosomal abnormalities, all cases at the onset of disease should be investigated [40] in order to monitor each patient by using a specific target.
Nevertheless, despite the great variability in the number of RNA transcripts produced, not all fusion genes are suitable for MRD evaluation [37]. In cases lacking a suitable MRD marker, alternative targets should be examined.
Given that the splicing process produces in all patients the same fusion transcript or a few splicing variants, the better material to detect these abnormalities is RNA. In this way, a small number of RQ-PCR assays is enough [41].
MRD assessment using fusion genes as a molecular marker is performed on a standard curve built on serial dilutions of a cell line or plasmid DNA (i.e., BCR-ABL1+).
The sensitive range reached is about one leukemic cell in 100,000 (10−5) normal lymphoid cells [37].
Despite its rapid and easy application, the accuracy of this analysis is altered by the variability in RNA transcripts, as already mentioned.

3. Novel Molecular Techniques for MRD Monitoring

3.1. Digital Droplet PCR (ddPCR)

ddPCR [42] appears to be a feasible and attractive alternative method for MRD assessment. The technique is based on the principle of partitioning the sample into several nano-PCR containing single, few, or no target sequences. PCR partitions are read as negative or positive by thresholds based on their fluorescence amplitude, and then the target sequence concentration is calculated by applying Poisson’s statistics [43,44] that link the theoretical depth of analysis to the number of compartments generated [45]. ddPCR [46,47] allows the quantitation of nucleic acid targets without the need for calibration curves [48] and seems to have a more precise quantification than the standard method because, in each droplet, the ratio between the target DNA to reagents is higher and each PCR is amplified and read individually so that any changes in fluorescence signal are detected [49] (Figure 1). Several ddPCR systems have been developed [50,51,52,53,54] thanks to their easy usage and applicability, adaptability, and saving of time.
At first, ddPCR was applied in the setting of molecular oncology [48,55] and prenatal diagnosis [56,57].
Currently, many reports on hematologic malignancies are also available [58,59,60,61,62,63,64]. The first studies established analytical parameters to investigate the applicability of ddPCR compared with RQ-PCR, and all reported a good concordance between the two techniques [58,59,60,61,62,63,64], underlining the sensitivity, accuracy, and reproducibility of ddPCR.
Regarding MRD evaluation, the discordances observed with RQ-PCR fell mostly at very low levels of disease. In this setting, the robustness of ddPCR to quantify RQ-PCR-PNQ samples and to identify false positive cases has been reported in several hematologic malignancies.
The Fondazione Italiana Linfomi (FIL) MRD Network has shown that non-Hodgkin’s lymphoma samples are defined as borderline because of alternating positive and negative results regardless of the method used (nested-PCR or RQ-PCR), and the type of rearrangement did not have interlaboratory discordance by ddPCR analysis [65]. Drandi et al. [66] compared ddPCR with RQ-PCR in mantle cell lymphoma MRD samples, showing very good concordance with both methods, particularly for samples with at least a 0.01% positivity. However, ddPCR presented a more robust quantification for low positive samples.
In ALL, few studies have explored the utility of ddPCR application for MRD assessment. In 2016, our group compared the two quantification techniques in 50 ALL adolescent patients, reporting a sensitivity and accuracy for ddPCR superimposable to that of RQ-PCR [63]. In 2019, we sought to analyze only adult ALL samples with RQ-PCR MRD levels ≤10−4 by both ddPCR/NGS [67]. The comparison showed a concordance rate of 57% and 52% for RQ-PCR/ddPCR and RQ-PCR/NGS, respectively, for samples concordantly positive or negative, while ddPCR and NGS also allowed to identify an MRD signal in very low positive samples, with a concordant MRD result among ddPCR and NGS in 87% of samples. The combined use of ddPCR and NGS significantly reduced the cohort of PNQ samples compared with RQ-PCR analysis and helped to predict three relapses in patients who resulted PNQ/NEG by RQ-PCR.
Recently, we also demonstrated that by increasing the number of patients and samples from a single trial, ddPCR was able to improve the rate of quantification in critically low positive samples [68]; moreover, the rate of concordance between ddPCR and NGS compared with RQ-PCR was also improved (92% vs. 87%).
These data demonstrated that in ALL, ddPCR is an extremely accurate tool when the RQ-PCR quantitative range is inferior to 10−4 thanks to its greater amplification efficiency and reproducibility. Moreover, the combined use of new technologies could better discriminate low positive samples that the standard method fails to detect or quantify the disease level [68].
Along the same line, Schwinghammer et al. [69] confirmed the high reproducibility and accuracy of ddPCR, particularly in very low positivity ranges and in quantitative ranges of higher positivity levels.
As for Ph+ ALL, three studies employed ddPCR for MRD monitoring [61,70,71]. In 2014, Iacobucci et al. [70] analyzed 60 ALL Ph+ samples, showing that ddPCR had a sensitivity to detect the disease comparable to that of the conventional approach.
A second study conducted by Coccaro and colleagues [61] proved that the ddPCR assay for p190 was able to quantify low disease levels by loading a high quantity of cDNA in different wells and combining the counts from multiple replicates. The higher ddPCR sensitivity was advantageous, translating in a timely manner for patient cure and predicting molecular relapse [61].
Finally, Ansuinelli and colleagues [71] tested 10 Ph+ ALL patients enrolled in the GIMEMA LAL 2116 trial [72], showing the ability of ddPCR to reduce the rate of PNQ samples and increase the proportion of quantifiable ones (46% of cases, p < 0.0001) significantly. Moreover, the authors highlighted as five cases that were RQ-PCR-negative and ddPCR-positive during the follow-up, 4/5 experienced a relapse [71].
As a step forward, there is growing interest in Ph+ ALL to monitor MRD using a “double-hit” strategy by using IG/TR gene rearrangements and the BCR-ABL1 fusion transcript as molecular markers [73,74,75]. In this context, we are testing both targets by ddPCR in adult Ph+ ALL with the aim of defining the concordance rate during MRD monitoring in terms of sensitivity and specificity (ongoing) and once follow-up data will be mature, to finally dissect which marker is more reliable.
In a study conducted on a pediatric cohort of ALL patients, it was shown that ddPCR MRD quantification at a particular time point (i.e., day +78) was associated with relapse and, in line with this, ddPCR MRD negative or PNQ results at the same time point were positively associated with a better outcome, highlighting the possible clinical significance of the method [76].
Currently, there are no guidelines for ddPCR MRD application. However, the EuroMRD Consortium is actively working to rapidly achieve this goal. SOPs (standard operating procedures) are published as a guide for digital analysis in lymphoid malignancies [77,78,79].

3.2. Next-Generation Sequencing (NGS)

NGS technologies have, in general, represented a step forward in genomic analysis.
Their application can be subdivided into two main purposes: the first one is to identify novel molecular markers that could help toward improvement in disease biology comprehension, and a clinical practical phase for detecting alterations that could contribute to better disease management by improving risk stratification and disease monitoring for the benefit of patients.
The NGS approach is a very complex analytic method capable of providing a large amount of data. Over the years, several sequencing techniques have been developed with different applications: whole genome sequencing (WGS), transcriptome sequencing (RNA-seq), whole exome sequencing (WES), and targeted gene sequencing.
Beyond the abovementioned goals, a great effort is ongoing to define whether the genomic lesions identified by NGS technologies can be used for MRD monitoring.
NGS of IG/TR gene rearrangements (IG/TR-NGS) [21] allows for the quick screening of all patients with lymphoid malignancies without the need to use patient-specific assays for MRD analysis. The IG/TR-NGS clonality assay is based on a multiplex PCR to amplify the target regions and by subsequent ligation of adapters for sequencing. After purification of the PCR products, the library preparation is performed, followed by sequencing on the platform (e.g., Ion Torrent or Illumina) [80]. The Ion Torrent platform [81] makes use of the electrochemical detection of hydrogen ions that are released during DNA synthesis [82]. In contrast, Illumina employs fluorescently labeled nucleotides that are incorporated during complementary DNA strand synthesis. Depending on the type of Illumina sequencer, this can be a 2-channel (e.g., MiniSeq, NextSeq, Nova-Seq) or a 4-channel chemistry (e.g., MiSeq, HiSeq). Ion Torrent and Illumina sequencing technologies require specific adapters for sequencing and barcodes for sample identification: for Ion Torrent sequencing, the adapters and barcodes are ligated to the amplicon, while for Illumina sequencing platforms, the sequencing adapters need to be incorporated in the amplicon primers [82,83]. These methods allow to detect and sequence any possible IG/TR gene rearrangements with a sensitivity that ranges from 10−4 to 10−6 [21,84,85] (Figure 2); to reach higher sensitive levels, a higher amount of DNA is needed [86,87]. Thanks to its great sensitivity, this approach can identify more gene rearrangements compared with the standard method. Oligoclonality is a well-known phenomenon in ALL that hampers conventional IG/TR MRD [88] assessment, but this can be better identified by NGS. Moreover, multiple IG/TR gene rearrangements in ALL result from both continuing rearrangement processes and from secondary rearrangements [89,90,91,92].
To perform an accurate MRD analysis by NGS, it is mandatory to define which rearrangements, among those identified at diagnosis, should be selected [93] because diagnostic clonal sequences can be detected in follow-up samples.
Moreover, it must be reminded that by NGS, it is possible to detect early clonal evolution, which is a probable event in ALL relapsed cases [94,95]; however, at relapse, the most representative clone is the diagnostic one in the majority of cases.
A comparative analysis of NGS and RQ-PCR using IG/TR gene rearrangements is reported by some authors, showing a good correlation of the two methods but with higher sensitivity and specificity of NGS for MRD quantification [85,86,96]. Importantly, NGS analysis does not define cases as PNQ because, unlike standard methods, its quantitative range is always superimposable to the sensitive one [86]. Finally, the predictive value after induction therapy and after allogenic SCT of NGS evaluation has also been reported [85,96].
At present, however, there are no international guidelines for NGS MRD analysis.
In the last years, the EuroClonality-NGS working group has published several papers on the development and standardization of IG/TR-NGS assays, focusing on marker identification protocols for subsequent MRD analysis, with a multicenter validation across many expert European laboratories [97,98,99,100].
Lastly, the first NGS assay (ClonoSEQ) for MRD evaluation in ALL or multiple myeloma patients using IG gene rearrangements has been approved by the FDA [101].
Finally, NGS LymphoTrack assays to study IG/TR gene rearrangements have been developed by Invivoscribe (Invivoscribe, San Diego, CA, USA). Different reports have demonstrated the clonality concordance between the LymphoTrack system and other molecular approaches and with disease monitoring, allowing to define the tool as sensitive and accurate for diagnostic testing and disease monitoring in B and T cell tumors [102,103,104].
Moreover, compared with the standard method, NGS assays show superior detection in cases with a lower tumor burden, allowing a higher resolution of clonal rearrangements partially hidden in a polyclonal background [105].
Thus overall, the higher sensitivity, specificity, and application of NGS for MRD monitoring compared with standard approaches make its implementation in the clinical practice one of the most important next goals to achieve. The availability of skilled bioinformatics, however, is required.
Some authors have compared ddPCR and NGS approaches in the setting of SCT [106,107], defining the methods as easy to perform and user-friendly in all molecular biology laboratories and highlighting a high correlation and concordance between the results [107].
In our experience, as already mentioned in the previous section, the combined use of both ddPCR and NGS significantly reduced the number of critical samples defined as PNQ by RQ-PCR, which is a challenge in clinical practice, showing great concordance with MRD quantification [66,67].

4. Novel Compartments for MRD Monitoring

MRD assessment is usually carried out on BM cells. In T-ALL, PB may be a reliable source for MRD monitoring since there appears not to be significant differences with the BM and could therefore be used as an alternative source. On the contrary, in B-lineage ALL, MRD levels tend to be 1 to 3 logs lower in PB compared with BM [108,109].
The study of cell-free DNA (cfDNA) and other PB components (known as “liquid biopsies”) is promising and has been investigated, especially in solid tumors [110]. A “liquid biopsy” can define the molecular profile of a tumor by analyzing PB using different methods: cell-free circulating nucleic acids (DNAs, mRNAs, micro-RNAs, or noncoding RNAs), circulating tumor cells (CTCs), or exosomes [111] (Figure 3).
cfDNA, composed of extracellular short double-stranded DNA fragments and detectable in almost all body fluids, including the blood, is involved in various physiologic and pathologic phenomena [112]. The majority of cfDNA found in the plasma originates from leukocytes, and only a small fraction is tumor-derived, defined as circulating tumor DNA (ctDNA). The concentration of ctDNA varies among patients and differs according to the type, localization, and stage of cancer.
MicroRNAs (miRNAs) are the most abundant RNA molecules in the blood. They present high stability and are very important in tumor growth and treatment resistance [113].
These cell-free circulating nucleic acids (cfDNAs and miRNAs) have been investigated in solid cancers and in non-Hodgkin’s lymphomas and are currently being investigated in ALL.
CTCs derive from an early step of tumor blood dissemination and are detected at low levels in most patients.
Studies of both animal models and cancer patients have demonstrated that CTCs have the potential to facilitate the early detection, prognosis, therapeutic target selection, and monitoring of response to treatment [114].
Exosomes are detectable in some types of cancer, carrying proteins and nucleic acids.
They are analyzed through their RNA content [115]. Compared with cfDNAs and miRNAs, CTCs and exosomes have found applications in other hematologic diseases, such as lymphomas, multiple myeloma, and chronic lymphocytic leukemia.
Recently, many studies also evaluated the potential use of cfDNA for risk stratification, monitoring tumor burden, and response to treatment in hematologic malignancies [116,117,118,119,120], with the caveat that the amount of cfDNA can be very low and variable across patients, and this can represent a challenge for MRD analysis. Its usage in ALL has been explored in some studies.
Schwarz and colleagues [121] conducted a study aimed at testing the feasibility of MRD measurements employing cfDNA from children with ALL. The authors concluded that despite low concentrations, cfDNA is a feasible tool to measure MRD kinetics during ALL induction therapy.
Likewise, Cheng et al. [122] evaluated the feasibility of MRD measurement by IG/TR gene rearrangement quantification on cfDNA, comparing it to flow cytometry analysis on BM. The study showed not only the feasibility of using cfDNA as a tool for treatment monitoring in leukemia patients but also demonstrated its prognostic significance.
Indeed, one of the most intriguing applications of cfDNA is represented by the evaluation of extramedullary relapses, in which leukemic clones are rarely sampled by standard BM aspirate/biopsy [123,124,125].
While liquid biopsy is an extremely attractive tool, it must be kept in mind that several preanalytic factors affect ctDNA amount and quality, thus hampering, at present, usage for molecular analysis [126,127] and highlighting the need to develop a standardized protocol to improve the impact of preanalytical step variability during plasma sample processing and, at the technological level, to allow accurate, robust, and reproducible results of ctDNA genotyping and quantification.

5. Conclusions

MRD is a powerful and independent predictor of outcomes in both children and adult ALL. MRD can provide different information according to the timing in which it is performed: very early during treatment, after induction/consolidation, or before and after SCT. Currently, RQ-PCR of IG/TR gene rearrangements and gene fusions is the most broadly applied and consolidated method for molecular MRD monitoring. Despite its broad applicability, the method, however, presents intrinsic limitations that must necessarily be overcome. ddPCR and NGS appear to be feasible and attractive alternative approaches for MRD assessment, more sensitive and accurate than the standard method that can better discriminate critical samples by RQ-PCR analysis, particularly in certain subsets, such as early T-ALL and pro-B ALL, often lacking a sensitive molecular marker. Given their intrinsic technical features, the combined use of both approaches could provide an accurate MRD analysis for virtually all patients. A major effort is ongoing at the international level to standardize and better understand how these new techniques could be incorporated into clinical trials.
During the last years, great interest has focused on the possibility of investigating ctDNA as a noninvasive, real-time, and sensitive tool. ctDNA may, in fact, provide a more comprehensive molecular overview of the genetic heterogeneity of a given tumor at presentation and may potentially enable sequential blood testing during the course of the disease. However, to prove the feasibility and reproducibility of ctDNA for the monitoring of MRD in patients with hematologic malignancies, some preanalytical biases that affect analysis need to be overcome.
MRD analyses are becoming progressively more refined and represent the main tool to design treatment protocols in ALL, always aimed at a more profound eradication of the disease and at curing more patients.
A word of concern is required, as the management of ALL patients of all ages is always becoming more laboratory-driven, from the diagnostic and prognostic work-up to the close monitoring of MRD during the different phases of the disease, leading to progressively more personalized treatment strategies. How doable is this worldwide? This aspect cannot be ignored, as this complex approach is paramount in order to offer optimal management to ALL patients and to increase the likelihood of a cure.

Author Contributions

I.D.S. wrote the manuscript; L.A.D.N., L.E., V.B. and M.B., performed RQ-PCR and ddPCR analysis; R.S. and D.C. performed next-generation sequencing analysis; S.C. revised the manuscript; A.G. and R.F. revised and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The review is based on results obtained thanks to the support of the Associazione Italiana per la Ricerca sul Cancro (AIRC), Special 5 × 1000 Programs “Metastatic disease: the key unmet need in oncology” (No. 21198), Milan (Italy), to R.F. and Progetti di Rilevante Interesse Nazionale (PRIN) Italia, 2017PPS2X4 project, to S.C.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Inaba, H.; Greaves, M.; Mullighan, C.G. Acute lymphoblastic leukaemia. Lancet 2013, 381, 1943–1955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Faderl, S.; O’Brien, S.; Pui, C.H.; Stock, W.; Wetzler, M.; Hoelzer, D.; Kantarjian, H.M. Adult acute lymphoblastic leukemia: Concepts and strategies. Cancer 2010, 116, 1165–1176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Redaelli, A.; Laskin, B.L.; Stephens, J.M.; Botteman, M.F.; Pashos, C.L. A systematic literature review of the clinical and epidemiological burden of acute lymphoblastic leukaemia (ALL). Eur. J. Cancer Care 2005, 14, 53–62. [Google Scholar] [CrossRef]
  4. Pui, C.H.; Evans, W.E. Treatment of acute lymphoblastic leukemia. N. Engl. J. Med. 2006, 354, 166–178. [Google Scholar] [CrossRef]
  5. Siegel, R.; Miller, K.; Jemal, A. Cancer statistics 2015. CA Cancer J. Clin. 2015, 65, 5–29. [Google Scholar] [CrossRef] [PubMed]
  6. Gökbuget, N.; Raff, R.; Brüggemann, M.; Flohr, T.; Scheuring, U.; Pfeifer, H.; Bartram, C.R.; Kneba, M.; Hoelzer, D. Risk/MRD adapted GMALL trials in adult ALL. Ann. Hematol. 2004, 83 (Suppl. S1), S129–S131. [Google Scholar] [PubMed]
  7. Conter, V.; Bartram, C.R.; Valsecchi, M.G.; Schrauder, A.; Panzer-Grumayer, R.; Moricke, A.; Aricò, M.; Zimmermann, M.; Mann, G.; De Rossi, G.; et al. Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: Results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood 2010, 115, 3206–3214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Stary, J.; Zimmermann, M.; Campbell, M.; Castillo, L.; Dibar, E.; Donska, S.; Gonzalez, A.; Izraeli, S.; Janic, D.; Jazbec, J.; et al. Intensive chemotherapy for childhood acute lymphoblastic leukemia: Results of the randomized intercontinental trial ALL IC-BFM 2002. J. Clin. Oncol. 2014, 32, 174–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Vora, A.; Goulden, N.; Wade, R.; Mitchell, C.; Hancock, J.; Hough, R.; Rowntree, C.; Richards, S. Treatment reduction for children and young adults with low-risk acute lymphoblastic leukaemia defined by minimal residual disease (UKALL 2003): A randomised controlled trial. Lancet Oncol. 2013, 14, 199–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Pieters, R.; de Groot-Kruseman, H.; Van der Velden, V.; Fiocco, M.; van den Berg, H.; de Bont, E.; Maarten Egeler, R.; Hoogerbrugge, P.; Kaspers, G.; Van der Schoot, E.; et al. Successful therapy reduction and intensification for childhood acute lymphoblastic leukemia based on minimal residual disease monitoring: Study ALL10 From the Dutch Childhood Oncology Group. J. Clin. Oncol. 2016, 34, 2591–2601. [Google Scholar] [CrossRef]
  11. Tzoneva, G.; Perez-Garcia, A.; Carpenter, Z.; Khiabanian, H.; Tosello, V.; Allegretta, M.; Paietta, E.; Racevskis, J.; Rowe, J.M.; Tallman, M.S.; et al. Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL. Nat. Med. 2013, 19, 368–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Einsiedel, H.G.; Von Stackelberg, A.; Hartmann, R.; Fengler, R.; Schrappe, M.; Janka-Schaub, G.; Mann, G.; Hählen, K.; Göbel, U.; Klingebiel, T.; et al. Long-term outcome in NGS for MRD Assessment in ALL 489 children with relapsed ALL by risk-stratified salvage therapy: Results of trial Acute Lymphoblastic Leukemia-Relapse Study of the Berlin-Frankfurt-Mu¨nster Group 87. J. Clin. Oncol. 2005, 23, 7942–7950. [Google Scholar] [CrossRef] [PubMed]
  13. Fielding, A.K.; Richards, S.M.; Chopra, R.; Lazarus, H.M.; Litzow, M.R.; Buck, G.; Durrant, I.J.; Luger, S.M.; Marks, D.I.; Franklin, I.M.; et al. Medical Research Council of the United Kingdom Adult ALL Working Party; Eastern Cooperative Oncology Group. Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study. Blood 2007, 109, 944–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gökbuget, N.; Stanze, D.; Beck, J.; Diedrich, H.; Horst, H.A.; Hüttmann, A.; Kobbe, G.; Kreuzer, K.A.; Leimer, L.; Reichle, A.; et al. German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia. Outcome of relapsed adult lymphoblastic leukemia depends on response to salvage chemotherapy, prognostic factors, and performance of stem cell transplantation. Blood 2012, 120, 2032–2041. [Google Scholar] [CrossRef]
  15. Bassan, R.; Pavoni, C.; Intermesoli, T.; Spinelli, O.; Tosi, M.; Audisio, E.; Marmont, F.; Cattaneo, C.; Borlenghi, E.; Cortelazzo, S.; et al. Updated risk-oriented strategy for acute lymphoblastic leukemia in adult patients 18–65 years: NILG ALL 10/07. Blood Cancer J. 2020, 10, 119. [Google Scholar] [CrossRef]
  16. Foà, R.; Chiaretti, S. Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2022, 386, 2399–2411. [Google Scholar] [CrossRef]
  17. Cavé, H.; van der Werff ten Bosch, J.; Suciu, S.; Guidal, C.; Waterkeyn, C.; Otten, J.; Bakkus, M.; Thielemans, K.; Grandchamp, B.; Vilmer, E. European Organization for Research and Treatment of Cancer–Childhood Leukemia Cooperative Group. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia. European Organization for Research and Treatment of Cancer—Childhood Leukemia Cooperative Group. N. Engl. J. Med. 1998, 339, 591–598. [Google Scholar]
  18. Borowitz, M.J.; Devidas, M.; Hunger, S.P.; Bowman, W.P.; Carroll, A.J.; Carroll, W.L.; Linda, S.; Martin, P.L.; Pullen, D.J.; Viswanatha, D.; et al. Children’s Oncology Group. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: A Children’s Oncology Group study. Blood 2008, 111, 5477–5485. [Google Scholar] [CrossRef] [Green Version]
  19. Raff, T.; Gökbuget, N.; Lüschen, S.; Reutzel, R.; Ritgen, M.; Irmer, S.; Böttcher, S.; Horst, H.A.; Kneba, M.; Hoelzer, D.; et al. GMALL Study Group. Molecular relapse in adult standard-risk ALL patients detected by prospective MRD monitoring during and after maintenance treatment: Data from the GMALL 06/99 and 07/03 trials. Blood 2007, 109, 910–915. [Google Scholar] [CrossRef] [Green Version]
  20. Gökbuget, N.; Kneba, M.; Raff, T.; Trautmann, H.; Bartram, C.R.; Arnold, R.; Fietkau, R.; Freund, M.; Ganser, A.; Ludwig, W.D.; et al. German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia. Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies. Blood 2012, 120, 1868–1876. [Google Scholar] [CrossRef] [Green Version]
  21. van Dongen, J.J.M.; van der Velden, V.H.J.; Brüggemann, M.; Orfao, A. Minimal residual disease diagnostics in acute lymphoblastic leukemia: Need for sensitive, fast, and standardized technologies. Blood 2015, 125, 3996–4009. [Google Scholar] [CrossRef] [PubMed]
  22. Nyvold, C.G. Critical methodological factors in diagnosing minimal residual disease in hematological malignancies using quantitative PCR. Expert Rev. Mol. Diagn. 2015, 15, 581–584. [Google Scholar] [CrossRef] [PubMed]
  23. van der Velden, V.H.; Hochhaus, A.; Cazzaniga, G.; Szczepanski, T.; Gabert, J.; van Dongen, J.J. Detection of minimal residual disease in hematologic malignancies by real-time quantitative PCR: Principles, approaches, and laboratory aspects. Leukemia 2003, 17, 1013–1034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. van der Velden, V.H.; Cazzaniga, G.; Schrauder, A.; Hancock, J.; Bader, P.; Panzer-Grumayer, E.R.; Flohr, T.; Sutton, R.; Cave, H.; Madsen, H.O.; et al. European Study Group on MRD detection in ALL (ESG-MRD-ALL). Analysis of minimal residual disease by Ig/TCR gene rearrangements: Guidelines for interpretation of real-time quantitative PCR data. Leukemia 2007, 21, 604–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Campana, D. Status of minimal residual disease testing in childhood haematological malignancies. Br. J. Haematol. 2008, 143, 481–489. [Google Scholar] [CrossRef]
  26. Flohr, T.; Schrauder, A.; Cazzaniga, G.; Panzer-Grümayer, R.; van der Velden, V.; Fischer, S.; Stanulla, M.; Basso, G.; Niggli, F.K.; Schäfer, B.W.; et al. International BFM Study Group (I-BFM-SG). Minimal residual disease directed risk stratification using real-time quantitative PCR analysis of immunoglobulin and T-cell receptor gene rearrangements in the international multicenter trial AIEOP-BFM ALL 2000 for childhood acute lymphoblastic leukemia. Leukemia 2008, 22, 771–782. [Google Scholar]
  27. Dworzak, M.N.; Gaipa, G.; Ratei, R.; Veltroni, M.; Schumich, A.; Maglia, O.; Karawajew, L.; Benetello, A.; Pötschger, U.; Husak, Z.; et al. Standardization of flow cytometric minimal residual disease evaluation in acute lymphoblastic leukemia: Multicentric assessment is feasible. Cytom. B Clin. Cytom. 2008, 74, 331–340. [Google Scholar] [CrossRef]
  28. Cazzaniga, G.; Valsecchi, M.G.; Gaipa, G.; Conter, V.; Biondi, A. Defining the correct role of minimal residual disease tests in the management of acute lymphoblastic leukaemia. Br. J. Haematol. 2011, 155, 45–52. [Google Scholar] [CrossRef]
  29. Pfeifer, H.; Cazzaniga, G.; van der Velden, V.H.J.; Cayuela, J.M.; Schäfer, B.; Spinelli, O.; Akiki, S.; Avigad, S.; Bendit, I.; Borg, K.; et al. Standardisation and consensus guidelines for minimal residual disease assessment in Philadelphia-positive acute lymphoblastic leukemia (Ph + ALL) by real-time quantitative reverse transcriptase PCR of e1a2 BCR-ABL1. Leukemia 2019, 33, 1910–1922. [Google Scholar] [CrossRef]
  30. van der Velden, V.H.J.; van Dongen, J.J.M. MRD detection in acute lymphoblastic leukemia patients using Ig/TCR gene rearrangements as targets for real-time quantitative PCR. Methods Mol. Biol. 2009, 538, 115–150. [Google Scholar]
  31. Langerak, A.W.; Szczepański, T.; van der Burg, M.; Wolvers-Tettero, I.L.; van Dongen, J.J.M. Heteroduplex PCR analysis of rearranged T cell receptor genes for clonality assessment in suspect T cell proliferations. Leukemia 1997, 11, 2192–2199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Germano, G.; del Giudice, L.; Palatron, S.; Giarin, E.; Cazzaniga, G.; Biondi, A.; Basso, G. Clonality profile in relapsed precursor-B-ALL children by GeneScan and sequencing analyses. Consequences on minimal residual disease monitoring. Leukemia 2003, 17, 1573–1582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Verhagen, O.J.; Willemse, M.J.; Breunis, W.B.; Wijkhuijs, A.J.; Jacobs, D.C.; Joosten, S.A.; van Wering, E.R.; van Dongen, J.J.; van der Schoot, C.E. Application of germline IGH probes in real-time quantitative PCR for the detection of minimal residual disease in acute lymphoblastic leukemia. Leukemia 2000, 14, 1426–1435. [Google Scholar] [CrossRef] [PubMed]
  34. Szczepański, T.; van der Velden, V.H.; Raff, T.; Jacobs, D.C.; van Wering, E.R.; Brüggemann, M.; Kneba, M.; van Dongen, J.J.M. Comparative analysis of T-cell receptor gene rearrangements at diagnosis and relapse of T-cell acute lymphoblastic leukemia (T-ALL) shows high stability of clonal markers for monitoring of minimal residual disease and reveals the occurrence of second T-ALL. Leukemia 2003, 17, 2149–2156. [Google Scholar] [CrossRef] [Green Version]
  35. Eckert, C.; Flohr, T.; Koehler, R.; Hagedorn, N.; Moericke, A.; Stanulla, M.; Kirschner-Schwabe, R.; Cario, G.; Stackelberg, A.; Bartram, C.R.; et al. Very early/early relapses of acute lymphoblastic leukemia show unexpected changes of clonal markers and high heterogeneity in response to initial and relapse treatment. Leukemia 2011, 25, 1305–1313. [Google Scholar] [CrossRef] [Green Version]
  36. van Dongen, J.J.; Macintyre, E.A.; Gabert, J.A.; Delabesse, E.; Rossi, V.; Saglio, G.; Gottardi, E.; Rambaldi, A.; Dotti, G.; Griesinger, F.; et al. Standardized RT-PCR analysis of fusion gene transcripts from chromosome aberrations in acute leukemia for detection of minimal residual disease. Report of the BIOMED-1 Concerted Action: Investigation of minimal residual disease in acute leukemia. Leukemia 1999, 13, 1901–1928. [Google Scholar] [CrossRef]
  37. Gabert, J.; Beillard, E.; van der Velden, V.H.; Bi, W.; Grimwade, D.; Pallisgaard, N.; Barbany, G.; Cazzaniga, G.M.; 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] [Green Version]
  38. Chiaretti, S.; Vitale, A.; Cazzaniga, G.; Orlando, S.M.; Silvestri, D.; Fazi, P.; Valsecchi, M.G.; Elia, L.; Testi, A.M.; Mancini, F.; et al. Clinico-biological features of 5202 patients with acute lymphoblastic leukemia enrolled in the Italian AIEOP and GIMEMA protocols and stratified in age cohorts. Haematologica 2013, 98, 1702–1710. [Google Scholar] [CrossRef]
  39. D’Angiò, M.; Valsecchi, M.G.; Testi, A.M.; Conter, V.; Nunes, V.; Parasole, R.; Colombini, A.; Santoro, N.; Varotto, S.; Caniglia, M.; et al. Clinical features and outcome of SIL/TAL1-positive T-cell acute lymphoblastic leukemia in children and adolescents: A 10-year experience of the AIEOP group. Haematologica 2015, 100, e10–e13. [Google Scholar] [CrossRef]
  40. Elia, L.; Mancini, M.; Moleti, L.; Meloni, G.; Buffolino, S.; Krampera, M.; De Rossi, G.; Foà, R.; Cimino, G. A multiplex reverse transcriptase polymerase chain reaction strategy for the diagnostic molecular screening of chimeric genes: A clinical evaluation on 170 patients with acute lymphoblastic leukemia. Haematologica 2003, 88, 275–279. [Google Scholar]
  41. Campana, D. Determination of minimal residual disease in leukaemia patients. Br. J. Haematol. 2003, 121, 823–838. [Google Scholar] [CrossRef] [PubMed]
  42. Huggett, J.F.; Whale, A. Digital PCR as a novel technology and its potential implications for molecular diagnostics. Clin. Chem. 2013, 59, 1691–1693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Dube, S.; Qin, J.; Ramakrishnan, R. Mathematical analysis of copy number variation in a DNA sample using digital PCR on a nanofluidic device. PLoS ONE 2008, 3, e2876. [Google Scholar] [CrossRef] [Green Version]
  44. Whale, A.S.; Cowen, S.; Foy, C.A.; Huggett, J.F. Methods for applying accurate digital PCR analysis on low copy DNA samples. PLoS ONE 2013, 8, e58177. [Google Scholar] [CrossRef] [PubMed]
  45. Williams, R.; Peisajovich, S.G.; Miller, O.J.; Magdassi, S.; Tawfik, D.S.; Griths, A.D. Amplification of complex gene libraries by emulsion PCR. Nat. Methods 2006, 3, 545–550. [Google Scholar] [CrossRef] [PubMed]
  46. Hindson, B.J.; Ness, K.D.; Masquelier, D.A.; Belgrader, P.; Heredia, N.J.; Makarewicz, A.J.; Bright, I.J.; Lucero, M.Y.; Hiddessen, A.L.; Legler, T.C.; et al. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal. Chem. 2011, 83, 8604–8610. [Google Scholar] [CrossRef] [PubMed]
  47. Pinheiro, L.B.; Coleman, V.A.; Hindson, C.M.; Herrmann, J.; Hindson, B.J.; Bhat, S.; Emslie, K.R. Evaluation of a droplet digital polymerase chain reaction format for DNA copy number quantification. Anal. Chem. 2012, 84, 1003–1011. [Google Scholar] [CrossRef]
  48. Sanders, R.; Huggett, J.F.; Bushell, C.A.; Cowen, S.; Scott, D.J.; Foy, C.A. Evaluation of digital PCR for absolute DNA quantification. Anal. Chem. 2011, 83, 6474–6484. [Google Scholar] [CrossRef]
  49. Vincent, M.E.; Liu, W.; Haney, E.B.; Ismagilov, R.F. Microfluidic stochastic confinement enhances analysis of rare cells by isolating cells and creating high density environments for control of diffusible signals. Chem. Soc. Rev. 2010, 39, 974–984. [Google Scholar] [CrossRef] [Green Version]
  50. Goh, H.G.; Lin, M.; Fukushima, T.; Saglio, G.; Kim, D.; Choi, S.Y.; Kim, S.H.; Lee, J.; Lee, Y.S.; Oh, S.M.; et al. Sensitive quantitation of minimal residual disease in chronic myeloid leukemia using nanofluidic digital polymerase chain reaction assay. Leuk. Lymphoma 2011, 52, 896–904. [Google Scholar] [CrossRef]
  51. Alikian, M.; Whale, A.S.; Akiki, S.; Piechocki, K.; Torrado, C.; Myint, T.; Cowen, S.; Griths, M.; Reid, A.G.; Apperley, J.; et al. RT-qPCR and RT-digital PCR: A comparison of dierent platforms for the evaluation of residual disease in chronic myeloid leukemia. Clin. Chem. 2017, 63, 525–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Jennings, L.J.; George, D.; Czech, J.; Yu, M.; Joseph, L. Detection and quantification of BCR-ABL1 fusion transcripts by droplet digital PCR. J. Mol. Diagn. 2014, 16, 174–179. [Google Scholar] [CrossRef] [PubMed]
  53. Madic, J.; Zoetic, A.; Sengis, V.; Fradet, E.; Andre, B.; Muller, S.; Dangla, R.; Droniou, M.E. Three-color crystal digital PCR. Biomol. Detect. Quantif. 2016, 10, 34–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Low, H.; Chan, S.J.; Soo, G.H.; Ling, B.; Tan, E.L. Clarity TM digital PCR system: A novel platform for absolute quantification of nucleic acids. Anal. Bioanal. Chem. 2017, 409, 1869–1875. [Google Scholar] [CrossRef]
  55. Whale, A.S.; Huggett, J.F.; Cowen, S.; Speirs, V.; Shaw, J.; Ellison, S.; Foy, C.A.; Scott, D.J. Comparison of microfluidic digital PCR and conventional quantitative PCR for measuring copy number variation. Nucleic Acids Res. 2012, 40, e82. [Google Scholar] [CrossRef]
  56. Tsui, N.B.; Kadir, R.A.; Chan, K.C.; Chi, C.; Mellars, G.; Tuddenham, E.G.; Leung, T.Y.; Lau, T.K.; Chiu, R.W.K.; Lo, Y.M.D. Non-invasive prenatal diagnosis of hemophilia by microfluidics digital PCR analysis of maternal plasma DNA. Blood 2011, 117, 3684–3691. [Google Scholar] [CrossRef] [Green Version]
  57. Pornprasert, S.; Prasing, W. Detection of alpha (0)-thalassemia South-East Asian-type deletion by droplet digital PCR. Eur. J. Haematol. 2014, 92, 244–248. [Google Scholar] [CrossRef]
  58. Stahl, T.; Böhme, M.U.; Kröger, N.; Fehse, B. Digital PCR to assess hematopoietic chimerism after allogeneic stem cell transplantation. Exp. Hematol. 2015, 43, 462–468. [Google Scholar] [CrossRef]
  59. Zagaria, A.; Anelli, L.; Coccaro, N.; Tota, G.; Casieri, P.; Cellamare, A.; Impera, L.; Brunetti, C.; Minervini, A.; Minervini, C.F.; et al. BCR-ABL1 e6a2 transcript in chronic myeloid leukemia: Biological features and molecular monitoring by droplet digital PCR. Virchows Arch. 2015, 467, 357–363. [Google Scholar] [CrossRef]
  60. Albano, F.; Zagaria, A.; Anelli, L.; Coccaro, N.; Tota, G.; Brunetti, C.; Minervini, C.F.; Impera, L.; Minervini, A.; Cellamare, A.; et al. Absolute quantification of the pre-treatment PML-RARA transcript defines the relapse risk in acute promyelocytic leukemia. Oncotarget 2015, 6, 13269–13277. [Google Scholar] [CrossRef] [Green Version]
  61. Coccaro, N.; Anelli, L.; Zagaria, A.; Casieri, P.; Tota, G.; Orsini, P.; Impera, L.; Minervini, A.; Minervini, C.F.; Cumbo, C.; et al. Droplet digital PCR is a robust tool for monitoring minimal residual disease in adult Philadelphia-positive acute lymphoblastic leukemia. J. Mol. Diagn. 2018, 20, 474–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Drandi, D.; Kubiczkova-Besse, L.; Ferrero, S.; Dani, N.; Passera, R.; Mantoan, B.; Gambella, M.; Monitillo, L.; Saraci, F.; Ghione, P.; et al. Minimal residual disease detection by droplet digital PCR in multiple myeloma, mantle cell lymphoma, and follicular lymphoma: A comparison with real-time PCR. J. Mol. Diagn. 2015, 17, 652–660. [Google Scholar] [CrossRef] [PubMed]
  63. Della Starza, I.; Nunes, V.; Cavalli, M.; De Novi, L.A.; Ilari, C.; Apicella, V.; Vitale, A.; Testi, A.M.; Del Giudice, I.; Chiaretti, S.; et al. Comparative analysis between RQ-PCR and digital-droplet-PCR of immunoglobulin/Tcell receptor gene rearrangements to monitor minimal residual disease in acute lymphoblastic leukaemia. Br. J. Haematol. 2016, 174, 541–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cavalli, M.; De Novi, L.A.; Della Starza, I.; Cappelli, L.V.; Nunes, V.; Pulsoni, A.; Del Giudice, I.; Guarini, A.; Foa, R. Comparative analysis between RQ-PCR and digital droplet PCR of BCL2/IGH gene rearrangement in the peripheral blood and bone marrow of early-stage follicular lymphoma. Br. J. Haematol. 2017, 177, 588–596. [Google Scholar] [CrossRef] [PubMed]
  65. Della Starza, I.; Cavalli, M.; De Novi, L.A.; Genuardi, E.; Mantoan, B.; Drandi, D.; Barbero, D.; Ciabatti, E.; Grassi, S.; Gazzola, A.; et al. Minimal residual disease (MRD) in non-Hodgkin lymphomas: Interlaboratory reproducibility on marrow samples with very low levels of disease within the FIL (Fondazione Italiana Linfomi) MRD Network. Hematol. Oncol. 2019, 37, 368–374. [Google Scholar] [CrossRef] [PubMed]
  66. Drandi, D.; Alcantara, M.; Benmaad, I.; Söhlbrandt, A.; Lhermitte, L.; Zaccaria, G.; Ferrante, M.; Genuardi, E.; Mantoan, B.; Villarese, P.; et al. Droplet digital PCR quantification of mantle cell lymphoma follow-up samples from four prospective trials of the European MCL network. HemaSphere 2020, 4, e347. [Google Scholar] [CrossRef] [PubMed]
  67. Della Starza, I.; De Novi, L.A.; Santoro, A.; Salemi, D.; Tam, W.; Cavalli, M.; Menale, L.; Soscia, R.; Apicella, V.; Ilari, C.; et al. Digital droplet PCR and next-generation sequencing refine minimal residual disease monitoring in acute lymphoblastic leukemia. Leuk. Lymphoma 2019, 60, 2838–2840. [Google Scholar] [CrossRef]
  68. Della Starza, I.; De Novi, L.A.; Santoro, A.; Salemi, D.; Spinelli, O.; Tosi, M.; Soscia, R.; Paoloni, F.; Cappelli, L.V.; Cavalli, M.; et al. Digital Droplet PCR Is a Reliable Tool to Improve Minimal Residual Disease Stratification in Adult Philadelphia-Negative Acute Lymphoblastic Leukemia. J. Mol. Diagn. 2022, 24, 893–900. [Google Scholar] [CrossRef]
  69. Schwinghammer, C.; Koopmann, J.; Chitadze, G.; Karawajew, L.; Brüggemann, M.; Eckert, C. A Droplet Digital PCR.: New View on Minimal Residual Disease Quantification in Acute Lymphoblastic Leukemia. J. Mol. Diagn. 2022, 24, 856–866. [Google Scholar] [CrossRef]
  70. Iacobucci, I.; Lonetti, A.; Venturi, C.; Ferrari, A.; Papayannidis, C.; Ottaviani, E.; Abbenante, M.C.; Paolini, S.; Bresciani, P.; Potenza, L.; et al. Use of a high sensitive nanofluidic array for the detection of rare copies of BCR-ABL1 transcript in patients with Philadelphia-positive acute lymphoblastic leukemia in complete response. Leuk. Res. 2014, 38, 581–585. [Google Scholar] [CrossRef]
  71. Foà, R.; Bassan, R.; Vitale, A.; Elia, L.; Piciocchi, A.; Puzzolo, M.C.; Canichella, M.; Viero, P.; Ferrara, F.; Lunghi, M.; et al. Dasatinib-Blinatumomab for Ph-Positive Acute Lymphoblastic Leukemia in Adults. N. Engl. J. Med. 2020, 383, 1613–1623. [Google Scholar] [CrossRef] [PubMed]
  72. Ansuinelli, M.; Della Starza, I.; Lauretti, A.; Elia, L.; Siravo, V.; Messina, M.; De Novi, L.A.; Taherinasab, A.; Canichella, M.; Guarini, A.; et al. Applicability of droplet digital polymerase chain reaction for minimal residual disease in Philadelphia-positive acute lymphoblastic leukaemia. Hematol. Oncol. 2021, 39, 680–686. [Google Scholar] [CrossRef] [PubMed]
  73. Hovorkova, L.; Zaliova, M.; Venn, N.C.; Bleckmann, K.; Trkova, M.; Potuckova, E.; Vaskova, M.; Linhartova, J.; Machova Polakova, K.; Fronkova, E.; et al. Monitoring of childhood ALL using BCR-ABL1 genomic breakpoints identifies a subgroup with CML-like biology. Blood 2017, 129, 2771–2781. [Google Scholar] [CrossRef] [PubMed]
  74. Cazzaniga, G.; De Lorenzo, P.; Alten, J.; Röttgers, S.; Hancock, J.; Saha, V.; Castor, A.; O Madsen, H.; Gandemer, V.; Cavé, H.; et al. Predictive value of minimal residual disease in Philadelphia-chromosome-positive acute lymphoblastic leukemia treated with imatinib in the European intergroup study of post-induction treatment of Philadelphia-chromosome-positive acute lymphoblastic leukemia, based on immunoglobulin/T-cell receptor and BCR/ABL1 methodologies. Haematologica 2018, 103, 107–115. [Google Scholar] [PubMed]
  75. Clappier, E.; Kim, R.; Cayuela, J.M.; Rousselot, P.; Chalandon, Y.; Passet, M.; Thomas, X.; Havelange, V.; Chevallier, P.; Huguet, F.; et al. Persistent BCR-ABL1 clonal hematopoiesis after blast clearance identifies a CML-like subgroup of patients with Philadelphia chromosome-positive (Ph+) ALL: Interim results from GRAAPH-2014 trial. In Proceedings of the 23rd EHA Annual Congress, Stockolm, Sweden, 14–17 June 2018; p. S1568. [Google Scholar]
  76. Della Starza, I.; Nunes, V.; Lovisa, V.; Silvestri, D.; Cavalli, M.; Garofalo, A.; Campeggio, M.; De Novi, L.A.; Soscia, R.; Oggioni, C.; et al. Droplet Digital PCR Improves IG-/TR-based MRD Risk Definition in Childhood B-cell Precursor Acute Lymphoblastic Leukemia. Hemasphere 2021, 5, e543. [Google Scholar] [CrossRef] [PubMed]
  77. Drandi, D.; Ferrero, S.; Ladetto, M. Droplet Digital PCR for Minimal Residual Disease Detection in Mature Lymphoproliferative Disorders. Methods Mol. Biol. 2018, 1768, 229–256. [Google Scholar] [PubMed]
  78. Whale, A.S.; De Spiegelaere, W.; Trypsteen, W.; Nour, A.A.; Bae, Y.K.; Benes, V.; Burke, D.; Cleveland, M.; Corbisier, P.; Devonshire, A.S.; et al. The Digital MIQE Guidelines Update: Minimum Information for Publication of Quantitative Digital PCR Experiments for 2020. Clin. Chem. 2020, 66, 1012–1029. [Google Scholar]
  79. Della Starza, I.; Eckert, C.; Drandi, D.; Cazzaniga, G.; on behalf of the EuroMRD Consortium. Minimal residual Disease Analysis by Monitoring Immunoglobulin and T-Cell Receptor Gene rearrangements by Quantitative PCR and Droplet Digital PCR. Methods Mol. Biol. 2022, 2453, 79–89. [Google Scholar]
  80. van Bladel, D.A.G.; van der Last-Kempkes, J.L.M.; Scheijen, B.; Groenen, P.J.T.A.; on behalf of the EuroClonality Consortium. Next-Generation Sequencing-Based Clonality Detection of Immunoglobulin Gene Rearrangements in B-Cell Lymphoma. Methods Mol. Biol. 2022, 2453, 7–42. [Google Scholar]
  81. Scheijen, B.; Meijers, R.W.J.; Rijntjes, J.; van der Klift, M.Y.; Möbs, M.; Steinhilber, J.; Reigl, T.; van den Brand, M.; Kotrova, M.; Ritter, J.M.; et al. Next-generation sequencing of immunoglobulin gene rearrangements for clonality assessment: A technical feasibility study by EuroClonality-NGS. Leukemia 2019, 33, 2227–2240. [Google Scholar] [CrossRef] [Green Version]
  82. Merriman, B.; Rothberg, J.M. Progress in ion torrent semiconductor chip-based sequencing. Electrophoresis 2012, 33, 3397–3417. [Google Scholar] [CrossRef] [PubMed]
  83. Bentley, D.R.; Balasubramanian, S.; Swerdlow, H.P.; Smith, G.P.; Milton, J.; Brown, C.G.; Hall, K.P.; Evers, D.J.; Barnes, C.L.; Bignell, H.R.; et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature 2008, 456, 53–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Wu, D.; Sherwood, A.; Fromm, J.R.; Winter, S.S.; Dunsmore, K.P.; Loh, M.L.; Greisman, H.A.; Sabath, D.E.; Wood, B.L.; Robins, H. High-throughput sequencing detects minimal residual disease in acute T lymphoblastic leukemia. Sci. Transl. Med. 2012, 4, 134ra63. [Google Scholar] [CrossRef] [PubMed]
  85. Kotrova, M.; Muzikova, K.; Mejstrikova, E.; Novakova, M.; Bakardjieva-Mihaylova, V.; Fiser, K.; Stuchly, J.; Giraud, M.; Salson, M.I.; Pott, C.; et al. The predictive strength of next-generation sequencing MRD detection for relapse compared with current methods in childhood ALL. Blood 2015, 126, 1045–1047. [Google Scholar] [CrossRef] [PubMed]
  86. Ladetto, M.; Brïggemann, M.; Monitillo, L.; Ferrero, S.; Pepin, F.; Drandi, D.; Barbero, D.; Palumbo, A.; Passera, R.; Boccadoro, M.; et al. Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders. Leukemia 2014, 28, 1299–1307. [Google Scholar] [CrossRef]
  87. Logan, A.C.; Zhang, B.; Narasimhan, B.; Carlton, V.; Zheng, J.; Moorhead, M.; Krampf, M.R.; Jones, C.D.; Waqar, A.N.; Faham, M.; et al. Minimal residual disease quantification using consensus primers and high-throughput IGH sequencing predicts post-transplant relapse in chronic lymphocytic leukemia. Leukemia 2013, 27, 1659–1665. [Google Scholar] [CrossRef] [PubMed]
  88. van Dongen, J.J.M.; Seriu, T.; Panzer-Grumayer, E.R.; Biondi, A.; Pongers-Willemse, M.J.; Corral, L.; Stolz, F.; Schrappe, M.; Masera, G.; Kamps, W.A.; et al. Prognostic value of minimal residual disease in acute lymphoblastic leukaemia in childhood. Lancet 1998, 352, 1731–1738. [Google Scholar] [CrossRef]
  89. Gawad, C.; Pepin, F.; Carlton, V.E.H.; Klinger, M.; Logan, A.C.; Miklos, D.B.; Faham, M.; Dahl, G.; Lacayo, N. Massive evolution of the immunoglobulin heavy chain locus in children with B precursor acute lymphoblastic leukemia. Blood 2012, 120, 4407–4417. [Google Scholar] [CrossRef] [Green Version]
  90. Kitchingman, G.R. Immunoglobulin heavy chain gene VH-D junctional diversity at diagnosis in patients with acute lymphoblastic leukemia. Blood 1993, 81, 775–782. [Google Scholar] [CrossRef] [Green Version]
  91. de Haas, V.; Verhagen, O.J.; von dem Borne, A.E.; Kroes, W.; van den Berg, H.; van der Schoot, C.E. Quantification of minimal residual disease in children with oligoclonal B-precursor acute lymphoblastic leukemia indicates that the clones that grow out during relapse already have the slowest rate of reduction during induction therapy. Leukemia 2001, 15, 134–140. [Google Scholar] [CrossRef] [Green Version]
  92. Theunissen, P.M.J.; van Zessen, D.; Stubbs, A.P.; Faham, M.; Zwaan, C.M.; van Dongen, J.J.M.; Van Der Velden, V.H.J. Antigen receptor sequencing of paired bone marrow samples shows homogeneous distribution of acute lymphoblastic leukemia subclones. Haematologica 2017, 102, 1869–1877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bashford-Rogers, R.J.M.; Nicolaou, K.A.; Bartram, J.; Goulden, N.J.; Loizou, L.; Koumas, L.; Chi, J.; Hubank, M.; Kellam, P.; Costeas, P.A.; et al. Eye on the B-ALL: B-cell receptor repertoires reveal persistence of numerous B-lymphoblastic leukemia subclones from diagnosis to relapse. Leukemia 2016, 30, 2312–2321. [Google Scholar] [CrossRef] [PubMed]
  94. Salson, M.; Giraud, M.; Caillault, A.; Grardel, N.; Duployez, N.; Ferret, Y.; Duez, M.; Herbert, R.; Rocher, T.; Sebda, S.; et al. High-throughput sequencing in acute lymphoblastic leukemia: Follow-up of minimal residual disease and emergence of new clones. Leuk. Res. 2017, 53, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Theunissen, P.M.J.; de Bie, M.; van Zessen, D.; de Haas, V.; Stubbs, A.P.; van der Velden, V.H.J. Next-generation antigen receptor sequencing of paired diagnosis and relapse samples of B-cell acute lymphoblastic leukemia: Clonal evolution and implications for minimal residual disease target selection. Leuk. Res. 2019, 76, 98–104. [Google Scholar] [CrossRef] [PubMed]
  96. Kotrova, M.; van der Velden, V.H.J.; van Dongen, J.J.M.; Formankova, R.; Sedlacek, P.; Brüggemann, M.; Zuna, J.; Stary, J.; Trka, J.; Fronkova, E. Next-generation sequencing indicates false-positive MRD results and better predicts prognosis after SCT in patients with childhood ALL. Bone Marrow Transpl. 2017, 52, 962–968. [Google Scholar] [CrossRef] [Green Version]
  97. Bruggemann, M.; Kotrova, M.; Knecht, H.; Bartram, J.; Boudjogrha, M.; Bystry, V.; Fazio, G.; Froňková, E.; Giraud, M.; Grioni, A.; et al. Standardized next-generation sequencing of immunoglobulin and T-cell receptor gene recombinations for MRD marker identification in acute lymphoblastic leukaemia; a EuroClonality-NGS validation study. Leukemia 2019, 33, 2241–2253. [Google Scholar] [CrossRef] [Green Version]
  98. Knecht, H.; Reigl, T.; Kotrova, M.; Appelt, F.; Stewart, P.; Bystry, V.; Krejci, A.; Grioni, A.; Pal, K.; Stranska, K.; et al. Quality control and quantification in IG/TR next-generation sequencing marker identification: Protocols and bioinformatic functionalities by EuroClonality-NGS. Leukemia 2019, 33, 2254–2265. [Google Scholar] [CrossRef]
  99. Stewart, J.P.; Gazdova, J.; Darzentas, N.; Wren, D.; Proszek, P.; Fazio, G.; Songia, S.; Alcoceba, M.; Sarasquete, M.E.; Villarese, P.; et al. Validation of the EuroClonality-NGS DNA capture panel as an integrated genomic tool for lymphoproliferative disorders. Blood Adv. 2021, 5, 3188–3198. [Google Scholar]
  100. Bystry, V.; Reigl, T.; Krejci, A.; Demko, M.; Hanakova, B.; Grioni, A.; Knecht, H.; Schlitt, M.; Dreger, P.; Sellner, L.; et al. ARResT/Interrogate: An interactive immunoprofiler for IG/TR NGS data. Bioinformatics 2017, 33, 435–437. [Google Scholar] [CrossRef] [Green Version]
  101. ClonoSEQ Cleared for Residual Cancer Testing. Cancer Discov. 2018, 8, OF6. [CrossRef] [Green Version]
  102. Lay, L.; Stroup, B.; Payton, J.E. Validation and interpretation of IGH and TCR clonality testing by Ion Torrent S5 NGS for diagnosis and disease monitoring in B and T cell cancers. Pract. Lab. Med. 2020, 22, e00191. [Google Scholar] [CrossRef] [PubMed]
  103. Ho, C.C.; Tungy, J.K.; Zehnder, J.L.; Zhangy, B.M. Validation of a Next-Generation Sequencing Based T-Cell Receptor Gamma Gene Rearrangement Diagnostic Assay Transitioning from Capillary Electrophoresis to Next-Generation Sequencing. J. Mol. Diagn. 2021, 23, 805–815. [Google Scholar] [CrossRef] [PubMed]
  104. Ha, J.; Lee, H.; Shin, S.; Cho, H.; Chung, H.; Jang, J.E.; Kim, S.J.; Cheong, J.W.; Lee, S.T.; Kim, J.S.; et al. Ig Gene Clonality Analysis Using Next Generation Sequencing for Improved Minimal Residual Disease Detection with Significant Prognostic Value in Multiple Myeloma Patients. J. Mol. Diagn. 2022, 24, 48–56. [Google Scholar] [CrossRef] [PubMed]
  105. Arcila, M.E.; Yu, W.; Syed, M.; Kim, H.; Maciag, L.; Yao, J.J.; Ho, C.; Petrova, K.; Moung, C.; Salazar, P.; et al. Establishment of Immunoglobulin Heavy (IGH) Chain Clonality Testing by Next-Generation Sequencing for Routine Characterization of B-Cell and Plasma Cell Neoplasms. J. Mol. Diagn. 2019, 21, 330–342. [Google Scholar] [CrossRef] [Green Version]
  106. Takamatsu, H. Comparison of Minimal Residual Disease Detection by Multiparameter Flow Cytometry, ASO-qPCR, Droplet Digital PCR, and Deep Sequencing in Patients with Multiple Myeloma Who Underwent Autologous Stem Cell Transplantation. J. Clin. Med. 2017, 6, 91. [Google Scholar] [CrossRef] [Green Version]
  107. Pedini, P.; Cherouat, N.; Basire, A.; Simon, S.; Budon, L.; Pourtein, M.; Grondin, S.; Moskovtchenko, P.; Chiaroni, J.; Michel, G.; et al. Evaluation of Next-Generation Sequencing and Crystal Digital PCR for Chimerism Monitoring of Post-Allogeneic Hematopoietic Stem Cell Transplantation. Transplant. Cell. Ther. 2021, 27, 89.e1–89.e10. [Google Scholar] [CrossRef]
  108. Brüggemann, M.; Kotrova, M. Minimal residual disease in adult ALL: Technical aspects and implications for correct clinical interpretation. Blood Adv. 2017, 1, 2456–2466. [Google Scholar] [CrossRef]
  109. Coustan-Smith, E.; Sancho, J.; Hancock, M.L.; Razzouk, B.I.; Ribeiro, C.R.; Rivera, G.K.; Rubnitz, J.E.; Sandlund, J.T.; Pui, C.H.; Campana, D. Use of peripheral blood instead of bone marrow to monitor residual disease in children with acute lymphoblastic leukemia. Blood 2002, 100, 2399–2402. [Google Scholar] [CrossRef]
  110. Alix-Panabières, C.; Schwarzenbach, H.; Pantel, K. Circulating Tumor Cells and Circulating Tumor DNA. Annu. Rev. Med. 2012, 63, 199–215. [Google Scholar] [CrossRef]
  111. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating Liquid Biopsies into the Management of Cancer. Nat. Rev. Clin. Oncol. 2017, 14, 531–548. [Google Scholar] [CrossRef]
  112. Gorgannezhad, L.; Umer, M.; Islam, M.N.; Nguyen, N.T.; Shiddiky, M.J.A. Circulating tumor DNA and liquid biopsy: Opportunities, challenges, and recent advances in detection technologies. Lab Chip 2018, 18, 1174–1196. [Google Scholar] [CrossRef] [PubMed]
  113. Salehi, M.; Sharifi, M. Exosomal MiRNAs as Novel Cancer Biomarkers: Challenges and Opportunities. J. Cell. Physiol. 2018, 233, 6370–6380. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, L.; Bode, A.M.; Dong, Z. Circulating Tumor Cells: Moving Biological Insights into Detection. Theranostics 2017, 7, 2606–2619. [Google Scholar] [CrossRef] [PubMed]
  115. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Buedts, L.; Vandenberghe, P. Circulating cell-free DNA in hematological malignancies. Haematologica 2016, 101, 997–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Rossi, D.; Diop, F.; Spaccarotella, E.; Monti, S.; Zanni, M.; Rasi, S.; Deambrogi, C.; Spina, V.; Bruscaggin, A.; Favini, C.; et al. Diffuse large B-cell lymphoma genotyping on the liquid biopsy. Blood 2017, 129, 1947–1957. [Google Scholar] [CrossRef] [PubMed]
  118. Lim, J.K.; Kuss, B.; Talaulikar, D. Role of cell-free DNA in haematological malignancies. Pathology 2021, 53, 416–426. [Google Scholar] [CrossRef]
  119. Bingham, N.; Spencer, A. The role of cell free DNA and liquid biopsies in haematological conditions. Cancer Drug Resist. 2020, 3, 521–531. [Google Scholar] [CrossRef]
  120. Colmenares, R.; Álvarez, N.; Barrio, S.; Martinez-López, J.; Ayala, R. The Minimal Residual Disease Using Liquid Biopsies in Hematological Malignancies. Cancers 2022, 14, 1310. [Google Scholar] [CrossRef]
  121. Schwarz, A.K.; Stanulla, M.; Cario, G.; Flohr, T.; Sutton, R.; Möricke, A.; Anker, P.; Stroun, M.; Welte, K.; Bartram, C.R.; et al. Quantification of Free Total Plasma DNA and Minimal Residual Disease Detection in the Plasma of Children with Acute Lymphoblastic Leukemia. Ann. Hematol. 2009, 88, 897–905. [Google Scholar] [CrossRef] [Green Version]
  122. Cheng, S.H.; Lau, K.M.; Li, C.K.; Chan, N.P.H.; Ip, R.K.L.; Cheng, C.K.; Lee, V.; Shing, M.M.K.; Leung, A.W.K.; Ha, S.Y.; et al. Minimal Residual Disease-Based Risk Stratification in Chinese Childhood Acute Lymphoblastic Leukemia by Flow Cytometry and Plasma DNA Quantitative Polymerase Chain Reaction. PLoS ONE 2013, 8, e69467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Ikeuchi, K.; Doki, N.; Takao, A.; Hishima, T.; Yamada, Y.; Konishi, T.; Nagata, A.; Takezaki, T.; Kaito, S.; Kurosawa, S.; et al. Extramedullary Gastric Relapse at the Time of Bone Marrow Relapse of Acute Lymphoblastic Leukemia after Allogeneic Bone Marrow Transplantation. Intern. Med. 2017, 56, 3215–3217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Aldoss, I.; Song, J.Y. Extramedullary relapse of KMT2A(MLL)-rearranged acute lymphoblastic leukemia with lineage switch following blinatumomab. Blood 2018, 131, 2507. [Google Scholar] [CrossRef] [PubMed]
  125. Demosthenous, C.; Lalayanni, C.; Iskas, M.; Douka, V.; Pastelli, N.; Anagnostopoulos, A. Extramedullary relapse and discordant CD19 expression between bone marrow and extramedullary sites in relapsed acute lymphoblastic leukemia after blinatumomab treatment. Curr. Probl. Cancer 2019, 43, 222–227. [Google Scholar] [CrossRef] [PubMed]
  126. Bronkhorst, A.J.; Aucamp, J.; Pretorius, P.J. Cell-free DNA: Preanalytical variables. Clin. Chim. Acta 2015, 450, 243–253. [Google Scholar] [CrossRef] [PubMed]
  127. Cavallone, L.; Aldamry, M.; Lafleur, J.; Lan, C.; Gonzalez Ginestet, P.; Alirezaie, N.; Ferrario, C.; Aguilar-Mahecha, A.; Basik, M. A Study of Pre-Analytical Variables and Optimization of Extraction Method for Circulating Tumor DNA Measurements by Digital Droplet PCR. Cancer Epidemiol. Biomark. Prev. 2019, 28, 909–916. [Google Scholar] [CrossRef] [PubMed]
Figure 1. ddPCR technology. This figure reports an analytical diagram for a ddPCR experiment. The mix reaction and the DNA samples are partitioned into 20,000 droplets (1-step), and then both are amplified in a thermal cycler (2-step). Finally, each droplet is individually analyzed by a droplet reader (3-step). Each droplet is plotted on the graph of fluorescence intensity, and the concentration is calculated on the fraction of droplets that does not contain any target DNA using software. The positive droplets are fitted to a Poisson algorithm to determine absolute copy number expressed as copies per 1 μL.
Figure 1. ddPCR technology. This figure reports an analytical diagram for a ddPCR experiment. The mix reaction and the DNA samples are partitioned into 20,000 droplets (1-step), and then both are amplified in a thermal cycler (2-step). Finally, each droplet is individually analyzed by a droplet reader (3-step). Each droplet is plotted on the graph of fluorescence intensity, and the concentration is calculated on the fraction of droplets that does not contain any target DNA using software. The positive droplets are fitted to a Poisson algorithm to determine absolute copy number expressed as copies per 1 μL.
Cancers 15 00374 g001
Figure 2. NGS analysis. A library is prepared by fragmentation and conjugation with adaptive sequences, composed with few nucleotides (1-step), and subsequently, it is amplified and sequenced, with the production of so-called ≪reads≫ (2-step). Data analysis is performed through the use of bioinformatic tools, matching the results to a reference genome (3-step).
Figure 2. NGS analysis. A library is prepared by fragmentation and conjugation with adaptive sequences, composed with few nucleotides (1-step), and subsequently, it is amplified and sequenced, with the production of so-called ≪reads≫ (2-step). Data analysis is performed through the use of bioinformatic tools, matching the results to a reference genome (3-step).
Cancers 15 00374 g002
Figure 3. cfDNA as novel compartment for molecular MRD monitoring in ALL.
Figure 3. cfDNA as novel compartment for molecular MRD monitoring in ALL.
Cancers 15 00374 g003
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

Della Starza, I.; De Novi, L.A.; Elia, L.; Bellomarino, V.; Beldinanzi, M.; Soscia, R.; Cardinali, D.; Chiaretti, S.; Guarini, A.; Foà, R. Optimizing Molecular Minimal Residual Disease Analysis in Adult Acute Lymphoblastic Leukemia. Cancers 2023, 15, 374. https://doi.org/10.3390/cancers15020374

AMA Style

Della Starza I, De Novi LA, Elia L, Bellomarino V, Beldinanzi M, Soscia R, Cardinali D, Chiaretti S, Guarini A, Foà R. Optimizing Molecular Minimal Residual Disease Analysis in Adult Acute Lymphoblastic Leukemia. Cancers. 2023; 15(2):374. https://doi.org/10.3390/cancers15020374

Chicago/Turabian Style

Della Starza, Irene, Lucia Anna De Novi, Loredana Elia, Vittorio Bellomarino, Marco Beldinanzi, Roberta Soscia, Deborah Cardinali, Sabina Chiaretti, Anna Guarini, and Robin Foà. 2023. "Optimizing Molecular Minimal Residual Disease Analysis in Adult Acute Lymphoblastic Leukemia" Cancers 15, no. 2: 374. https://doi.org/10.3390/cancers15020374

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