Molecular Minimal Residual Disease Monitoring in Acute Myeloid Leukemia

Molecular Minimal Residual Disease Monitoring in Acute Myeloid Leukemia

Accepted Manuscript Molecular Minimal Residual Disease Monitoring in Acute Myeloid Leukemia: Challenges and Future Directions Adrian Selim, Andrew S. ...

NAN Sizes 0 Downloads 88 Views

Accepted Manuscript Molecular Minimal Residual Disease Monitoring in Acute Myeloid Leukemia: Challenges and Future Directions Adrian Selim, Andrew S. Moore PII:

S1525-1578(17)30485-3

DOI:

10.1016/j.jmoldx.2018.03.005

Reference:

JMDI 689

To appear in:

The Journal of Molecular Diagnostics

Accepted Date: 27 March 2018

Please cite this article as: Selim A, Moore AS, Molecular Minimal Residual Disease Monitoring in Acute Myeloid Leukemia: Challenges and Future Directions, The Journal of Molecular Diagnostics (2018), doi: 10.1016/j.jmoldx.2018.03.005. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

ACCEPTED MANUSCRIPT

Molecular minimal residual disease monitoring in acute myeloid leukaemia: challenges and future directions Adrian Selim* and Andrew S. Moore*†‡

RI PT

From the The University of Queensland Diamantina Institute,* Translational Research Institute, Brisbane; the Oncology Services Group,† Children’s Health Queensland Hospital and Health Service, Brisbane; and the UQ Child Health Research Centre,‡ The University of

SC

Queensland, Brisbane, Australia

M AN U

Correspondence: Andrew S. Moore Children’s Health Queensland Research Directorate Level 7, Centre for Children’s Health Research

62 Graham St, South Brisbane, Queensland, 4101, Australia

TE D

Email: [email protected]

EP

Running Head: Molecular MRD in AML

AC C

Funding: A.S.M. is supported by the Children’s Hospital Foundation, Queensland. Disclosures: None declared.

2

ACCEPTED MANUSCRIPT Abstract

The ability to sensitively monitor minimal residual disease (MRD) has played a key role in improving the management and outcomes for a number of leukaemias, particularly acute promyelocytic leukaemia and childhood acute lymphoblastic leukaemia. In contrast, MRD

RI PT

monitoring in acute myeloid leukaemia (AML) has been limited by variable assay methodologies and a relative paucity of patient-specific MRD markers. Inter- and intra-tumor genetic heterogeneity pose significant challenges for the identification of molecular markers

SC

suitable for MRD monitoring in AML, particularly for those cases without structural

M AN U

chromosomal rearrangements associated with fusion genes. Furthermore, the need to discriminate which mutations may be suitable for MRD monitoring creates additional complexity. The mainstay of current molecular MRD monitoring is real-time quantitative PCR, targeting fusion genes, mutations, and gene over-expression. New technologies, particularly next-generation sequencing approaches, offer new ways to overcome these

TE D

limitations. Here, we review the techniques available for molecular MRD monitoring in AML

AC C

EP

and discuss their utility in clinical practice.

3

ACCEPTED MANUSCRIPT Introduction

Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by diverse genetic abnormalities and variable morphology, immunophenotypes, and clinical outcomes1. Genetic heterogeneity exists not only between different AML cases but within each individual case

RI PT

and mutations in AML may pre-date leukaemia-initiating events, occur after initiation, and contribute to the founding AML clone, or occur sub-clonally after leukaemogenesis2. Thus, the clonal composition of the leukaemia can vary significantly, as sub-clones (along with

SC

their late acquired mutations) are subjected to natural and treatment-induced selection

M AN U

pressures, expanding, diminishing, or even being cleared3-5. These factors present challenges when selecting genetic abnormalities as markers of post-treatment minimal residual disease (MRD) in AML. Despite this, the impetus for MRD monitoring in AML is ever-present, as countless studies have established the role of MRD as a dynamic predictor of relapse risk and overall survival, often stronger than any pre-treatment variables1. Moreover, the ultimate aim

TE D

of MRD monitoring in leukaemia is to be able to utilize this dynamic marker to personalize therapy, such that treatments and their associated toxicities are curbed in low-risk patients,

EP

and intensified in those at high risk of relapse.

The characteristics of a suitable MRD marker can be defined by ‘3 Ss’; a marker must be

AC C

‘specific’, ‘sensitive’, and ‘stable’. Moreover, some would argue for a fourth characteristic, ‘stemness’, whereby a marker is present only in those cells capable of self-renewal and hence relapse, and not in bulk leukaemic cells1. Although such markers are being actively pursued in the context of multicolor flow cytometric MRD determination6, it remains a largely philosophical ideal in the molecular context. The terms ‘sensitive’ and ‘specific’ can be a source of confusion. As characteristics of the MRD marker, a highly sensitive marker is one that is present in all leukemic cells, and a highly specific marker is one that is only present in leukaemic cells while being absent in non-leukemic/pre-leukemic cells. A lack of sensitivity

4

ACCEPTED MANUSCRIPT

will produce false negative MRD, whereas a lack of specificity will produce false positives. The second use of the term sensitivity is the analytical sensitivity, a performance characteristic of the assay, more appropriately described as the lower limit of detection (LOD). A commonly accepted benchmark in LOD in AML MRD assays is 10-3 (0.1%),

RI PT

meaning one leukemic cell (or equivalent) in 1,000 cells measured, although some molecular MRD assays have LODs of 10-5 to 10-7. The theoretical benefit of utilizing more sensitive assays is an ability to identify more patients at risk of relapse, and to quantitatively track

SC

minute changes in low-level MRD. Despite a lack of strong evidence for MRD-directed therapeutic approaches in non-APL AML, by employing a sequential MRD surveillance

M AN U

strategy, one could expect better outcomes when therapy is initiated at the time of ‘molecular relapse’ rather than frank haematological relapse. Risk-adapted treatment protocols taking into account sequential MRD results have shown favorable results in paediatric AML (AML027), although the study relied on comparison to historical cohorts. Hopefully, with the

TE D

results of prospective, randomized clinical trials assessing the value of MRD-directed therapy, such as the United Kingdom’s NCRI AML17 trial, which randomized patients with informative molecular markers to “MRD monitoring” vs “no MRD monitoring” strategies,

EP

the question of whether modification of post-remission treatment intensity based on MRD

AC C

status in AML will be answered8.

The mainstay of molecular MRD monitoring at present are real-time quantitative PCR (RQPCR)–based methods, which can be employed when a suitable target is present, afford high analytical sensitivity, and, to a significant extent, allow for standardization between different laboratories. Targets for RQ-PCR as well as other molecular methods can be divided into three categories: i) chimeric fusion gene transcripts; ii) gene mutations; and iii) overexpressed genes.

5

ACCEPTED MANUSCRIPT

PCR-based assays of fusion genes and NPM1 mutations have the potential to collectively monitor approximately 60% of AML presenting in children and younger adults; however, this proportion steadily declines with increasing age, encompassing only 32% of those over 609 . PCR of over-expressed genes, most commonly WT1, can be applied in a significant

RI PT

proportion of AML cases (ranging from 13% to 73% in different studies depending on whether PB or BM is used and the threshold for defining over-expression10, 11), but with its limited dynamic range, is generally restricted to those who lack other molecular markers.

SC

The nucleic acid source for molecular assays may be genomic DNA or RNA, each with a set

M AN U

of advantages and disadvantages. DNA is more stable and easier to extract than RNA, making it favorable for collection, processing, transport, and storage. In the case of fusion genes, breakpoints at the gDNA level are unique, providing patient-specific markers of MRD, reducing the risk of cross-contamination between different samples in the laboratory, and essentially eliminating the possibility of amplifying non-leukaemic clones12,

13

. Moreover,

TE D

DNA provides an absolute quantitation of the number of leukaemic cells in a sample, as opposed to RNA-based assays, which are also affected by mRNA expression levels and

EP

mRNA stability. On the other hand, use of DNA requires sequencing and individualized primer/probe design, significantly increasing complexity, time of implementation, and cost,

AC C

compared with RNA transcripts which lack intronic segments and are often restricted to a small number of common variants amenable to standardized primer/probe sets for screening. Moreover, gene over-expression assays utilize the differential expression of genes between normal and leukaemic cells and must therefore be RNA-based. These considerations have resulted in limited interest in DNA-based monitoring in routine practice, with RNA-based RQ-PCR methods predominating.13 Sample quality is particularly important for RNA, and quantitation of a control gene in parallel with the target of interest provides insight into sample quality and reverse transcriptase efficiency. 14 Overall, the accuracy of an MRD test is

6

ACCEPTED MANUSCRIPT

determined by the diagnostic sensitivity and specificity of the marker, the prevalence of the leukemic cell in the sampled tissue (ie, bone marrow or peripheral blood), the sample size collected (as well as its quality), and the lower limit of detection of the assay. 15 RQ-PCR–based MRD approaches are highly amenable to standardization, as demonstrated

RI PT

over a decade ago by the Europe Against Cancer collaboration, whereby quantitative assays for nine of the most common fusion genes in leukemias were standardized across 26

SC

laboratories, and later for WT1 assays by the European LeukemiaNet.10, 16

As with any clinical laboratory testing, cost, throughput, and turn-around-times are critical

M AN U

considerations. Compared with multiparametric flow cytometry–based MRD, in which each sample is assayed individually, with results potentially available within one day, most molecular MRD tests must be batched to be economically viable.

Here we review the three classes of molecular targets mentioned above, including factors

TE D

which affect their identification, as well as their suitability for MRD monitoring, and discuss future directions in molecular MRD monitoring with a particular focus on next-generation

Fusion genes

EP

sequencing (NGS) applications. A summary is presented in Table 1.17-23

AC C

The monitoring of fusion genes is possible in AML cases characterized by a recurrent chromosomal rearrangement, which allows fusion gene transcripts such as RUNX1RUNX1T1, CBFB-MYH11, DEK-NUP214, NUP98-NSD1, and a myriad of KMT2A (MLL) fusions to be quantitated, most commonly by RNA-based RQ-PCR. These rearrangements have provided the most robust molecular MRD markers, and have allowed monitoring for approximately 50% of children, 30% of adults aged 15 to 60, and 10% of adults aged >60.23 Assay sensitivities are among the highest available, often approaching 10-5. From a practical viewpoint, the number of probe/primer pairs required to screen for these abnormalities is

7

ACCEPTED MANUSCRIPT

variable, and determined by the level of conservation of breakpoints for each respective rearrangement. For example, there is only a single type of RUNX1-RUNX1T1 transcript, greatly simplifying its screening. On the other hand, in screening for its CBFB counterpart (CBFB-MYH11), three different reverse primers along with a common forward primer and

RI PT

probe are required to identify the three commonest break-points to cover approximately 95% of cases.16

In current routine practice, a subset of AML cases with fusion events suitable for MRD

SC

monitoring will be missed, including cytogenetically cryptic, uncommon, and novel fusions.

M AN U

That is because we currently rely upon the diagnostic karyotype ± a limited FISH and/or a screening PCR panel for their identification, thereby limiting detection based on prior knowledge and common abnormalities. Moreover, in many cases the diagnostic services required to identify or confirm rearrangements by FISH, PCR, or sequencing and/or identify breakpoints to design PCR-based MRD assays are unavailable. As an example, there are at

TE D

least 28 different rearrangements of the NUP98 gene in haematopoietic malignancies.24 In AML, the commonest of these is the NUP98-NSD1 fusion, which is enriched in paediatric AML cases, and produces a cytogenetically cryptic t(5;11), with frequency reported to range

EP

from 3.6% to 7.4% in unselected paediatric AML series25. Traditionally, to identify a specific

AC C

NUP98 rearrangement, it first needs to be screened for by a FISH break-apart probe, then confirmation of the partner gene would require further FISH analysis if a suitable probe was available, PCR if a putative gene was suspected, or if truly novel, would require sequencing. Other factors also reduce FISH probe hybridization such as deletion within the target region, as was seen in two out of 22 NUP98 rearranged cases in one study, in which the NUP98 FISH probe failed, prompting RNA sequencing for further characterization25. It is therefore evident that in order to identify as many fusion events suitable for MRD monitoring as possible, an agnostic method with a high sensitivity for fusion gene detection is highly

8

ACCEPTED MANUSCRIPT

desirable. One promising approach is RNA-based NGS assays which are discussed in detail later.

RI PT

Gene Mutations Currently only one recurrent mutation that of the NPM1 gene has emerged as a suitable MRD marker in AML. The commonest mutation in adult AML, NPM1 mutations (NPM1mut), are

SC

present in 30% to 35% of all AML cases and 50% to 60% of AML with normal karyotype26. Ivey et al elegantly showed that monitoring NPM1mut MRD by RQ-PCR using a mutation-

M AN U

specific primer with a common primer and probe afforded a median sensitivity of 10-5, reliably predicted relapse on sequential monitoring, was stably present at relapse in 99% of cases and gave highly informative information to guide the decision to undertake stem cell transplantation.20

TE D

To understand what constitutes a suitable gene mutation for MRD purposes, a basic understanding of AML leukaemogenesis is required. The majority of mutations in an AML genome are actually random events that occurred within a haematopoietic stem/progenitor

EP

cell prior to the acquisition of a leukaemia initiating event, or in many cases a few initiating,

AC C

or so-called driver mutations. Subsequently, the progenitor acquires a number of cooperating mutations, and gives rise to the founding clone that is detected at diagnosis.2 Further cooperating mutations do occur and can contribute to disease progression/relapse but often only in a subset of leukemic cells, giving rise to sub-clones. Therefore, a mutation suitable for MRD monitoring is one occurring between the leukaemia-initiating event and the emergence of the founding clone, visually represented in Figure 1. Related to this concept is that of the variant allele frequency (VAF) of a mutation, which is the fraction of tumor cells that harbor a specific mutation, assuming a relatively pure

9

ACCEPTED MANUSCRIPT

leukemic sample and a diploid genome, variations of which reflect intra-tumoral heterogeneity.27 The archetypal initiating events are the aforementioned recurrent structural rearrangements which produce fusion genes, and are present in almost all leukaemic cells, resulting in a VAF of approximately 0.5. On the other hand, it is challenging to identify

RI PT

driver mutations in cases such as AML with normal karyotype. In this setting, the VAF can be helpful, as initiating/driver mutations should have VAF approaching 0.5 for heterozygous mutations (sensitive), persist within sub-clones, and therefore detectable at relapse (stable).

SC

However, for some early driver mutations, specificity becomes a problem, due to the existence of the mutation in a pre-leukaemic state. This is particularly relevant for mutations

M AN U

in epigenetic modifiers, such as DNTM3A and IDH1/2 which are common in adult AML. In NPM1-mutated cases, Ivey et al found many cases of persistence of DNMT3A and IDH mutations at high levels in patients who achieved NPM1-MRD negativity and maintained long-term remission.20 Furthermore, in five of 15 de novo AML cases, Wong et al

TE D

demonstrated “rising clones” in one or more remission samples that were not detected by enhanced exome sequencing at diagnosis, enriched for mutations in TP53, TET2, DNMT3A, and ASXL1, all of which were shown to be present in rare cells before therapy by more

EP

sensitive techniques28. These findings are in line with our current understanding of clonal haematopoiesis of indeterminate potential (CHIP), and the use of these mutations in the MRD

AC C

context would therefore lead to false positivity in many instances.20,29 Unfortunately, many of the most common recurrent mutations in AML such as those within tyrosine kinase genes such as FLT3 and KIT, or in the RAS/MAPK/MEK pathway such as NRAS and PTPN11, are often present at lower VAF, suggesting that these mutations are often late, sub-clonal events.3, 5 Taking the above factors into account, it is becoming increasingly clear that to identify gene mutations suitable for monitoring in AML other than NPM1, a patient-specific approach is

10

ACCEPTED MANUSCRIPT

required, which can only be afforded by NGS-based technologies. With the above issues limiting the use of many recurrent mutations in AML, it is important to note that when a leukaemia-specific mutation is defined as a mutation occurring between the leukaemiainitiating event and the emergence of the founding clone, the mutation need not be common,

SC

discussion of genomic DNA-based NGS below.

RI PT

a driver mutation, or even pathogenic. Identifying such mutations is outlined further in the

Gene Overexpression

M AN U

In patients who lack a leukaemia-specific target, quantitation of gene transcripts overexpressed in AML has aimed to provide a ‘universal’ MRD target. The best characterized of these is WT1, which is overexpressed in the majority of AML cases.20 The sensitivity of assays taking this approach is limited by baseline levels of normal expression in

TE D

peripheral blood and bone marrow. In the case of WT1, unlike most molecular assays, peripheral blood is more sensitive than bone marrow due to its lower baseline WT1 expression. The performance of early WT1 assays was limited by factors which reduced their

EP

specificity and sensitivity, which led to the European LeukaemiaNet (ELN) collaborative effort to select an assay with optimal performance10. It was noted that mutations within the

AC C

WT1 gene, occurring in 10% of AML patients, tended to cluster within the 3’ regions of the gene, leading to false negativity of assays that amplified this region. Other assays had problems with cross-reactivity with genomic DNA. Finally, the best performing of nine WT1 assays was selected. From a cohort of 129 uniformly treated adults, only 46% of patients had sufficiently high diagnostic WT1 expression levels in PB to allow discrimination of at least a 2-log reduction in expression. Despite this limitation, the ELN study and others have demonstrated increased relapse risk for those with sub-optimal reductions in or re-emergence

11

ACCEPTED MANUSCRIPT

of WT1 transcripts at early time-points, pre-HSCT and post-HSCT.11, 30,31 Moreover, Marani et al also demonstrated that incorporation of WT1 log reduction was able to further refine relapse risk in patients with MRD detectable by flow cytometry post-induction, highlighting that orthogonal MRD methods may be complementary when used together rather than

RI PT

provide the same information.

Another contemporary approach to improve the performance of MRD detection by overexpression was evaluated by Steinbach et al, who measured the expression of seven

SC

genes known to be overexpressed in AML (including WT1), along with three housekeeping

M AN U

genes, on the same TaqMan Low Density Array (TLDA)32. Although WT1 was the best performing single marker of MRD positivity, of the 42 follow-up samples that were classified as MRD positive, WT1 was only positive in 20 (48%) of the cases, which means that the gene panel allowed for detection of more than twice the number of MRD-positive cases than WT1

TE D

alone. Moreover, the TLDA provided for a simple, fast, and standardized methodology.

Future directions in molecular MRD monitoring

EP

Droplet digital PCR

AC C

Droplet digital PCR (ddPCR) is a recently commercialized technology that allows precise quantification of target nucleic acids in a sample by first partitioning it into nanoliter-sized, highly uniform, water-in-oil droplets33. This limiting dilution results in either one or zero target molecules in most reactions. After end-point PCR using fluorescently quenched oligonucleotide probes, measurement of fluorescence of each partition individually allows absolute nucleic acid quantification by Poisson statistical analysis of the number of positive and negative reactions34-36. Although the technology may not play a role in the identification of suitable markers for MRD, several characteristics of ddPCR, compared to conventional

12

ACCEPTED MANUSCRIPT

RQ-PCR make it very promising for use in MRD monitoring. Most notable of these is the capacity for absolute quantification independent of a reference standard, robustness to variations in PCR efficiency, and overall simplicity34. There is growing interest in the use of ddPCR for MRD measurement in many haematological malignancies. One such example is

RI PT

in chronic myeloid leukaemia (CML), in which monitoring BCR-ABL1 fusion transcripts by RQ-PCR has become standard of care. In this disease, where second generation tyrosine kinase inhibitors (TKI’s) are resulting in deeper molecular responses, the lower limit of

SC

detection (LOD) of the RQ-PCR method may be insufficient, especially when determining which patients may be eligible for TKI cessation37. Several groups have now demonstrated

M AN U

improved assay sensitivity as well as superior precision with ddPCR of BCR-ABL1 transcripts compared to a conventional RQ-PCR assay37,38. In fact, the LOD can be adjusted to the precise requirement of any application by modifying the amount of input RNA and the number of replicates39, making ddPCR an exciting addition to the MRD armamentarium.

TE D

Recently, a novel application of ddPCR for MRD monitoring of NPM1mut AML was reported40. In this study, common forward primer and probes were used as previously described37, along with a degenerate reverse primer capable of generating oligonucleotides

EP

with all possible tetranucleotide insertions in exon 12 position 863, characteristic of 95% of

AC C

NPM1 mutations. The multiplex assay was tested against mutation-specific ddPCR assays using a multitude of common and rare mutations from patient and cell line samples, with overall excellent concordance between the two assays as well as with RQ-PCR. This could potentially benefit clinical laboratories in testing of rare NPM1 mutations, obviating the need for patient-specific assays, as well as in testing for patients for which the mutant sequence is unknown. In the post allogeneic transplant setting, Brambati et al published their experience performing ddPCR MRD monitoring for DNMT3A and IDH1/2 mutations41. In their series of 89 patients,

13

ACCEPTED MANUSCRIPT

at least one of these mutations was present in 33.7% of the cases. By performing monthly BM evaluation in the first three months, then once every three months for the first year and yearly thereafter, six of the nine patients who relapsed had MRD positivity in the BM immediately preceding relapse, with a median time from first positivity to relapse of 60 days.

RI PT

Conceivably, this provides a feasible time frame for pre-emptive therapy and the high sensitivity (significantly greater than molecular chimerism studies and FISH) should allow

SC

more patients to be treated with low level disease.

M AN U

Next-Generation Sequencing (NGS)

Due to rapidly improving technology and falling cost, the prospect of using NGS for both identification and monitoring of MRD targets is becoming increasingly promising. Again, both genomic DNA and RNA can be utilized and each can be subjected to comprehensive or

TE D

targeted NGS assays. With their inherent strengths and limitations, selection of the correct platform and subsequently the correct mutation(s) to monitor is pivotal.

EP

Genomic DNA sequencing

Klco et al demonstrated the potential to use genomic mutation clearance to define groups

AC C

with differing relapse risk. Although not designed for highly sensitive molecular MRD detection, the study illustrated some of the factors that must be considered in setting up an NGS-based MRD assay.42 For 25 adult AML cases, cryopreserved diagnostic and postinduction samples were subjected to enhanced exome sequencing. From another 25 patients who had previous exome or genome sequencing, DNA extracted from day 30 FFPE BM tissue was used to generate short amplicons (using AmpliseqTM) for deep sequencing of a mean of 12.1 mutations from each case. Although the enhanced exome sequencing provided a mean coverage of leukaemia-associated variants at day 30 of 256X, the deep sequencing of

14

ACCEPTED MANUSCRIPT

the FFPE BM tissue, which was of significantly poorer quality, afforded a mean coverage on day 30 of 14,780X, more than 50 times greater. Factors influencing the sensitivity of an NGS-based MRD assay include sequencing coverage, DNA quantity, and the type of target mutation23. At diagnosis, when identifying

RI PT

leukaemia-specific mutations, use of a comprehensive sequencing platform such as wholeexome sequencing may be suitable, since, while read depth of individual mutations may be low, breadth of coverage may allow many mutations with high VAF to be detected. On

SC

follow-up studies on the other hand, read depth in the regions of interest must be increased

M AN U

substantially to allow for highly sensitive MRD detection, meaning that the sequencing must be targeted.

Another important consideration when deciding which NGS platform is to be used, is the type of mutations to be identified. For example, if the intention is to detect single nucleotide

TE D

polymorphisms (SNPs) and small insertions/deletions (indels) in coding regions, wholeexome sequencing (WES) will provide greater coverage than whole-genome sequencing (WGS). Conversely, if fusion genes are the primary target, WGS is more suited than WES,

EP

which can only detect these structural variations in the subset of rearrangements with breakpoints in or near exons, significantly reducing sensitivity for fusion gene detection.43

AC C

Moreover, fusion events are identified by WGS only through intensive computational analysis, and the large amount of sequencing needed may be restrictive in terms of cost and throughput44. On the other hand, RNA sequencing can be used to interrogate gene fusions as long as they are sufficiently expressed, with the added capacity for targeted assay development, making these the most promising NGS-based methods for identifying as well as potentially monitoring MRD of fusion genes. 12, 43

15

ACCEPTED MANUSCRIPT

Most studies in NGS-based MRD have focused on the monitoring of mutations in the genes most commonly mutated in the AML genome, which has highlighted several shortcomings to this approach. Firstly, as mentioned previously, common mutations often include those which are often present in pre-leukaemic clones (eg, TP53, DNMT3A, and ASXL1), rendering them

RI PT

unsuitable as MRD markers, unless monitoring in the context of allogeneic haematopoietic stem cell transplant (allo-HSCT), where tracking the clearance of this pre-leukaemic clone as a marker of recipient haematopoietic origin may be useful. Secondly, other common,

SC

recurrently mutated genes in AML (eg, FLT3, PTPN11, and NRAS) are often sub-clonal and thus unreliably present at relapse, as noted in several studies.3,

42,45

Furthermore, although

M AN U

mutations in epigenetic regulators are among the commonest mutations in adult AML, studies in childhood AML have demonstrated them to be rare, and in general childhood cases display a relative paucity of mutations, making identification of mutations for MRD even more difficult in this population.3, 46 Thus, it is evident that in many cases NGS is needed to define

TE D

patient- and leukaemia-specific markers. An example of this approach was published by Malmberg and colleagues, who performed WES on FACS-sorted leukaemic cells and lymphocytes from 17 AML patients.23 In doing so, they were able to filter both germline and

EP

early HPSC passenger mutations, and subsequently removed mutations with 95% CI of VAF

AC C

below 0.5, in effect selecting leukaemia-specific mutations as defined earlier. This approach yielded a median of 11 leukaemia-specific mutations per case, with DNMT3A mutations being excluded. Using targeted deep sequencing in one patient and selecting three SNP mutations and a type A NPM1 mutation, they were able to monitor MRD in subsequent samples with high sensitivity (significance threshold set at VAF 0.027% for SNPs and 0.006% for NPM1), and a high level of correlation was demonstrated between mutational loads of each target mutation at the various time-points, as well as with concurrent NPM1mut DNA-based RQ-PCR measurements. Furthermore, all mutations were present at high

16

ACCEPTED MANUSCRIPT

mutational load at relapse. This approach may prove to be a realistic alternative in those patients without other sensitive MRD markers. Finally, one remaining question to be addressed is whether NGS-based MRD has advantages over, or is able to provide additional information to MRD detected by RQ-PCR or other

RI PT

methods. One study which illustrates this potential, involved comparison of MRD detection in NPM1mut AML by targeted deep sequencing and flow cytometry in 22 samples from six patients.22 The first finding here was increased sensitivity, with a limit of detection of

SC

approximately 0.001% MRD in cell dilution studies, an order of magnitude greater than

M AN U

observed in concurrent flow cytometry studies, and the detection of MRD in all clinical samples which were negative by flow cytometry. Secondly, the specificity of leukaemic blast detection is potentially much greater by NGS, owing to immunophenotypic overlap with regenerative blasts in flow cytometry, which could lead to either over or under-estimation of MRD, when compared with the non-limiting specificity of NPM1 mutations in this study,

TE D

with no positive reads in the PB of 20 normal blood donors. Moreover, in addition to the effect of specificity, quantitation of bone marrow MRD is also conceivably more accurate by

EP

NGS as the DNA of early erythroid precursors is included, whereas this is not possible by flow cytometry due to erythroid lysis protocols typically used. NGS-based identification of

AC C

NPM1 mutant sequences for subsequent MRD monitoring is more streamlined than the multistep conventional approaches. However, possibly the most striking finding seen in this study was NPM1-mutant clonal heterogeneity, seen in two of the six patients studied. In one of these patients, although still detectable, the level of the index mutation was overtaken by an alternative NPM1 mutation, which would not be reflected by a mutant-specific PCR assay (although potentially quantitated accurately by the multiplex ddPCR assay described earlier40) and therefore lead to falsely reduced quantitation of disease burden.

17

ACCEPTED MANUSCRIPT RNA Sequencing

As mentioned earlier, RNA-based NGS approaches offer the potential to be both high yield in terms of fusion gene detection, and streamlined – that is, in one step identifying the partner genes as well as provide the specific breakpoint required for design of PCR-based MRD

RI PT

detection. Alternatively, the identified fusion could be monitored by a targeted sequencing assay. Whole transcriptome sequencing can be used to identify both known and novel fusion transcripts through computational tools44. However, target-enrichment strategies may further

SC

enhance detection sensitivity while reducing the cost of sequencing47. Amplicon-based target

M AN U

enrichment strategies require prior knowledge of fusion partners for opposing primer design and are therefore unable to identify novel fusion genes. This and other limitations in cancer fusion gene detection prompted the development of the Anchored Multiplex PCR (AMP) targeted sequencing method, which, from small amounts of input RNA is able to uncover unknown sequences adjacent to a known nucleic acid sequence by using a one-sided nested

TE D

primer approach, enabling fusion detection with only one partner gene targeted 48. AMPenriched targeted RNA sequencing (Archer® FusionPlex®, ArcherDx, Boulder, CO) has

EP

recently been evaluated for MLL-fusion breakpoint identification in childhood leukaemia cases, and compared to long-distance inverse-PCR (LDI-PCR), a gold-standard technique

AC C

which provides patient-specific genomic DNA breakpoints for subsequent MRD monitoring by RQ-PCR21, 49. Of the 39 samples tested harboring nine different MLL fusions, all but one fusion identified by LDI-PCR were detected by AMP, one fusion unable to be characterized by LDI-PCR was identified by AMP, and furthermore, AMP identified multiple MLL-fusion transcript isoforms in 47% of the patients. Importantly, implementation of LDI-PCR for MLL breakpoint identification is still a decision based on prior knowledge or suspicion of an MLL rearrangement usually by cytogenetics/FISH, whereas AMP-enriched sequencing could be applied to unselected patients to identify MLL or any fusion genes expressed in the leukaemia

18

ACCEPTED MANUSCRIPT

for subsequent MRD monitoring. The study provided strong evidence to support the potential use of AMP-enriched targeted RNA sequencing for highly sensitive MRD detection – by performing serial dilutions of MV4-11 (MLL-AFF1) cells in Kasumi-1 cells from 10-2 to 10-7, and subjecting the extracted RNA to AMP-enrichment, the MLL fusion was detectable down

RI PT

to the 10-7 dilution. Moreover, as an NGS assay, leveraging the base pair resolution to

identify sequence variants (point mutations, small insertions/deletions) of relevance to AML is also promising. A pitfall of this technology however, is a significant artefactual false-

SC

positive fusion call rate, requiring rigorous review of breakpoints and extensive test

M AN U

validation.50

Conclusion

MRD monitoring in haematological malignancies is a burgeoning field as it uniquely

TE D

provides a dynamic assessment of relapse risk independent of baseline prognostic evaluation. The experience in both acute promyelocytic leukaemia and childhood acute lymphoblastic leukaemia have demonstrated that MRD-based risk adapted therapies can improve outcomes

EP

in those diseases; however, we eagerly await similar evidence in AML51,52. However, the inter- and intra-tumor genetic heterogeneity in AML has posed challenges in defining

AC C

specific, sensitive, and stable molecular MRD markers for all patients, and with many different assays required, standardization has also proven challenging. As outlined in this review, improving technology, particularly through next-generation sequencing, coupled with an improved understanding of AML leukaemogenesis, is leading to new methods to both identify and monitor suitable MRD targets in this challenging malignancy.

19

ACCEPTED MANUSCRIPT References 1. Hourigan CS, Gale RP, Gormley NJ, Ossenkoppele GJ, Walter RB: Measurable residual disease testing in acute myeloid leukaemia. Leukemia 2017.

2. Welch J, Ley T, Link D, Miller C, Larson D, Koboldt D, Wartman L, Lamprecht Tamara l, Liu F, Xia J,

RI PT

Kandoth C, Fulton Robert s, McLellan Michael d, Dooling David j, Wallis John w, Chen K, Harris Christopher c, Schmidt Heather k, Kalicki-Veizer Joelle m, Lu C, Zhang Q, Lin L, O’laughlin

Michelle d, McMichael Joshua f, Delehaunty Kim d, Fulton Lucinda a, Magrini Vincent j, McGrath

SC

Sean d, Demeter Ryan t, Vickery Tammi l, Hundal J, Cook Lisa l, Swift Gary w, Reed Jerry p,

Alldredge Patricia a, Wylie Todd n, Walker Jason r, Watson Mark a, Heath Sharon e, Shannon

M AN U

William d, Varghese N, Nagarajan R, Payton Jacqueline e, Baty Jack d, Kulkarni S, Klco Jeffery m, Tomasson Michael h, Westervelt P, Walter Matthew j, Graubert Timothy a, Dipersio John f, Ding L, Mardis Elaine r, Wilson Richard k: The Origin and Evolution of Mutations in Acute Myeloid Leukemia. Cell 2012, 150:264-278.

TE D

3. Farrar JE, Schuback HL, Ries RE, Wai D, Hampton OA, Trevino LR, Alonzo TA, Guidry Auvil JM, Davidsen TM, Gesuwan P, Hermida L, Muzny DM, Dewal N, Rustagi N, Lewis LR, Gamis AS, Wheeler DA, Smith MA, Gerhard DS, Meshinchi S: Genomic Profiling of Pediatric Acute Myeloid

EP

Leukemia Reveals a Changing Mutational Landscape from Disease Diagnosis to Relapse. Cancer research 2016, 76:2197.

AC C

4. Metzeler KH, Herold T, Rothenberg-Thurley M, Amler S, Sauerland MC, Görlich D, Schneider S, Konstandin NP, Dufour A, Bräundl K, Ksienzyk B, Zellmeier E, Hartmann L, Greif PA, Fiegl M, Subklewe M, Bohlander SK, Krug U, Faldum A, Berdel WE, Wörmann B, Büchner T, Hiddemann W, Braess J, Spiekermann K: Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood 2016, 128:686. 5. Patel JL, Schumacher JA, Frizzell K, Sorrells S, Shen W, Clayton A, Jattani R, Kelley TW: Coexisting and cooperating mutations in NPM1-mutated acute myeloid leukemia. Leukemia Research 2017, 56:7-12.

20

ACCEPTED MANUSCRIPT 6. Jaso JM, Wang SA, Jorgensen JL, Lin P: Multi-color flow cytometric immunophenotyping for detection of minimal residual disease in AML: past, present and future. Bone Marrow Transplantation 2014. 7. Rubnitz JE, Inaba H, Dahl G, Ribeiro RC, Bowman WP, Taub J, Pounds S, Razzouk BI, Lacayo NJ,

RI PT

Cao X, Meshinchi S, Degar B, Airewele G, Raimondi SC, Onciu M, Coustan-Smith E, Downing JR, Leung W, Pui C-H, Campana D: Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial. Lancet Oncology 2010, 11:543-552.

in acute myeloid leukemia. Blood, 2015. pp. 2331-2335.

SC

8. Kayser S, Schlenk R, Grimwade D, Yosuico V, Walter R: Minimal residual disease-directed therapy

M AN U

9. Grimwade D, Freeman SD: Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for "prime time"? Blood 2014, 124:3345.

10. Cilloni D, Renneville A, Hermitte F, Hills RK, Daly S, Jovanovic JV, Gottardi E, Fava M, Schnittger S, Weiss T, Izzo B, Nomdedeu J, van Der Heijden A, van Der Reijden BA, Jansen JH, van Der Velden

TE D

VHJ, Ommen H, Preudhomme C, Saglio G, Grimwade D: Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: a European LeukemiaNet study. Journal of clinical

EP

oncology : official journal of the American Society of Clinical Oncology 2009, 27:5195. 11. Marani C, Clavio M, Grasso R, Colombo N, Guolo F, Kunkl A, Ballerini F, Giannoni L, Ghiggi C,

AC C

Fugazza G, Ravetti J-L, Gobbi M, Miglino M: Integrating post induction WT1 quantification and flow- cytometry results improves minimal residual disease stratification in acute myeloid leukemia. Leukemia Research 2013, 37:1606-1611. 12. Alikian M, Gale RP, Apperley JF, Foroni L: Molecular techniques for the personalised management of patients with chronic myeloid leukaemia. Biomolecular detection and quantification 2017, 11:4-20. 13. Duployez N, Nibourel O, Marceau-Renaut A, Willekens C, Helevaut N, Caillault A, Villenet C, CelliLebras K, Boissel N, Jourdan E, Dombret H, Figeac M, Preudhomme C, Renneville A: Minimal

21

ACCEPTED MANUSCRIPT

residual disease monitoring in t(8;21) acute myeloid leukemia based on RUNX1-RUNX1T1 fusion quantification on genomic DNA. American Journal of Hematology 2014, 89:610-615. 14. Ross DM, Branford S, Melo JV, Hughes TP: Reply to 'What do we mean by sensitivity when we talk about detecting minimal residual disease?' by Steinbach and Debatin.(Letters to

RI PT

the Editor)(Report). Leukemia 2009, 23:819. 15. Butturini A, Klein J, Gale RP: Modeling minimal residual disease (MRD)-testing. Leukemia Research 2003, 27:293-300.

SC

16. Gabert J, Beillard E, Velden VHJVD, Bi W, Grimwade D, Pallisgaard N, Barbany G, Cazzaniga G, Cayuela JM, Cavé H, Pane F, Aerts JLE, Micheli DD, Thirion X, Pradel V, González M, Viehmann S,

M AN U

Malec M, Saglio G, Dongen JJMV: 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.

TE D

17. Hokland P, Ommen HB, Nyvold CG, Roug AS: Sensitivity of minimal residual disease in acute myeloid leukaemia in first remission – methodologies in relation to their clinical situation. 2012. pp. 569-580.

EP

18. Willekens C, Blanchet O, Renneville A, Cornillet-Lefebvre P, Pautas C, Guieze R, Ifrah N, Dombret H, Jourdan E, Preudhomme C, Boissel N: Prospective long-term minimal residual disease

AC C

monitoring using RQ-PCR in RUNX1-RUNX1T1-positive acute myeloid leukemia: results of the French CBF-2006 trial. Haematologica 2016, 101:328. 19. Ommen HB: Monitoring minimal residual disease in acute myeloid leukaemia: a review of the current evolving strategies. Therapeutic Advances in Hematology 2016, 7:3-16. 20. Ivey A, Hills RK, Simpson MA, Jovanovic JV, Gilkes A, Grech A, Patel Y, Bhudia N, Farah H, Mason J, Wall K, Akiki S, Griffiths M, Solomon E, McCaughan F, Linch DC, Gale RE, Vyas P, Freeman SD, Russell N, Burnett AK, Grimwade D: Assessment of Minimal Residual Disease in Standard-Risk AML. The New England Journal of Medicine 2016, 374:422-433.

22

ACCEPTED MANUSCRIPT 21. Afrin S, Zhang CR, Meyer C, Stinson CL, Pham T, Bruxner TJ, Venn NC, Trahair TN, Sutton R,

Marschalek R, Fink JL, Moore AS: Targeted Next-Generation Sequencing for Detecting MLL Gene Fusions in Leukemia. Molecular Cancer Research 2018, 16:279-285. 22. Stephen JS, Jonathan RF, Jay S, Brent LW, David W: Detection of minimal residual disease in

RI PT

NPM1-mutated acute myeloid leukemia by next-generation sequencing. Modern Pathology 2014, 27.

23. Malmberg EB, Ståhlman S, Rehammar A, Samuelsson T, Alm SJ, Kristiansson E, Abrahamsson J,

SC

Garelius H, Pettersson L, Ehinger M, Palmqvist L, Fogelstrand L: Patient-tailored analysis of

minimal residual disease in acute myeloid leukemia using next generation sequencing. European

M AN U

Journal of Haematology 2016.

24. Gough SM, Slape C, Aplan PD: NUP98 gene fusions and hematopoietic malignancies: common themes and new biologic insights. Blood, 2011. pp. 6247-6257.

25. Struski S, Lagarde S, Bories P, Puiseux C, Prade N, Cuccuini W, Pages MP, Bidet A, Gervais C,

TE D

Lafage-Pochitaloff M, Roche-Lestienne C, Barin C, Penther D, Nadal N, Radford-Weiss I, CollongeRame MA, Gaillard B, Mugneret F, Lefebvre C, Bart-Delabesse E, Petit A, Leverger G, Broccardo C, Luquet I, Pasquet M, Delabesse E: NUP98 is rearranged in 3.8% of pediatric AML forming a

EP

clinical and molecular homogenous group with a poor prognosis. Leukemia 2017, 31:565. 26. Karam A-I, Aziz N: Molecular landscape in acute myeloid leukemia: where do we stand in 2016.

AC C

Cancer Biology & Medicine 2016, 13:474. 27. Sallman DA, Padron E: Integrating mutation variant allele frequency into clinical practice in myeloid malignancies. Hematology/Oncology and Stem Cell Therapy 2016, 9:89-95. 28. Wong TN, Miller CA, Klco JM, Petti A, Demeter R, Helton NM, Li T, Fulton RS, Heath SE, Mardis ER, Westervelt P, Dipersio JF, Walter MJ, Welch JS, Graubert TA, Wilson RK, Ley TJ, Link DC: Rapid expansion of preexisting nonleukemic hematopoietic clones frequently follows induction therapy for de novo AML. Blood 2016, 127:893.

23

ACCEPTED MANUSCRIPT 29. Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA, Hasserjian RP, Ebert BL: Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood 2015, 126:9.

30. Buckley SA, Wood BL, Othus M, Hourigan CS, Ustun C, Linden MA, Defor TE, Malagola M, Anthias

RI PT

C, Valkova V, Kanakry CG, Gruhn B, Buccisano F, Devine B, Walter RB: Minimal residual disease prior to allogeneic hematopoietic cell transplantation in acute myeloid leukemia: a metaanalysis. Haematologica 2017:haematol.2016.159343.

SC

31. Duléry R, Nibourel O, Gauthier J, Elsermans V, Behal H, Coiteux V, Magro L, Renneville A,

Marceau A, Boyer T, Quesnel B, Preudhomme C, Duhamel A, Yakoub-Agha I: Impact of Wilms'

M AN U

tumor 1 expression on outcome of patients undergoing allogeneic stem cell transplantation for AML. Bone marrow transplantation 2017.

32. Steinbach D, Bader P, Willasch A, Bartholomae S, Debatin K-M, Zimmermann M, Creutzig U, Reinhardt D, Gruhn B: Prospective validation of a new method of monitoring minimal residual

TE D

disease in childhood acute myelogenous leukemia. Clinical cancer research : an official journal of the American Association for Cancer Research 2015, 21:1353. 33. Pinheiro LB, Coleman VA, Hindson CM, Herrmann J, Hindson BJ, Bhat S, Emslie KR: Evaluation of

EP

a droplet digital polymerase chain reaction format for DNA copy number quantification. Analytical Chemistry 2012, 84:1003.

AC C

34. Hindson CM, Chevillet JR, Briggs HA, Gallichotte EN, Ruf IK, Hindson BJ, Vessella RL, Tewari M: Absolute quantification by droplet digital PCR versus analog real-time PCR. Nature Methods 2013, 10:1003.

35. Della Starza I, Nunes V, Cavalli M, De Novi LA, Ilari C, Apicella V, Vitale A, Testi AM, Del Giudice I, Chiaretti S, Foà R, Guarini A: Comparative analysis between RQ-PCR and digital-droplet-PCR of immunoglobulin/T-cell receptor gene rearrangements to monitor minimal residual disease in acute lymphoblastic leukaemia. British Journal of Haematology 2016, 174:541-549.

24

ACCEPTED MANUSCRIPT 36. Miotke L, Lau BT, Rumma RT, Ji HP: High sensitivity detection and quantitation of DNA copy

number and single nucleotide variants with single color droplet digital PCR. Analytical chemistry 2014, 86:2618. 37. Jennings LJ, George D, Czech J, Yu M, Joseph L: Detection and Quantification of BCR-ABL1 Fusion

RI PT

Transcripts by Droplet Digital PCR. The Journal of Molecular Diagnostics 2014, 16:174-179. 38. Maier J, Franke G, Schubert K, Wildenberger K, Cross M, Niederwieser D, Lange T: A Comparison of Droplet Digital PCR and Quantitative RT-PCR for Low Level BCR-ABL in CML Patients with

SC

Molecular Responses. Blood 2014, 124.

39. Brunetti C, Anelli L, Zagaria A, Minervini A, Minervini CF, Casieri P, Coccaro N, Cumbo C, Tota G,

M AN U

Impera L, Orsini P, Specchia G, Albano F: Droplet Digital PCR Is a Reliable Tool for Monitoring Minimal Residual Disease in Acute Promyelocytic Leukemia. The Journal of Molecular Diagnostics 2017.

40. Mencia-Trinchant N, Hu Y, Alas MA, Ali F, Wouters BJ, Lee S, Ritchie EK, Desai P, Guzman ML,

TE D

Roboz GJ, Hassane DC: Minimal Residual Disease Monitoring of Acute Myeloid Leukemia by Massively Multiplex Digital PCR in Patients with NPM1 Mutations. The Journal of Molecular Diagnostics 2017, 19:537-548.

EP

41. Brambati C, Galbiati S, Xue E, Toffalori C, Crucitti L, Greco R, Sala E, Crippa A, Chiesa L, Soriani N, Mazzi B, Tresoldi C, Stanghellini M, Peccatori J, Carrabba MG, Bernardi M, Ferrari M, Lampasona

AC C

V, Ciceri F, Vago L: Droplet digital polymerase chain reaction for DNMT3A and IDH1/2 mutations to improve early detection of acute myeloid leukemia relapse after allogeneic hematopoietic stem cell transplantation. Haematologica 2016, 101. 42. Klco J, Miller C, Griffith M, Petti A, Spencer D, Ketkar-Kulkarni S, Wartman L, Christopher M, Lamprecht T, Helton N, Duncavage E, Payton J, Baty J, Heath S, Griffith O, Shen D, Hundal J, Chang G, Fulton R, O'Laughlin M, Bs C, Magrini V, Demeter R, Larson D, Kulkarni S, Ozenberger B, Welch J, Walter M, Graubert T, Westervelt P, Radich J, Link D, Mardis E, Dipersio J, Wilson R, Ley

25

ACCEPTED MANUSCRIPT T: Association Between Mutation Clearance After Induction Therapy and Outcomes in Acute Myeloid Leukemia. Journal of the American Medical Association 2015, 314:811-811.

43. Yang L, Lee M-S, Lu H, Oh D-Y, Kim Yeon j, Park D, Park G, Ren X, Bristow Christopher a, Haseley Psalm s, Lee S, Pantazi A, Kucherlapati R, Park W-Y, Scott Kenneth l, Choi Y-L, Park Peter j:

RI PT

Analyzing Somatic Genome Rearrangements in Human Cancers by Using Whole-Exome Sequencing. The American Journal of Human Genetics 2016, 98:843-856.

44. Wang Q, Xia J, Jia P, Pao W, Zhao Z: Application of next generation sequencing to human gene

SC

fusion detection: computational tools, features and perspectives. Briefings in Bioinformatics 2013, 14:506-519.

M AN U

45. Krönke J, Bullinger L, Teleanu V, Tschürtz F, Gaidzik VI, Kühn MWM, Rücker FG, Holzmann K, Paschka P, Kapp-Schwörer S, Späth D, Kindler T, Schittenhelm M, Krauter J, Ganser A, Göhring G, Schlegelberger B, Schlenk RF, Döhner H, Döhner K: Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia. Blood 2013, 122:100.

TE D

46. Liang D-C, Liu H-C, Yang C-P, Jaing T-H, Hung I-J, Yeh T-C, Chen S-H, Hou J-Y, Huang Y-J, Shih Y-S, Huang Y-H, Lin T-H, Shih L-Y: Cooperating gene mutations in childhood acute myeloid leukemia with special reference on mutations of ASXL1, TET2, IDH1, IDH2, and DNMT3A. Blood 2013,

EP

121:2988.

47. Netto GJ, Schrijver I: Genomic Applications in Pathology. New York, NY: Springer New York: New

AC C

York, NY, 2015. DOI https://doi.org/10.1007/978-1-4939-0727-4. 48. Zheng Z, Liebers M, Zhelyazkova B, Cao Y, Panditi D, Lynch KD, Chen J, Robinson HE, Shim HS, Chmielecki J, Pao W, Engelman JA, Iafrate AJ, Le LP: Anchored multiplex PCR for targeted nextgeneration sequencing. Nature Medicine 2014;20:1479-1484. 49. Meyer C, Burmeister T, Gröger D, Tsaur G, Fechina L, Renneville S, Sutton R, Venn NC, Emerenciano M, Pombo-De-Oliveira MS, Blunck CB, Lopes BA, Zuna J, Trka J, Ballerini P, Lapillonne H, Braekeleer MD, Cazzaniga G, Abascal LC, Velden VHJVD, Delabesse E, Park TS, Oh SH, Silva MLM, Lund-Aho T, Juvonen V, Moore AS, Heidenreich O, Vormoor J, Zerkalenkova E,

26

ACCEPTED MANUSCRIPT Olshanskaya Y, Bueno C, Menendez P, Teigler-Schlegel S, Stadt UZ, Lentes J, Göhring G,

Kustanovich S, Aleinikova O, Schäfer BW, Kubetzko S, Madsen HO, Gruhn B, Duarte X, Gameiro P, Lippert E, Bidet S, Cayuela JM, Clappier E, Alonso CN, Zwaan CM, Heuvel-Eibrink MMVD, Izraeli S, Trakhtenbrot L, Archer P, Hancock J, Möricke A, Alten J, Schrappe M, Stanulla M, Strehl S,

RI PT

Attarbaschi A, Dworzak M, Haas OA, Panzer-Grümayer R, Sedék L, Szczepański T, Caye S, Suarez L, Cavé H, Marschalek R: The MLL recombinome of acute leukemias in 2017. Leukemia 2018, 32:273-284

SC

50. Guseva NV, Tanas MR, Stence AA, Sompallae R, Schade JC, Bossler AD, Bellizzi AM, Ma D: The NAB2–STAT6 gene fusion in solitary fibrous tumor can be reliably detected by anchored

M AN U

multiplexed PCR for targeted next-generation sequencing. Cancer Genetics 2016, 209:303-312. 51. Grimwade D, Jovanovic JV, Hills RK, Nugent EA, Patel Y, Flora R, Diverio D, Jones K, Aslett H, Batson E, Rennie K, Angell R, Clark RE, Solomon E, Lo-Coco F, Wheatley K, Burnett AK: Prospective minimal residual disease monitoring to predict relapse of acute promyelocytic

27:3650-3658.

TE D

leukemia and to direct pre-emptive arsenic trioxide therapy. Journal of clinical oncology 2009,

52. Vora A, Goulden N, Mitchell C, Hancock J, Hough R, Rowntree C, Moorman AV, Wade R:

EP

Augmented post-remission therapy for a minimal residual disease-defined high-risk subgroup of children and young people with clinical standard-risk and intermediate-risk acute lymphoblastic

AC C

leukaemia (UKALL 2003): a randomised controlled trial. The Lancet oncology 2014, 15:809-818.

27

ACCEPTED MANUSCRIPT Figure Legend

Figure 1. Over time, haematopoietic stem/progenitor cells (HSPC) acquire passenger mutations (P), most of which are benign and may be found in mature progeny. Later, this HSPC acquires a leukaemia-initiating mutation (i). If this initiating/early driver mutation commonly exists in a pre-

RI PT

leukaemic state (such as DNMT3A mutations), the mutation may potentially be present in mature progeny. The progenitor acquires further co-operating mutations (c), as well as passenger mutations (p), which gives rise to the founding AML clone (X). Subsequent co-operating and passenger

SC

mutations (c1, c2, p1, p2) occur in subsets of leukaemic cells, giving rise to sub-clones (1 and 2). Thus, suitable mutations for minimal residual disease monitoring are those that are acquired between the

M AN U

leukaemia-initiating event and the emergence of the founding clone (with the exception of mutations commonly present in the pre-leukaemic state). Adapted from Welch et al2, with

AC C

EP

TE D

permission from Elsevier.

28

ACCEPTED MANUSCRIPT Table 1. Characteristics of selected molecular MRD assays. Assay

Nucleic

Analytical Sensitivity

Reference

0.025-0.05%

Hokland et al (2012)17

0.001%

Willekens et al (2016)18

0.025-0.05%

Hokland et al (2012)17

acid

RNA

Fusion gene DEK-NUP214 RUNX1-RUNX1T1 MLL-MLLT3

M AN U

RNA

Gene-

SC

RQ-PCR

RI PT

source

overexpression

>1:100 in 23% *

WT1

Ommen (2016)19

>1:200 in 7 % *

>1:100 in 59% †

TE D

Steinbach 7 gene set

Ommen (2016)19

DNA or

Gene Mutation

RNA NPM1mut

Ivey et al (2016)20

EP

0.001%‡

RNA or

at least equivalent to

DNA

RQ-PCR

AC C

ddPCR

NGS-based

Fusion gene

RNA

10-7 §

Afrin et al (2018)21

NPM1mut DNA

0.001%

Salipante et al (2014)22

Patient-specific SNVs DNA

0.025%

Malmberg et al (2016)23

MLL-AFF1

Gene mutations

DNA

29

ACCEPTED MANUSCRIPT *Adult AML, compared with median normal expression. †Childhood AML, compared with 90% upper normal limit. ‡Median sensi^vity for 27 different NPM1 muta^ons.

AC C

EP

TE D

M AN U

SC

RI PT

§Cellular dilution of MV4-11 cell line in Kasumi-1 cell line. Anchored Multiplex PCR–enriched RNA sequencing (Archer® FusionPlex®Heme panel).

RI PT

ACCEPTED MANUSCRIPT

       

 

SC

 

HSPC

1

Founding clone 

 

P + i 



   

?

P + i + c + p  (X)

P + i 

TE D

     P 

M AN U

 

 

X + c1 + p1

AC C

EP

Mature progeny

Subclones

2 X + c2 + p2 

P = passenger mutations in HSPC  i = initiating mutation(s)  c = co‐operating mutations in founding clone  p = passenger mutations in founding clone   X = P + i + c + p  c1= cooperating mutations in subclone 1  c2= cooperating mutations in subclone 2  p1 = passenger mutations in subclone 1  p2 = passenger mutations in subclone 2