Sunteți pe pagina 1din 14

614529

research-article2015
TAH0010.1177/2040620715614529Therapeutic Advances in HematologyHB Ommen

Therapeutic Advances in Hematology Review

Monitoring minimal residual disease in


Ther Adv Hematol

2016, Vol. 7(1) 316

acute myeloid leukaemia: a review of the DOI: 10.1177/


2040620715614529

current evolving strategies


The Author(s), 2015.
Reprints and permissions:
http://www.sagepub.co.uk/
journalsPermissions.nav

Hans Beier Ommen

Abstract: Several disease-monitoring techniques are available for the physician treating acute
myeloid leukaemia (AML). Besides immunohistochemistry assisted light microscopy, the past
20 years have seen the development and preclinical perfection of a number of techniques,
most notably quantitative polymerase chain reaction (PCR) and multicolor flow cytometry.
Late additions to the group of applicable assays include next generation sequencing and
digital PCR. In this review the principles of use of these modalities at three different time
points during the AML disease course are discussed, namely at the time of treatment
evaluation, pretransplantation and postconsolidation. The drawbacks and pitfalls of each
different technique are delineated. The evidence or lack of evidence for minimal residual
disease guided treatment decisions is discussed. Lastly, future strategies in the MRD field are
suggested and commented upon.

Keywords: Minimal residual disease, Preemotive treatment, AML, flow cytometry, qPCR, NGS

Introduction hematologist [Cornelissen et al. 2012; Dohner Correspondence to:


Hans Beier Ommen, MD,
Upon diagnosis, a patient with acute myeloid leu- etal. 2010]. PhD
kaemia (AML) faces the ordeal of his or her life. Department of
Hematology, Aarhus
Several courses of intensive chemotherapy, com- Aiding the hematologist in these decisions are a University Hospital, Tage-
plicating infections, and possibly allogeneic bone number of prognostic scoring systems, including Hansens gade 2, Aarhus C,
8000, Denmark
marrow transplantation awaits, and even then the leukaemia- and patient-specific data, that is, the hansomme@rm.dk
prognosis is no better than a 5-year survival of European Leukaemia Net (ELN) prognostic sys-
50% even in young adults [Burnett etal. 2013]. tem [Dohner et al. 2010] the revised Grimwade
For the hematologist aiding the patient through criteria [Grimwade etal. 2010], and the Wheatley
his or her disease course, evidence supports the score of the UK Medical Research Council
choice of front-line induction therapy but not (MRC) program [Wheatley etal. 1999], to name
much else. In most centers, for the few patients a few. To different degrees, these systems include
with an especially good prognosis [i.e. core the rapidly growing amount of data on the differ-
binding factor leukaemias (CBFs), RUNX1 ent molecular phenotypes of AML. More sophis-
RUNX1T1 and CBFBMYH11 positive AMLs] ticated systems will probably emerge in the
consolidation treatment does not include alloge- coming years as prognostic values of the multi-
neic bone marrow transplantation. The some- tudes of different genetic aberration combina-
what more common patients with an especially tions recently discovered can be included.
bad prognosis invariably are considered candi-
dates for transplantation, at least if comorbidities However, presently, these systems cannot foresee
and donor availability permits. Treatment besides the treatment response at the level of the individ-
this, such as the optimal consolidation treatment ual patient with AML. The sheer complexity of
for the large majority of intermediate risk patients both the genetic (and epigenetic) background of
or the correct treatment upon refractory disease the leukaemia as well as of the patient preclude
or relapse, still very much depends on the treating that we anytime soon will be able to predict this.

http://tah.sagepub.com 3
Therapeutic Advances in Hematology 7(1)

Thus, during and after treatment, patients are highest degree. These include when the leukemic
monitored to observe the treatment responses. In clone is disappearing, such as during treatment;
this discipline also, a rapidly expanding set of when it is imperative that the leukemic burden is
options is available. The choice between different low, such as before allogeneic treatment; or when
disease-monitoring techniques and the possibili- the leukaemia is reappearing after either chemo-
ties and caveats of these in the treatment and fol- therapy or transplantation consolidation.
low up of AML are the topics of this review. Acute
promyelocytic leukaemia presents a special case
that will not be covered by this review. Treatment efficacy evaluation
As the aim of cytoreductive treatment is to eradi-
cate the leukemic clone, it is natural to test the
Disease surveillance in the genomic age efficacy of this treatment by MRD detection tech-
Since 1961 [Freireich et al. 1961], the achieve- niques. In the early 2000s a large number of sin-
ment of the state where less than 5% of bone mar- gle-center studies confirmed the prognostic value
row blasts present by morphological examination of a low MRD measurement during treatment,
by light microscopy [morphological complete either in a selected cohort based on single molec-
remission (CR)] has been considered to be of key ular aberrations [Schnittger et al. 2003, 2007;
importance in the long-term prognostication of Stentoft etal. 2006] or using more broadly appli-
patients with AML. Since then, technical develop- cable MRD markers [Ommen etal. 2008; Cilloni
ment has produced a number of advanced tech- etal. 2008, 2009]. In the last 58 years the results
niques to further assess the presence of residual of a number of pivotal multicenter studies have
disease. In the 1980s and early 1990s, the term been published as well [Freeman et al. 2013;
minimal residual disease (MRD) was coined to Terwijn et al. 2013; Kronke et al. 2011]. While
distinguish residual disease as assessed morpho- some studies identify a threshold below which
logically from that detected with more sensitive relapse risk is low, most commonly 0.1% or 1%,
techniques in patients in CR [Campana and Pui, others were powered to show the linear relation-
1995]. Thus, any technique with a sensitivity ship between declining MRD levels and progno-
beyond that of (immunohistochemistry-assisted) sis. Also controversial is the optimal time point
light microscopy can in the traditional definition for MRD testing. While testing after the first
be used as an MRD detection technique. This course of chemotherapy is most convenient if
dichotomy in disease status as evaluable either by intensification of treatment is necessary, data
standard methods or as MRD is reflected in the from the HOVON group suggest that some
current consensus response criteria report from patients, in CR but MRD positive after the first
2003 [Cheson et al. 2003] in which molecular course of chemotherapy, actually achieve a suffi-
response is included as a provisional response cri- cient MRD decrease after the second course of
terion only [Cheson etal. 2003]. With the steadily chemotherapy, allowing the inclusion of these
growing body of evidence that MRD measure- patients in the MRD-based low-risk arm [Terwijn
ments have prognostic value in AML and with tri- etal. 2013]. Further studies will show if this ben-
als incorporating MRD measurements in decision efit makes up for the obvious risks of administer-
making slowly emerging, we advocate the merging ing an additional course of standard chemotherapy
of the concepts of morphological residual disease to the high-risk patients when prompt treatment
and MRD into the concept of measureable resid- intensification could be advocated.
ual disease to reflect the joining of the MRD con-
cept into standard disease surveillance practice
[Hokland et al. 2015]. Several studies (e.g. the Postchemotherapy early relapse detection
MRC AML17 and AML19 studies) are currently One of the most unsatisfying aspects of disease
investigating the beneficial role of MRD in disease surveillance based on standard methods is in the
surveillance in a large randomized manner to pro- follow up of patients after completion of chemo-
vide definitive evidence for this transition. therapy. Patients can be well, with normal hema-
tological counts, and 14 days later they can
The current use of MRD measurements reflects display signs of relapse with deteriorating bone
its auxiliary status compared with standard mor- marrow function. It is unsurprising that longitu-
phology. MRD measurements are most useful at dinal follow up of patients postchemotherapy
time points when the increased sensitivity com- with the prospect of early relapse detection was
pared with light microscopy is exploited to the an early venue of the MRD technology. Several

4 http://tah.sagepub.com
HB Ommen

points have been made in the last 15 years of test- by the transplanted immune system could change
ing and perfecting the techniques. the relapse pattern, rendering the relapses of two
molecularly similar leukaemias different in a trans-
First, evidence suggests that above a certain planted and a nontransplanted host.
threshold of MRD positivity, relapse is certain to
occur [Ommen etal. 2010; Yin etal. 2012]. This
condition is commonly referred to as molecular Methods
relapse. Second, surveillance can be done substi-
tuting bone marrow aspirations for blood sam- Polymerase chain reaction
ples, at least in some cases [Cilloni et al. 2008; The exponential nature of the polymerase chain
Yin et al. 2012; Ommen et al. 2010, 2014; reaction (PCR) techniques allows for very high
Abildgaard et al. 2013]. Third, diligence is sensitivities making these techniques ideal for
required in post-treatment sampling, as many MRD measurements, especially upon the intro-
relapses will occur too fast if patients are only duction of the quantitative PCR systems [Kubista
occasionally sampled [Ommen etal. 2008, 2010, etal. 2006]. Recently, development of microfluid-
2014, 2015; Weisser etal. 2005]. Taken together, ics-based systems, such as digital PCR, has
early relapse diagnostics is now possible, but evi- allowed for the development of assays with a sen-
dence is still scarce on how to act upon molecular sitivity that is even higher (up to 10 fold) than
relapse. that of the traditional Taqman probe (or similar)
based quantitative PCR (qPCR) systems
[Hindson et al. 2013]. Additionally, qPCR sys-
MRD in the allotransplant setting tems offer relatively easy standardization, and
The use of MRD measurements in the allotrans- standardized clinically tested assays [Kronke etal.
plant setting is a special case. Several studies 2011] exist for the commonest fusion transcripts
report the adverse effect of MRD positivity prior [Gabert etal. 2003], mutated genes and overex-
to receiving the transplant [Jacobsohn etal. 2009; pressed genes [Cilloni etal. 2009].
Walter et al. 2011; Rossi et al. 2015]. These
patients are invariably patients who have recently
been heavily treated with chemotherapy. Thus, Fusion transcripts
this is a group of patients with suboptimal In AMLs harboring chromosomal translocations
response to chemotherapy, similar to that noted such as t(8;21), inv(16), t(6;9), t(9;11) and oth-
in ordinary treatment response evaluation. In ers, the unique fusion transcripts arising from
contrast to treating these patients using chemo- these translocations (RUNX1RUNX1T1,
therapy alone [Terwijn et al. 2013], allogeneic CBFBMYH11, DEKNUP214, MLLMLLT3)
transplantation has the potential to save 1520% can be used as leukaemia-specific disease markers
of these patients [Terwijn etal. 2013]. Based on [Gabert et al. 2003; Ostergaard et al. 2004a;
this, MRD positivity will not preclude transplan- Tobal et al. 2004]. Unfortunately, only about
tation, but could in the future result in the imple- 30% of young adults and older adults with AML
mentation of additional measures reserved for harbor translocations [Grimwade and Freeman,
high-risk patients. 2014] and even the commonest fusion transcripts,
RUNX1RUNX1T1 and CBFBMYH11 are not
In the post-transplant period, the use of MRD seen in more than 5% of cases in population-
measurements is similar to that in patients who based surveys [Grimwade and Freeman, 2014].
have completed chemotherapy consolidation. A As such, a large panel of primer-probe sets is nec-
couple of important differences are important to essary to cover only a small fraction of patients
note. The cohort of higher-risk patients combined with AML, at least in adult AML. For childhood
with the delayed onset of the graft versus leukaemia AML, CBF leukaemias, but especially the MLL
(GvL) effect means that those patients who do (HUGO gene) translocations, are commoner,
relapse, relapse earlier than patients who have not allowing for a coverage of about 51% using fusion
received transplantation [Goswami et al. 2015]. transcripts alone [Grimwade and Freeman,
Also, the reconstitution of the donor immune sys- 2014]. Fusion-based MRD measurements are
tem in the new host and the instigation of the GvL among the most sensitive methods available. The
effect fundamentally change the conditions under sensitivity varies depending on the average num-
which the early relapse occurs. At least theoreti- ber of fusion transcripts per leukemic cell, but
cally, the enhanced immune surveillance exercised often reaches 1105 [Gabert etal. 2003].

http://tah.sagepub.com 5
Therapeutic Advances in Hematology 7(1)

Mutated genes Table 1. The use of overexpressed genes as MRD


Specific qPCR reaction can be constructed to markers.
create MRD measurement assays that have a Gene name Sensitivity Applicability
much broader applicability than that of fusion (%)
transcript based MRD assays, as all or virtually all
AMLs harbor some kind of genetic mutation. Wilms tumor gene 1:50* 56
1, WT1
However, only in some genes (e.g. NPM1, to
1:100$ 23
some extent DNMT3A) do mutational hotspots
exist to allow for the detection of the mutant allele 1:200$ 7
without having to design patient-specific primer Preferentially 1:50* 33
expressed antigen in
and probe sets [Gorello etal. 2006]. In most set-
melanoma, PRAME
tings, the additional human capital required to
1:100 26
produce patient-specific primer and probe sets is
Steinbach 7 gene set 1:30 74
not available. Thus, other commonly mutated
1:100 59
genes (e.g. RUNX1, TET2) that require the use of
Goswami 5 gene set 1:50* 87
patient-specific primer and probe sets as no muta-
tional hotspots exist are rarely used as MRD *Adult AML, compared with median normal expression
markers using qPCR-based techniques. [Goswami etal. 2015].
$Adult AML, compared with 95% upper normal limit

[Hokland etal. 2012].


Childhood AML, compared with 90% upper normal limit

Overexpressed genes [Steinbach etal. 2014].


Fusion transcripts and useful mutated genes do AML, acute myeloid leukaemia; MRD, minimal residual
disease.
not exist in all AMLs, especially not in childhood
AML in which the NPM1 mutation is rare
[Grimwade and Freeman, 2014]. Thus, much
effort has been put into developing additional that, even if overexpressed in up to 80% of
assays to cover these remaining patients. This patients [Ostergaard et al. 2004b], only offers
need is further increased by the fact that the com- 100-fold upregulation in 23% of adult patients
monest fusion transcripts, RUNX1RUNX1T1, [Hokland etal. 2012] and if this level of overex-
CBFBMYH11, as well as NPM1 all confer a pression is required, can only be used in that
favorable prognosis to patients, signifying that fraction of patients. In the postconsolidation
relapsed or resistant disease actually is rarer in follow-up situation, expression in healthy tissues
these patients. For patients who have the greatest presents a special problem. One can either estab-
need for disease surveillance, those at greatest risk lish a safe threshold above which expression is
of resistant disease or relapse, a far lower fraction not seen in healthy tissues [Ostergaard et al.
of patients can actually be followed by a fusion 2004; Cilloni and Saglio, 2004] or closely inter-
transcript or mutated gene based MRD marker. pret the changes in expression, when a dramatic
rise in transcript level will signify an impending
In the transformed homeostasis of the leukemic relapse [Kristensen et al. 2011]. Most com-
cell, several mRNA transcripts are expressed to a monly, the two methods are used in combina-
greater extent than in the healthy bone marrow tion [Ommen etal. 2008].
cell. In diagnosis samples from patients with
AML, one or more of these overexpressed genes With some effort and combination, good gene
will in the large majority of cases be several fold sets can be constructed to cover almost all leukae-
higher than in healthy bone marrow (Table 1). mias, allowing overexpressed genes to be used as
a universal MRD detection system [Goswami
The downside of using overexpressed genes as etal. 2015; Steinbach etal. 2006, 2014]. Another
MRD markers is that the level of expression of possibility is to use the best overexpressed genes
the marker in healthy hematopoiesis has to be (such as WT1) in cases when none of the other
taken into account as well. Thus, even for a PCR-based MRD detection methods (fusion
marker that is upregulated in virtually all cases of transcripts, mutated genes) are applicable to cre-
AML, the need for 50- or 100-fold upregulation ate an MRD detection system including all three
to obtain sufficient sensitivity results in a restric- types of MRD markers, possibly supplemented
tion in the number of patients who can be evalu- by flow cytometry when especially sensitive flow
ated. The classical example is the WT1 gene cytometric markers are present.

6 http://tah.sagepub.com
HB Ommen

The most commonly used genes as well as two presence of normal cells in the LAP. However, in
published sets of genes and the sensitivities that flow cytometry, the MRD analyst has a lot more
can be achieved using these are summarized in control of which cells are included in the com-
Table 1. parison to the malignant phenotype than in PCRs
when cells are lyzed in the nucleotide (DNA or
RNA) purification process. As immature cells are
Flow cytometry uncommon in healthy bone marrow, the addition
Flow cytometry becomes an obvious MRD meas- of an immature marker to the LAP usually results
uring tool based on the high-throughput nature of in normal cell pollution below 0.05%, effectively
the technique and the possibility for very exact eliminating the problem [Terwijn etal. 2013].
cell characterization offered by multicolor proto-
cols. The principle is to identify leukaemia-asso- In many cases surface antigen expression on the
ciated phenotypes (LAPs) when leukemic cells leukemic clone varies, probably depending on dif-
differ from the large majority of healthy hemat- ferent states of maturation of the individual clonal
opoiesis bone marrow or blood cells. The differ- cells. Thus, a given LAP will often not comprise
ing expression of different LAPs can be divided all leukemic cells. This raises the problem of
into four different types: cross-lineage expression, whether the cells selected actually have the poten-
when T-cell, B-cell or natural killer cell markers tial to give rise to a relapse. To amend this, the
are expressed on the myeloid blasts; overexpres- inclusion of stem cell markers in the LAPs has
sion, when normally expressed antigens are been proposed [Larsen etal. 2012; Van Rhenen
expressed to a higher degree on each individual etal. 2007]. This may hamper theoretical sensi-
cell; lack of expression, when normally expression tivity as stem cells in the leukemic clone are
antigens are lacking; or finally asynchronous uncommon, but has been shown to perform as
expression, when immature and mature myeloid well as MRD assays based on standard LAPs or
antigens are expressed together in an aberrant WT1 overexpression [Roug etal. 2013].
manner [Kern etal. 2010]. Finding a LAP to all
AMLs requires an extensive panel of antibodies
[Kern etal. 2010], but 8090% of patients can be Next generation sequencing
followed using a reasonably sized panel [Freeman Next generation sequencing (NGS) offers the
etal. 2013; Terwijn etal. 2013]. The most com- opportunity for detection and follow up of a large
mon antigens included in LAPs and the frequen- number of aberrations. This allows not only clas-
cies of their possible use are shown in Table 2. sic MRD follow up but also the inclusion of
potentially important changes on the subclonal
level. As virtually all AMLs harbor genetic muta-
LAP selection: immature and stem cell tions and mutational hotspots are not necessary
markers for NGS-based MRD measurements, NGS could
Flow cytometric based MRD detection uses the potentially be applicable in all AMLs. There is a
same principles as overexpressed gene MRD caveat, however. The current base error rate
detection and the sensitivity is restricted by the means that the sensitivity level is about 1%

Table 2. Antigen basis for LAPs in AML. Applicability and detection thresholds in table from Freeman etal. [2013].

LAP type Main aberrant antigens Detection threshold* Applicability (%)


Cross lineage CD7 0.020.1% 22
CD56 0.020.1% 24
CD19 0.020.1% <5$
Asynchronous CD13/CD33 0.1% 27
CD38/CD33 0.1% 14
Overexpression CD34 0.1% 15
Lack of expression HLA-DR 0.020.1% 30
*LAP only used if present in over 10% of blasts. As at least 20% of blasts are present at diagnosis, a detection threshold
of 0.1% compares to a sensitivity of at least 1:20, but often more than 1:100.
$Often associated with presence of RUNX1-RUNX1T1 [Gustafson etal. 2009].

AML, acute myeloid leukaemia; LAP, leukaemia-associated phenotype.

http://tah.sagepub.com 7
Therapeutic Advances in Hematology 7(1)

[Kohlmann etal. 2014; Luthra etal. 2014], which this will increase the need for broader antibody
cannot compete with any of the above-mentioned panels and the cost of the analysis. Stem cell
MRD measurements techniques, save perhaps marker based MRD follow up has been reported
the worst overexpressed gene-based assays. to be stable at relapse [Roug et al. 2013] and
Technical development is currently trying to cir- another possibility is to include some of these
cumvent this [Hokland et al. 2015] and when markers in the LAPs.
these endeavors are successful, NGS-based MRD
measurements could be a very powerful tool
indeed. Detection of premalignant clones
The opposite of the risk of using mutations not
present in the relapsing subclone as MRD marker
Pitfalls in MRD assessment is using a marker that is present in the premalig-
nant lesion of the patient.
Phenotype shifts
The dominating malignant clone at diagnosis is Thus, Ploen and colleagues found mutated
often genetically unstable and some genetic evo- DNMT3A to be present in the leukemic cells of a
lution will invariantly occur during disease pro- patient with AML, but also to be continually pre-
gression. Furthermore, subclone selection by sent even after eradication of the leukemic clone
treatment is unavoidable, allowing clones that [Ploen et al. 2014]. The DNMT3A mutation in
are only present in small proportions at diagnosis the described case is probably a marker of mono-
to dominate at relapse. The frequency of pheno- clonal hematopoiesis of unknown significance, as
type shifts differs substantially between different recently described [Jaiswal etal. 2014; Genovese
MRD targets. It is virtually unknown for fusion etal. 2014]. DNMT3A is a commonly seen muta-
transcript and NPM1 based MRD detection to tion in this context, as is TET2. Diligence is
display phenotype shifts, whereas other muta- advised if these MRD targets are used as MRD
tions such as FLT3-ITD and WT1 mutation dis- markers to ensure that recurring MRD positivity
appears or is added at relapse in up to 20% of actually represents recurrence of the leukemic
cases [Nyvold et al. 2006; Bachas et al. 2010]. clone.
The recent paper by Lindsey and colleagues
offers an explanation for this [Lindsey et al.
2015]. The authors divide AML mutations into Post-treatment positivity
MDS related (e.g. ASXL1, SRSF2), pan-AML Another observation is that when testing
(e.g. RUNX1, CEBPA, TET2, WT1, FLT3) and patients postconsolidation not all MRD-
de novo AML related (e.g. NPM1, CBF leukae- positive patients relapse [Yin et al. 2012;
mia translocations, MLL translocation). De novo Ommen etal. 2010, 2014, 2015]. There seems
AMLs contain relatively few mutations and these to be a certain threshold below which relapse is
mark the leukemogenic events in these patients, not certain to occur, both for fusion transcript
whereas pan-AML mutations can occur both as based [Yin et al. 2012; Ommen et al. 2010,
leukomogenic and secondary events, making 2014, 2015], mutation based [Kronke et al.
these markers more prone to loss because of 2011] and flow cytometry based MRD detec-
alternative clone outgrowth. This is not to say tion [Terwijn et al. 2013]. This phenomenon
that a mutation classified by Lindsey and col- could have several possible explanations. First,
leagues as pan-AML cannot be a very useful the PCR-based positivity could be simply due
MRD marker, merely to emphasize the power of to detection of more mature cells not capable of
this model to explain the findings that phenotype producing a relapse. If so, stem cell based flow
shifts are more common for some mutations than cytometric follow up should not display this
for others. feature. Further research will show whether this
is the case. Second, the low level of positivity
As immunophenotypic markers are not part of could represent a low level of residual leukae-
the malignant process but rather a product of the mia held in check by the immune system. The
disturbed cell homeostasis, these markers are observation that the threshold for relapse is
among those most commonly lost [Feller et al. higher in transplanted patients [Ommen et al.
2004; Voskova etal. 2004]. This can be amended 2014] could support this notion, but further
following patients using several markers to lessen research is needed before any firm conclusions
the risk of them all disappearing at once. However, can be drawn on this point.

8 http://tah.sagepub.com
HB Ommen

Status on the current evidence for the use of completed their assigned treatment (see Table 3)
MRD assessments in the clinic [Zhu et al. 2013]. Naturally, a study like this is
How can MRD diagnostics improve the manage- prone to selection bias, especially in the arm
ment of patients with AML in morphological CR? where patients did not receive transplantation
Consider a patient at high risk based on pretreat- despite their risk status recommending this treat-
ment factors, patient specific or leukaemia spe- ment. The authors showed that the groups were
cific. If this patient experiences a very good comparable based on age, sex and comorbidities,
molecular response to induction therapy should but as always, in an unrandomized study,
consolidation be deescalated and transplantation unknown biases are not accounted for.
avoided? Or is it in such patients that transplanta-
tion does actually have a chance to provide a cure? Currently the UK MRC AML18 trial is recruiting.
This trial contains true randomization between
Consider the patient consolidated with chemo- two different kinds of intensified treatment in
therapy experiencing a molecular relapse. Should patients who are in the MRD high-risk group and
preemptive treatment be started prior to frank has the potential to further research on MRD-
relapse? Or should this life-threatening therapy be directed treatment intensification significantly.
reserved for frank relapse?

A number of possible courses of action can be Relapse kinetics


envisioned based on MRD measurements. These In the evaluation of MRD measurements in
are summarized in Table 3. Each of these options patients who no longer receive chemotherapy, the
will have to be tested in a randomized manner, time factor in the possible reappearance of AML
and few of these studies have been published to becomes a concern. In the case of a negative MRD
date. Some evidence does exist, however, and this measurement this indicates the patient is free of
is described in the following section. relapse in the near future, but since different AML
subtypes relapse at very different relapse speeds
[Yin etal. 2012; Ommen etal. 2010, 2014, 2015],
Treatment intensification based on MRD this time period varies significantly for different
detection AML subtypes. In Figure 1, the median times
Rubnitz and colleagues performed a pivotal study from molecular to clinical relapse can be seen for
in childhood AML in which risk stratification those subtypes for which data are published. In
based on MRD analysis after the first course of the case of a molecular relapse, the relapse kinetics
chemotherapy determined treatment intensifica- data can aid in the decision on how often to con-
tion in the form of an early second course of trol the relapsing disease if no preemptive treat-
chemotherapy with the possible addition of gem- ment is chosen. If sufficient evidence to support
tuzumab ozogamicin. Furthermore, consolidation preemptive treatment is produced in the future, it
was determined based both on MRD measure- is conceivable that relapse kinetics data could be
ment and pretreatment risk factors. As is common helpful in the choice between different treatment
in childhood AML, the study was not randomized modalities (Figure 1).
between action upon MRD-directed treatment or
not, but the authors concluded that the long-term
survival rate is superior to most contemporary Preemptive treatment
protocols, assigning this success to the MRD- Several smaller studies (see Table 3) have tested
directed therapy design [Rubnitz etal. 2010]. the value of preemptive treatment in patients
experiencing a molecular relapse. In 2009,
In adult AML, Zhu and colleagues performed a Doubek and colleagues published their experi-
study in patients with CBF-positive AML. ences treating 21 molecular relapses of RUNX1
Whether patients received transplantation or not RUNX1T1, CBFBMYH11 or MLL gene
was based on achievement of MRD at the level of translocated AML with either gemtuzumab ozo-
1103. In both the high-risk and low-risk gamicin, 5+2 like chemotherapy or, in the case
groups, some patients opted not to follow the of patients who had undergone allogeneic trans-
treatment decision based on the MRD determi- plantation, prompt discontinuation of immuno-
nations and the authors proved that the patients suppression or donor lymphocyte infusion (DLI)
receiving treatment other than that assigned by (see Table 3) [Doubek et al. 2009]. Responses
the risk status did worse than those who were seen to all treatment modalities, but the

http://tah.sagepub.com 9
Therapeutic Advances in Hematology 7(1)

Figure 1. Relapse kinetics of different AML subtypes and relation to treatment onset.
Relapse kinetics from Ommen and colleagues (several papers) [Ommen etal. 2010, 2014, 2015] and Yin etal. [2012]. Time to
treatment onset for azacitidine from Silverman etal. [2011]. Time to treatment onset for chemotherapy and GvL effect based
on common clinical observations (disappearance of blasts after first course of chemotherapy, full chimerism often 3 months
post transplantation). AML, acute myeloid leukaemia; GvL, graft versus leukaemia.

treatments were too heterogeneous and the Another possibility is a demethylating agent, even
patient cohort too small to draw any firm conclu- if this option is only attractive in patients in whom
sion. Patients who re-entered molecular CR sufficient time is available from molecular to clini-
invariably relapsed, but the duration of response cal relapse for the treatment effect of these agents
was several hundred days in some cases. to commence (see Figure 1). Sockel and col-
leagues administered azacitidine upon molecular
In the venue of post-transplanted patients, Pozzi relapse of patients with NPM1 mutation. In 7 of
and colleagues were able to administer DLI in 10 patients, the authors observed a response,
17/38 patients with molecular relapse [Pozzi etal. either as renewed molecular remission or as a
2013]. The survival was superior in the DLI delayed time to clinical relapse [Sockel et al.
group but the study was not truly randomized; 2011]. As two-thirds of complete responses were
the reason for not administering DLI included seen in previously transplanted patients, the group
random factors such as donor unavailability but initiated the RELAZA trial in which transplanted
also treatment-related factors such as type of patients with molecular relapse were treated with
transplant (cord blood transplants could not azacitidine. Relapse was avoided in 4 of 20 patients
receive DLI). but was delayed a median of 230 days in patients
with relapsing disease [Platzbecker et al. 2012].
In another study of preemptive treatment in Thus, azacitidine seems promising; especially in
CBF-positive AML, seven of eight patients with the allogeneic transplant setting, but can probably
relapsing disease treated with dasatinib upon only cure a minority of patients on its own. To fol-
molecular relapse progressed quickly to clinical low up on this observation, the NOPHO group of
relapse at a speed (median time from molecular pediatric oncologists are currently starting a study
relapse to clinical relapse, 60 days) indistinguish- to test whether azacitidine can be useful as a bridg-
able from published median times from molecu- ing agent between a molecular relapse and an allo-
lar to clinical relapse [Yin et al. 2012; Ommen geneic transplantation.
etal. 2010; Boissel etal. 2011].
Finally, in the UK MRC AML17 and AML19
In conclusion, neither reduced dose chemother- studies, patients are randomized if they have a use-
apy nor tyrosine kinase inhibitor (TKI) seems an ful MRD marker to either MRD follow up or not.
attractive choice for preemptive treatment. The endpoint is overall survival. If successful, this

10 http://tah.sagepub.com
HB Ommen

Table 3. Possible MRD-directed actions.

Response evaluation Trial Result p


Treatment intensification, MRC AML 18 Pending
chemotherapy
Treatment intensification, Zhu etal. [2013] OS 72% in transplanted high-risk 0.007
allogeneic transplant patients
OS 27% in nontransplanted high-risk
patients
Treatment de-escalation Zhu etal. [2013] OS 76% in transplanted low-risk 0.013
patients
OS 100% in nontransplanted low-risk
patients
Molecular relapse Trial Result
Wait and watch AML17+ Pending
AML19
Initiation of donor search AML17+ Pending
AML19
Demethylating agent Sockel etal. [2011] 7/10 response
3/10 reentered CR
Platzbecker etal. [2012] 10/20 reentered CR
4/20 long term survivors
Leukaemia aberration Boissel etal. [2011] 1/8 long-term survivors
specific treatment
Discontinuation of Doubek etal. [2009] 1/3 long-term survivor
immunosuppression
Donor lymphocyte infusion Doubek etal. [2009] 3/3 response
1/3 long-term survivor
Pozzi etal. [2013] OS 44% 17 DLI-treated patients versus 0.004
14% 21 non-DLI-treated patients
Reduced dose standard Doubek etal. [2009] 3/15 response
chemotherapy No long-term survivors
Standard chemotherapy AML17+ Pending
AML19
Direct allogeneic none
transplantation
AML, acute myeloid leukaemia; CR, complete response; MRC, Medical Research Council; MRD, minimal residual dis-
ease; OS, overall survival.

study will probably significantly alter the way we Terwijn et al. 2013], but the use of RUNX1
treat molecular relapse as preemptive treatment RUNX1T1 and CBFBMYH11 [Yin etal. 2012]
will have to be seriously considered in these cases. and NPM1 [Kronke et al. 2011] has been vali-
dated as well. At treatment evaluation, phenotype
shifts are only a lesser concern as selection and
Future directions in the evaluation of MRD outgrowth of a secondary clone has not yet hap-
diagnostics pened. Thus, the expensive procedures of using
multiple LAPs requiring both expenditure of
Comparison of flow cytometry versus PCR multiple antibodies and a significant amount of
versus NGS human capital become less important. The higher
The different methods of MRD detection offer sensitivity of fusion transcript or mutated gene
different advantages and have different disadvan- based PCR assays is not necessarily useful in the
tages. In the case of the choice of method in treat- treatment assessment situation as some residual
ment evaluation, two large trials validated the disease is probably acceptable, at least after the
value of flow cytometry [Freeman et al. 2013; first course of chemotherapy [Terwijn etal. 2013].

http://tah.sagepub.com 11
Therapeutic Advances in Hematology 7(1)

In the patients without useful fusion transcripts or could be proposed, but further standardization
mutated genes, several works show that one over- and testing is awaited before implementation.
expressed gene or a combination of these can be
highly prognostically useful in treatment evalua-
tion. Few reports have diligently tested the use of The inclusion of relapse kinetics
flow cytometry in the post-chemotherapy follow When evaluating preemptive treatment options,
up and problems with phenotype shift are espe- the nature of the relapsing AML should be kept in
cially prevalent in this setting. However, at least mind. In the majority of published results for
with the current evidence, LAPs including stem patients treated with preemptive treatment,
cell markers do not seem marred by a great num- relapse was not avoided but delayed (Table 3). In
ber of LAP losses and the problem with low-level some patients not fit for intensive reinduction or
positivity should be reduced using this method, transplantation, or without a suitable donor,
even if evidence does not yet exist. delaying relapse by several months could be a
useful alternative to no treatment.
Flow cytometry and PCR measure two different
things, namely malignant cells and mRNA expres- It is very probable that all molecular relapses
sion in the malignant cells. Using immature or should not be treated in the same way. Slower
stem cell markers, flow cytometry has the poten- relapsing subtypes could be eligible for azaciti-
tial to differentiate between cells of the malignant dine or direct allogeneic transplantation. Others
clone with or without relapse potential. However, will have to be reinduced using intensive chemo-
PCR-based MRD measurement includes infor- therapy or experimental treatment by therapies
mation on cell homeostasis as metabolically active directed at the molecular aberrations in the
cells containing larger amounts of the targeted relapsing leukemic cells since not enough time is
RNA sequences are more easily detected. It is available for the initiation of the effect of the
impossible to theoretically predict which of these slower-acting treatment strategies.
two approaches discriminates best between risk
groups. Future studies will show if one type of
MRD measurement or the other is best at the dif- The perfect as the enemy of the good
ferent MRD measuring situations, but currently A large amount of literature exists to support the
flow cytometry and the PCR-based method are prognostic value of MRD measurements at several
probably equally useful at the treatment evalua- different locations. Current MRD detection tech-
tion time point whereas PCR is probably superior niques are highly sophisticated, allowing detection
in post-chemotherapy follow up. of minute amounts of residual AML cells. Our
current technologies are probably highly sufficient;
it is how we act upon a high MRD measurement
Maintenance therapy in AML that is uncertain. Evolving technologies such as
In patients not fit for transplantation, the use of NGS, digital PCR or other microfluidics-based
MRD measurements could provide the basis for assays or even cell-free DNA-based assays will
intensification of therapy in the form of addition probably refine our MRD measurements further,
of maintenance therapy, currently mainly used in but in the enthusiasm of the current molecular
the acute lymphoblastic leukaemia (ALL) setting. revolution we should not forget our well tested
Also, in frailer patients maintenance therapy could assays for which the possible time to clinical imple-
possibly replace intensive chemotherapy consoli- mentation is shorter. If we use our current under-
dation. Evidence for the use of MRD to direct this standing of MRD and if we get evidence as to how
is currently sparse, but in patients achieving CR to act upon MRD measurements, current tech-
but who are not fit for additional intensive treat- nologies are fully adequate to help our patients.
ment, this could definitely be an option.
Funding
The drugs to use in this setting remain to be deter- This research received no specific grant from any
mined, but both demethylating agents and tyros- funding agency in the public, commercial, or not-
ine kinase inhibitors with activity in AML could for-profit sectors.
be likely candidates. In the case of the use of dem-
ethylating agents, follow up by MRD detection Conflict of interest statement
methods targeting methylations rather than DNA The author declares that there is no conflict of
changes, gene overexpression or flow cytometry interest.

12 http://tah.sagepub.com
HB Ommen

References The European LeukemiaNet AML Working Party


Abildgaard, L., Ommen, H., Lausen, B., Hasle, H. consensus statement on allogeneic HSCT for patients
and Nyvold, C. (2013) A novel RT-qPCR assay for with AML in remission: an integrated-risk adapted
quantification of the MLL-MLLT3 fusion transcript approach. Nat Rev Clin Oncol 9: 579590.
in acute myeloid leukaemia. Eur J Haematol 91:
Dohner, H., Estey, E., Amadori, S., Appelbaum, F.,
394398.
Buchner, T., Burnett, A. etal. (2010) Diagnosis and
Bachas, C., Schuurhuis, G., Hollink, I., Kwidama, Z., management of acute myeloid leukemia in adults:
Goemans, B., Zwaan, C. etal. (2010) High-frequency recommendations from an international expert panel,
type I/II mutational shifts between diagnosis and on behalf of the European LeukemiaNet. Blood 115:
relapse are associated with outcome in pediatric AML: 453474.
implications for personalized medicine. Blood 116:
27522758. Doubek, M., Palasek, I., Pospisil, Z., Borsky, M.,
Klabusay, M., Brychtova, Y. etal. (2009) Detection
Boissel, N., Jourdan, E., Pigneux, A., Blanchet, O., and treatment of molecular relapse in acute myeloid
Renneville, A., Recher, C. et al. (2011) Single-agent leukemia with RUNX1 (AML1), CBFB, or MLL
dasatinib does not prevent hematological relapse in gene translocations: frequent quantitative monitoring
patients with core binding factor (CBF) acute myeloid of molecular markers in different compartments and
leukemia (AML) in first complete remission, but correlation with WT1 gene expression. Exp Hematol
persistent or re-appearing molecular minimal residual 37: 659672.
disease-results of the DASA-CBF trial from the
French AML intergroup. Blood 118: 1119. Feller, N., van der Pol, M., van Stijn, A., Weijers, G.,
Westra, A., Evertse, B. etal. (2004) MRD parameters
Burnett, A., Russell, N., Hills, R., Hunter, A., using immunophenotypic detection methods are
Kjeldsen, L., Yin, J. etal. (2013) Optimization of highly reliable in predicting survival in acute myeloid
chemotherapy for younger patients with acute myeloid leukaemia. Leukemia 18: 13801390.
leukemia: results of the Medical Research Council
AML15 trial. J Clin Oncol 31: 33603368. Freeman, S., Virgo, P., Couzens, S., Grimwade,
D., Russell, N., Hills, R. etal. (2013) Prognostic
Campana, D. and Pui, C. (1995) Detection
relevance of treatment response measured by flow
of minimal residual disease in acute leukemia:
cytometric residual disease detection in older patients
methodologic advances and clinical significance. Blood
with acute myeloid leukemia. J Clin Oncol 31:
85: 14161434.
41234131.
Cheson, B., Bennett, J., Kopecky, K., Buchner,
Freireich, E., Gehan, E., Sulman, D., Boggs, D.
T., Willman, C., Estey, E. etal. (2003) Revised
and Frei, E., 3rd (1961) The effect of chemotherapy
recommendations of the International Working Group
on acute leukemia in the human. J Chronic Dis 14:
for Diagnosis, Standardization of Response Criteria,
593608.
Treatment Outcomes, and Reporting Standards for
Therapeutic Trials in Acute Myeloid Leukemia. J Clin Gabert, J., Beillard, E., van der Velden, V., Bi,
Oncol 21: 46424649. W., Grimwade, D., Pallisgaard, N. etal. (2003)
Cilloni, D., Messa, F., Arruga, F., Defilippi, I., Standardization and quality control studies of real-
Gottardi, E., Fava, M. etal. (2008) Early prediction time quantitative reverse transcriptase polymerase
of treatment outcome in acute myeloid leukemia by chain reaction of fusion gene transcripts for residual
measurement of WT1 transcript levels in peripheral disease detection in leukemia a Europe Against
blood samples collected after chemotherapy. Cancer program. Leukemia 17: 23182357.
Haematologica 93: 921924. Genovese, G., Kahler, A., Handsaker, R., Lindberg,
Cilloni, D., Renneville, A., Hermitte, F., Hills, J., Rose, S., Bakhoum, S. etal. (2014) Clonal
R., Daly, S., Jovanovic, J. etal. (2009) Real-time hematopoiesis and blood-cancer risk inferred from
quantitative polymerase chain reaction detection blood DNA sequence. N Engl J Med 371: 24772487.
of minimal residual disease by standardized WT1
Gorello, P., Cazzaniga, G., Alberti, F., DellOro, M.,
assay to enhance risk stratification in acute myeloid
Gottardi, E., Specchia, G. etal. (2006) Quantitative
leukemia: a European LeukemiaNet study. J Clin
assessment of minimal residual disease in acute
Oncol 27: 51955201.
myeloid leukemia carrying nucleophosmin (NPM1)
Cilloni, D. and Saglio, G. (2004) WT1 as a universal gene mutations. Leukemia 20: 11031108.
marker for minimal residual disease detection
Goswami, M., McGowan, K., Lu, K., Jain, N.,
and quantification in myeloid leukemias and in
Candia, J., Hensel, N. etal. (2015) A multigene array
myelodysplastic syndrome. Acta Haematol 112: 7984.
for measurable residual disease detection in AML
Cornelissen, J., Gratwohl, A., Schlenk, R., Sierra, patients undergoing SCT. Bone Marrow Transplant 50:
J., Bornhauser, M., Juliusson, G. etal. (2012) 642651.

http://tah.sagepub.com 13
Therapeutic Advances in Hematology 7(1)

Grimwade, D. and Freeman, S. (2014) Defining acute myeloid leukaemia patients with increased
minimal residual disease in acute myeloid leukemia: sensitivity compared to WT1 expression. Eur J
which platforms are ready for Prime Time? Blood Haematol 87: 400408.
124: 33453355.
Kronke, J., Schlenk, R., Jensen, K., Tschurtz, F.,
Grimwade, D., Hills, R., Moorman, A., Walker, H., Corbacioglu, A., Gaidzik, V. etal. (2011) Monitoring
Chatters, S., Goldstone, A. etal. (2010) Refinement of minimal residual disease in NPM1-mutated acute
of cytogenetic classification in acute myeloid myeloid leukemia: a study from the German-Austrian
leukemia: determination of prognostic significance acute myeloid leukemia study group. J Clin Oncol 29:
of rare recurring chromosomal abnormalities among 27092716.
5876 younger adult patients treated in the United
Kubista, M., Andrade, J., Bengtsson, M., Forootan,
Kingdom Medical Research Council trials. Blood 116:
A., Jonak, J., Lind, K. etal. (2006) The real-time
354365.
polymerase chain reaction. Mol Aspects Med 27:
Gustafson, S., Lin, P., Chen, S., Chen, L., Abruzzo, 95125.
L., Luthra, R. etal. (2009) Therapy-related acute
Larsen, H., Roug, A., Just, T., Brown, G. and
myeloid leukemia with t(8;21) (q22;q22) shares
Hokland, P. (2012) Expression of the hMICL in acute
many features with de novo acute myeloid leukemia
myeloid leukemia a highly reliable disease marker at
with t(8;21)(q22;q22) but does not have a favorable
diagnosis and during follow-up. Cytometry
outcome. Am J Clin Pathol 131: 647655.
B Clin Cytom 82: 38.
Hindson, C., Chevillet, J., Briggs, H., Gallichotte,
Lindsley, R., Mar, B., Mazzola, E., Grauman, P.,
E., Ruf, I., Hindson, B. etal. (2013) Absolute
Shareef, S., Allen, S. etal. (2015) Acute myeloid
quantification by droplet digital PCR versus analog
leukemia ontogeny is defined by distinct somatic
real-time PCR. Nat Methods 10: 10031005.
mutations. Blood 125: 13671376.
Hokland, P., Ommen, H., Mule, M. and Hourigan,
Luthra, R., Patel, K., Reddy, N., Haghshenas, V.,
C. (2015) Advancing the minimal residual disease
Routbort, M., Harmon, M. etal. (2014) Next-
concept in acute myeloid leukemia. Semin Hematol 52:
generation sequencing-based multigene mutational
184192. doi:10.1053/j.seminhematol.2015.04.001
screening for acute myeloid leukemia using MiSeq:
Hokland, P., Ommen, H., Nyvold, C. and Roug, A. applicability for diagnostics and disease monitoring.
(2012) Sensitivity of minimal residual disease in acute Haematologica 99: 465473.
myeloid leukaemia in first remissionmethodologies in
Nyvold, C., Stentoft, J., Braendstrup, K., Melsvik,
relation to their clinical situation. Br J Haematol 158:
D., Moestrup, S., Juhl-Christensen, C. etal. (2006)
569580.
Wilms tumor 1 mutation accumulated during therapy
Jacobsohn, D., Tse, W., Chaleff, S., Rademaker, A., in acute myeloid leukemia: biological and clinical
Duerst, R., Olszewski, M. etal. (2009) High WT1 implications. Leukemia 20: 20512054.
gene expression before haematopoietic stem cell
Ommen, H., Hokland, P., Haferlach, T., Abildgaard,
transplant in children with acute myeloid leukaemia
L., Alpermann, T., Haferlach, C. etal. (2014) Relapse
predicts poor event-free survival. Br J Haematol 146:
kinetics in acute myeloid leukaemias with MLL
669674.
translocations or partial tandem duplications within
Jaiswal, S., Fontanillas, P., Flannick, J., Manning, A., the MLL gene. Br J Haematol 165: 618628.
Grauman, P., Mar, B. etal. (2014) Age-related clonal
Ommen, H., Nyvold, C., Braendstrup, K., Andersen,
hematopoiesis associated with adverse outcomes.
B., Ommen, I., Hasle, H. etal. (2008) Relapse
N Engl J Med 371: 24882498.
prediction in acute myeloid leukaemia patients in
Kern, W., Bacher, U., Haferlach, C., Schnittger, S. complete remission using WT1 as a molecular marker:
and Haferlach, T. (2010) The role of multiparameter development of a mathematical model to predict time
flow cytometry for disease monitoring in AML. Best from molecular to clinical relapse and define optimal
Pract Res Clin Haematol 23: 379390. sampling intervals. Br J Haematol 141: 782791.
Kohlmann, A., Nadarajah, N., Alpermann, T., Ommen, H., Schnittger, S., Jovanovic, J., Ommen,
Grossmann, V., Schindela, S., Dicker, F. etal. (2014) I., Hasle, H., Ostergaard, M. etal. (2010) Strikingly
Monitoring of residual disease by next-generation different molecular relapse kinetics in NPM1c, PML-
deep-sequencing of RUNX1 mutations can identify RARA, RUNX1-RUNX1T1, and CBFB-MYH11
acute myeloid leukemia patients with resistant disease. acute myeloid leukemias. Blood 115: 198205.
Leukemia 28: 129137.
Ommen, H., Touzart, A., MacIntyre, E., Kern, W.,
Kristensen, T., Moller, M., Friis, L., Bergmann, O. Haferlach, T., Haferlach, C. etal. (2015) The kinetics
and Preiss, B. (2011) NPM1 mutation is a stable of relapse in DEK-NUP214-positive acute myeloid
marker for minimal residual disease monitoring in leukemia patients. Eur J Haematol 95: 436441.

14 http://tah.sagepub.com
HB Ommen

Ostergaard, M., Stentoft, J. and Hokland, P. based on quantification of fusion transcripts. Blood
(2004a) A real-time quantitative RT-PCR assay for 102: 27462755.
monitoring DEK-CAN fusion transcripts arising from
Silverman, L., Fenaux, P., Mufti, G., Santini, V.,
translocation t(6;9) in acute myeloid leukemia. Leuk
Hellstrom-Lindberg, E., Gattermann, N. etal. (2011)
Res 28: 12131215.
Continued azacitidine therapy beyond time of first
Ostergaard, M., Olesen, L., Hasle, H., Kjeldsen, response improves quality of response in patients with
E. and Hokland, P. (2004b) WT1 gene expression: higher-risk myelodysplastic syndromes. Cancer 117:
an excellent tool for monitoring minimal residual 26972702.
disease in 70% of acute myeloid leukaemia patients
Sockel, K., Wermke, M., Radke, J., Kiani, A.,
results from a single-centre study. Br J Haematol 125:
Schaich, M., Bornhauser, M. etal. (2011) Minimal
590600.
residual disease-directed preemptive treatment
Platzbecker, U., Wermke, M., Radke, J., Oelschlaegel, with azacitidine in patients with NPM1-mutant
U., Seltmann, F., Kiani, A. etal. (2012) Azacitidine acute myeloid leukemia and molecular relapse.
for treatment of imminent relapse in MDS or Haematologica 96: 15681570.
AML patients after allogeneic HSCT: results of the
Steinbach, D., Bader, P., Willasch, A., Bartholomae,
RELAZA trial. Leukemia 26: 381389.
S., Debatin, K., Zimmermann, M. etal. (2014)
Ploen, G., Nederby, L., Guldberg, P., Hansen, M., Prospective validation of a new method of monitoring
Ebbesen, L., Jensen, U. etal. (2014) Persistence of minimal residual disease in childhood acute myeloid
DNMT3A mutations at long-term remission in adult leukemia. Clin Cancer Res 21: 13531359.
patients with AML. Br J Haematol 167: 478486.
Steinbach, D., Schramm, A., Eggert, A., Onda, M.,
doi:10.1111/bjh.13062
Dawczynski, K., Rump, A. etal. (2006) Identification
Pozzi, S., Geroldi, S., Tedone, E., Luchetti, S., of a set of seven genes for the monitoring of minimal
Grasso, R., Colombo, N. etal. (2013) Leukaemia residual disease in pediatric acute myeloid leukemia.
relapse after allogeneic transplants for acute myeloid Clin Cancer Res 12: 24342441.
leukaemia: predictive role of WT1 expression. Br J
Stentoft, J., Hokland, P., Ostergaard, M., Hasle, H.
Haematol 160: 503509.
and Nyvold, C. (2006) Minimal residual core binding
Rossi, G., Carella, A., Minervini, M., di Nardo, F., factor AMLs by real time quantitative PCRinitial
Waure, C., Greco, M. etal. (2015) Optimal time- response to chemotherapy predicts event free survival
points for minimal residual disease monitoring change and close monitoring of peripheral blood unravels the
on the basis of the method used in patients with kinetics of relapse. Leuk Res 30: 389395.
acute myeloid leukemia who underwent allogeneic
Terwijn, M., van Putten, W., Kelder, A., van der
stem cell transplantation: a comparison between
Velden, V., Brooimans, R., Pabst, T. etal. (2013)
multiparameter flow cytometry and Wilms tumor 1
High prognostic impact of flow cytometric minimal
expression. Leuk Res 39: 138143.
residual disease detection in acute myeloid leukemia:
Roug, A., Larsen, H., Nederby, L., Just, T., Brown, data from the HOVON/SAKK AML 42A study.
G., Nyvold, C. etal. (2013) hMICL and CD123 in J Clin Oncol 31: 38893897.
combination with a CD45/CD34/CD117 backbone
Tobal, K., Frost, L. and Liu Yin, J. (2004)
- a universal marker combination for the detection of
Quantification of DEK-CAN fusion transcript by
minimal residual disease in acute myeloid leukaemia.
real-time reverse transcription polymerase reaction
Br J Haematol 164: 212222.
in patients with t(6;9) acute myeloid leukemia.
Rubnitz, J., Inaba, H., Dahl, G., Ribeiro, R., Haematologica 89: 12671269.
Bowman, W., Taub, J. etal. (2010) Minimal residual
Van Rhenen, A., Moshaver, B., Kelder, A., Feller, N.,
disease-directed therapy for childhood acute myeloid
Nieuwint, A., Zweegman, S. etal. (2007) Aberrant
leukaemia: results of the AML02 multicentre trial.
marker expression patterns on the CD34+CD38-
Lancet Oncol 11: 543552.
stem cell compartment in acute myeloid leukemia
Schnittger, S., Kern, W., Schoch, C. and Haferlach, allows to distinguish the malignant from the normal
T. (2007) RT-PCR-based MRD detection in stem cell compartment both at diagnosis and in
NPM1 mutated AML: a prospective follow-up remission. Leukemia 21: 17001707.
study in 130 patients. 49th Annual Meeting of the
Voskova, D., Schoch, C., Schnittger, S., Hiddemann,
American Society of Hematology, 811 December
W., Haferlach, T. and Kern, W. (2004) Stability of
2007. Blood 16: abstract 545.
leukemia-associated aberrant immunophenotypes
Schnittger, S., Weisser, M., Schoch, C., Hiddemann, in patients with acute myeloid leukemia
W., Haferlach, T. and Kern, W. (2003) New score between diagnosis and relapse: comparison with
predicting for prognosis in PML-RARA+, AML1- cytomorphologic, cytogenetic, and molecular genetic
ETO+, or CBFBMYH11+ acute myeloid leukemia findings. Cytometry B Clin Cytom 62: 2538.

http://tah.sagepub.com 15
Therapeutic Advances in Hematology 7(1)

Walter, R., Gooley, T., Wood, B., Milano, F., myeloid leukaemia derived from the MRC AML 10
Fang, M., Sorror, M. etal. (2011) Impact of trial. United Kingdom Medical Research Councils
pretransplantation minimal residual disease, as Adult and Childhood Leukaemia Working Parties. Br
detected by multiparametric flow cytometry, on J Haematol 107: 6979.
outcome of myeloablative hematopoietic cell
Yin, J., OBrien, M., Hills, R., Daly, S., Wheatley,
transplantation for acute myeloid leukemia. J Clin
K. and Burnett, A. (2012) Minimal residual disease
Oncol 29: 11901197.
monitoring by quantitative RT-PCR in core binding
Weisser, M., Kern, W., Rauhut, S., Schoch, factor AML allows risk stratification and predicts
C., Hiddemann, W., Haferlach, T. etal. (2005) relapse: results of the United Kingdom MRC AML-
Prognostic impact of RT-PCR-based quantification 15 trial. Blood 120: 28262835.
of WT1 gene expression during MRD monitoring of
Zhu, H., Zhang, X., Qin, Y., Liu, D., Jiang,
acute myeloid leukemia. Leukemia 19: 14161423.
H., Chen, H. etal. (2013) MRD-directed risk
Wheatley, K., Burnett, A., Goldstone, A., Gray, stratification treatment may improve outcomes of
Visit SAGE journals online R., Hann, I., Harrison, C. etal. (1999) A simple, t(8;21) AML in the first complete remission: results
http://tah.sagepub.com
robust, validated and highly predictive index for from the AML05 multicenter trial. Blood 121:
SAGE journals the determination of risk-directed therapy in acute 40564062.

16 http://tah.sagepub.com

S-ar putea să vă placă și