Clinical Significance of Minimal Residual Disease in Patients With Acute Leukaemia Undergoing Haematopoietic Stem Cell Transplantation

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

review

Clinical significance of minimal residual disease in patients


with acute leukaemia undergoing haematopoietic stem cell
transplantation

Dario Campana1 and Wing Leung2,3


1
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 2Department of Bone
Marrow Transplantation and Cellular Therapy, St Jude Children’s Research Hospital, and 3Department of Pediatrics, College of
Medicine, University of Tennessee Health Science Center, Memphis, TN, USA

Summary insights have been gained into the pathogenesis of acute leu-
kaemia. In turn, this has led to a mounting arsenal of novel
In patients with acute leukaemia, the relative risk of relapse
treatment agents. Perhaps more than ever before, it is now
influences the choice between chemotherapy and haemato-
imperative to precisely identify patients who have high
poietic stem cell transplantation (HSCT). The demonstration
chances of cure with standard therapy, those who could be
that minimal residual disease (MRD) is the strongest overall
candidates for experimental therapies, and those for whom
prognostic indicator and can identify patients who are unli-
haematopoietic stem cell transplantation (HSCT) is the only
kely to be cured by standard chemotherapy has added a
possible curative option. In this regard, presenting clinical
powerful new factor to consider when making this decision.
and/or biological features can be helpful but their prognostic
There is substantial data indicating that the likelihood of
accuracy is limited.
relapse after transplant is directly correlated with levels of
Levels of residual disease at any time point during treat-
MRD before transplant. This knowledge can be used to
ment represent an essential parameter for drawing meaning-
adjust the timing of HSCT, and guide the selection of donor,
ful prognostic algorithms and selecting the best available
conditioning regimen, and post-HSCT strategies to maximize
treatment strategies. Achievement of morphological remission
the graft-versus-leukaemia effect. Because MRD emerging
after initial treatment is a prerequisite for cure but patients
post-transplant carries a dire prognosis, its detection can
who are in morphological remission may have varying,
trigger withdrawal of immunosuppression, additional cellular
sometimes high, levels of minimal residual disease (MRD), a
and molecular therapies, or preparations for a second HSCT.
concept proposed well before it was possible to detect it
Although it is not yet clear whether any of these actions will
(Mathe et al, 1966). Initial efforts to measure MRD tried to
significantly improve outcome, it is likely that they will be
exploit possible differences in the growth patterns of normal
most effective for patients with a relatively low tumour bur-
and leukaemia cells but results appeared to be largely subjec-
den, who can be identified only through MRD testing. In
tive and hard to reproduce (Harris & Freireich, 1970; Moore
this article, we review the clinical significance of MRD in the
et al, 1974; Mertelsmann et al, 1981). Cytogenetic studies can
context of autologous and allogeneic HSCT.
identify residual disease in some patients considered to be in
complete clinical and haematological remission, a finding
Keywords: acute lymphoblastic leukaemia, acute myeloid
that correlates with a higher risk of relapse (Freireich et al,
leukaemia, minimal residual disease, haematopoietic stem cell
1992; Chen et al, 2011). This method, however, has a limited
transplantation.
sensitivity, even when it is complemented by fluorescence
in situ hybridization (Bacher et al, 2006).
With the increasing understanding of the genetic lesions
Current approaches to measure MRD in acute
underlying leukaemogenesis and of the factors that influence
lymphoblastic leukaemia (ALL) rely primarily on polymerase
the growth and drug resistance of leukaemic cells, important
chain reaction (PCR) amplification of clonally rearranged
antigen-receptor genes and on flow cytometry (Bruggemann
et al, 2010; Campana, 2012). Measurements of MRD in acute
Correspondence: Dario Campana, Department of Paediatrics, Yong myeloid leukaemia (AML) are based on PCR amplification
Loo Lin School of Medicine, National University of Singapore, of leukaemia-associated genetic abnormalities or on flow
Centre for Translational Medicine, 14 Medical Drive, Level 9-02, cytometric detection of leukaemia-associated immunopheno-
Singapore 117599, Singapore. types (Shook et al, 2009; Buccisano et al, 2012). After HSCT,
E-mail: paedc@nus.edu.sg MRD can also be monitored by analysis of short tandem

ª 2013 John Wiley & Sons Ltd First published online 9 May 2013
British Journal of Haematology, 2013, 162, 147–161 doi:10.1111/bjh.12358
Review

repeats (STR) or variable number tandem repeats (VNTR) determined at diagnosis (Knechtli et al, 1998a). MRD was
chimerism (Antin et al, 2001). The characteristics of the ‘high’ in 12 patients, ‘low’ in 11 and ‘negative’ in 41; after
methodologies for MRD detection have been extensively excluding eight patients who had an isolated extramedullary
reviewed elsewhere and we refer the readers to the above relapse and were MRD-negative in the bone marrow prior to
articles for a list of relevant publications; Table I summarizes HSCT, the 2-year event-free survival (EFS) for the three
their main features. We here focus on the clinical importance groups was 0%, 36%, and 73%, respectively (P < 0001;
of MRD in the context of HCST. Knechtli et al, 1998a). Using a similar methodology, Bader
et al (2002) studied 41 children and adolescents who were in
first or subsequent remission at the time of HSCT: MRD was
Prognostic significance of MRD prior to HSCT
high in 17 patients, low in 10 and negative in 14; their 5-year
EFS was 23%, 48% and 78%, respectively (P = 0022). A
Acute lymphoblastic leukaemia
third pioneering study was performed by van der Velden
The prognostic significance of MRD levels prior to HSCT in et al (2001) who targeted antigen-receptor genes with a more
children and adolescents with ALL is well established contemporary approach, based on patient-specific primers
(Table II). Knechtli et al (1998a) evaluated MRD by PCR complementary to the unique junctional region identified at
amplification of antigen-receptor genes in 64 children with diagnosis (also known as allele-specific oligonucleotides,
ALL (19 in first remission) 6–81 d prior to HSCT. In this ASO); MRD was quantified by ‘real-time’ (RQ-) PCR: relapse
study, MRD was analysed by using consensus primers to post-HSCT occurred in four of the six MRD-positive and
amplify rearranged immunoglobulin and/or T-cell receptor two of the 11 MRD-negative patients (van der Velden et al,
(TCR) genes; the PCR products were then separated accord- 2001). The results of these three studies were combined
ing to their size by polyacrylamide gel electrophoresis and with unpublished data in a landmark analysis performed
transferred to a nylon membrane by electroblotting. High- by the Pre-BMT MRD Study Group including 140 children
level MRD (approximately 1–01%) was defined when a with ALL (85 in second or higher remission; Krejci et al,
clonal band was evident; low-level MRD (01–0001%) when 2003). The 5-year EFS for patients with high, low or
a band became evident only after probing the membrane undetectable MRD prior to HSCT was 21%, 41% and
with an oligonucleotide complementary to the junctional 75%, respectively (P < 0001); MRD was an independent
sequence of the predominantly rearranged gene as prognostic factor in a multivariable analysis.

Table I. Methods to monitor minimal residual disease in patients undergoing haematopoietic stem cell transplantation.

PCR of genetic Chimerism assay by


PCR of IG/TCR genes abnormalities Flow cytometry STR/VNTR

Sensitivity 10 4 to 10 5 10 4 to 10 5 10 3 to 10 5 10 2
Applicability B-lineage ALL: c. 95% B-lineage ALL: c. 35% B-lineage ALL: >95% 100%
T-lineage ALL: c. 95% T-lineage ALL: c. 15% T-lineage ALL: >95%
AML: <10% AML: c. 40% AML: c. 95%
Advantages  Sensitivity  Sensitivity  Applicable to most patients  Applicable to all patients
 Specificity  Specificity and  Rapid  No diagnostic sample required
 Accurate quantification stability of marker  Quantitative  Rapid
 Standardized  Fast  Established predictive value  Standardization possible
 Stability of DNA  Relatively cheap  Additional information on  Relatively cheap
 Established normal haematopoiesis  Additive information on
predictive value impending graft rejection
 Stability of DNA
Disadvantages  Not useful for AML  Predictive value less  Potential for phenotypic shifts  Limited sensitivity
 Establishment of PCR established  Relatively expensive  Applicable only after HSCT
conditions labour-intensive*  Variability in transcript:  Extensive expertise required
 Potential for clonal evolution* cell ratio
 Extensive expertise required  Not applicable to
 Relatively expensive most patients
 Instability of RNA

PCR, polymerase chain reaction; IG, immunoglobulin; TCR, T-cell receptor; STR, short tandem repeats; VNTR, variable number tandem repeats;
ALL, acute lymphoblastic leukaemia; AML, acute myeloid leukaemia; HSCT, haematopoietic stem cell transplantation.
*These limitations are overcome by newer methods to measure minimal residual disease using deep sequencing of IG/TCR genes, which can also
increase the sensitivity beyond 10 6 (Faham et al, 2012; Wu et al, 2012).

148 ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 147–161
Table II. Prognostic significance of minimal residual disease prior to haematopoietic stem cell transplantation in acute lymphoblastic leukaemia.

Outcome

MRD Pos

Type No. of MRD Low High


Study of HSCT patients Age group Method Estimate Neg MRD* MRD* P

Knechtli et al (1998a) Allo 64 Paediatric PCR IG/TCR 2-year EFS 73% 36% 0% <0001
Bader et al (2002) Allo 41 Paediatric PCR IG/TCR 5-year EFS 78% 48% 23% 0022

ª 2013 John Wiley & Sons Ltd


Dombret et al (2002) Allo 63 Adult RT-PCR BCR-ABL1 3-year incidence 41% 75% 001
of relapse
Mortuza et al (2002) Auto 13 Adult PCR IG/TCR Relapses/patients 0/6 6/7 0005‡
studied
Krejci et al (2003) Allo 140 Paediatric PCR and ASO 5-year EFS 75% 41% 21% <0001
RQ-PCR IG/TCR
Sramkova et al (2007) Allo 25 Paediatric ASO RQ-PCR IG/TCR Relapses/patients 1/17 6/8 0001‡

British Journal of Haematology, 2013, 162, 147–161


studied
Spinelli et al (2007) Allo 37 Adult ASO RQ-PCR 3-year incidence 0% 46% 0027
IG/TCR or RT of relapse
(RQ)-PCR fusion 3-year OS 80% 49% NS
transcripts
Paganin et al (2008) Allo 25 Paediatric ASO RQ-PCR IG/TCR Relapses/patients 2/14 8/11 0005‡
studied
Bader et al (2009) Allo 91 Paediatric ASO RQ-PCR IG/TCR 4-year incidence 11% 20%/64%† 57% <0001
of relapse
4-year EFS 64% 48%/19%† 31% 0036
Patel et al (2010) Allo 36 Adult PCR and ASO Relapses/patients 4/23 2/13 NS
RQ-PCR IG/TCR studied
5-year EFS 50% 52% NS
Patel et al (2010) Auto 25 Adult PCR and ASO 5-year LFS 77% 25% 001
RQ-PCR IG/TCR
Giebel et al (2010) Auto 103 Adult Flow cytometry or 5-year LFS 57% 17% <0001
ASO RQ-PCR
IG/TCR or
RT(RQ)-PCR fusion
transcripts
Elorza et al (2010) Allo 31 Paediatric Flow cytometry 2-year EFS 75% 20% <001
Leung et al (2011) Allo 64 Paediatric Flow cytometry 5-year incidence 6% 28% 003
of relapse
5-year OS 68% 30% 0008
Sanchez-Garcia et al (2012) Allo 102 Both Flow cytometry 5-year LFS 66% 43% 0% <0001
5-year OS 52% 29% 0% <0001

149
Review
Review

HSCT, haematopoietic stem cell transplantation; MRD, minimal residual disease; allo, allogeneic; auto, autologous; UCB, umbilical cord blood; PCR, polymerase chain reaction; IG, immunoglobulin
These early findings in childhood ALL were confirmed

genes; TCR, T-cell receptor genes; RT, reverse transcriptase; ASO, allele-specific oligonucleotides; RQ, real time; RT, reverse transcription; EFS, event-free survival; OS, overall survival; LFS, leukae-
0006

005

002
and extended in more recent studies (Sramkova et al, 2007;

P
Paganin et al, 2008; Elorza et al, 2010). A prospective study
of 91 children with relapsed ALL who received HSCT in sec-
ond or subsequent remission demonstrated the relationship

MRD*
High between MRD levels prior to HSCT as measured by ASO
RQ-PCR and risk of relapse: 4-year EFS was 31% for patients
MRD Pos

29% with MRD  01% (n = 33), 19% for patients with MRD

30%

30%
 001–<01% (n = 12), 48% for those with MRD detectable
below <001% (n = 10) and 64% for patients with undetect-
MRD*

able MRD (n = 36; P = 0036); cumulative incidence of


Low

relapse was 54%, 64%, 20% and 11%, respectively


(P < 0001; Bader et al, 2009). In this study, MRD was also
predictive of outcome among patients with different present-
MRD

54%

16%

55%

ing risk features; in a multivariate Cox proportional hazards


Neg

model including sex, age, remission status, time and site of


relapse, immunophenotype, donor type, T-cell depletion,
time to transplantation, and graft-versus-host disease
2-year incidence

(GvHD), MRD was the only independent prognostic factor


of relapse

(Bader et al, 2009). In an analysis of 64 patients with high-


4-year LFS

3-year LFS
Outcome

Estimate

risk ALL who underwent HSCT at St. Jude Children’s


Research Hospital, we found that MRD before transplant as
measured by flow cytometry was a significant and indepen-
dent prognostic factor: 5-year cumulative incidence of relapse
was 28% for the 30 patients who had MRD  001% and
or fusion transcripts§

†Two subgroups with relatively low levels of MRD were analysed in this study: <001% and 001% to <01%.

6% for the 34 with MRD <001% or undetectable


(P = 003); overall survival (OS) rates were 30% and 68%,
Flow cytometry;

Flow cytometry
PCR IG/TCR

respectively (P = 0008; Leung et al, 2011). Among patients


with detectable MRD, higher levels of MRD predicted a
Method

poorer outcome (Leung et al, 2012).


The prognostic importance of MRD prior to HSCT has
also been demonstrated for adult patients with ALL
(Table II). Detection of BCR-ABL1 transcripts prior to trans-
Age group

plant in patients with Philadelphia (Ph)-positive ALL was a


Paediatric

significant predictor of outcome in an early study: 3-year


‡Calculated by Fisher’s exact test based on the data reported in the article.
Both

incidence of relapse was 75% for patients with detectable


transcripts by semi-quantitative reverse-transcriptase (RT)
PCR (n = 39) and 41% for those with no detectable tran-
scripts (n = 24; P = 001; Dombret et al, 2002). More
patients
No. of

recently, it was reported that the degree of response to imati-


170

86

nib prior to transplant in patients with this leukaemia


subtype, as measured by RT(RQ)-PCR MRD monitoring,
was a major determinant of relapse after HSCT (Lee et al,
of HSCT

2012). MRD before transplant evaluated in 37 patients with


Type

UCB

UCB

either Ph-positive or Ph-negative ALL by using RT(RQ)-


mia-free survival; NS, not significant.

PCR targeting BCR-ABL1 or MLL-AFF1 fusion transcripts, or


§PCR methology used not stated.
*Definition varies among studies.

ASO RQ-PCR targeting rearrangements of antigen-receptor


genes yielded prognostic information: cumulative incidence
of relapse was 46% for MRD-positive (n = 25) and 0% for
Bachanova et al (2012)
Table II. (Continued)

MRD-negative patients (n = 12; P = 0027) after a median


Ruggeri et al (2012)

follow-up of 23 months (Spinelli et al, 2007). In another


study supporting the prognostic significance of pre-HSCT
MRD, this was measured by flow cytometry in 102 children
and adult patients (including 55 older than 14 years of age)
Study

and found to be >01% in 18, 001 to  01% in 12 and

150 ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 147–161
Review

undetectable in 72 patients; leukaemia-free survival (LFS) with Ph-negative ALL (Giebel et al, 2010). The 5-year LFS
rates were 0%, 43% and 66%, respectively (P < 0001); OS was 17% for patients with MRD  01% prior to HSCT by
rates were 0%, 29% and 52% (P < 0001), with MRD being either flow cytometry or ASO RQ-PCR amplification of anti-
the most significantly adverse prognostic factor in a multi- gen-receptor genes and 57% for patients with <01% or
variable analysis (Sanchez-Garcia et al, 2012). undetectable MRD (P = 00002). The difference was particu-
It should be noted that significant correlations between larly striking for patients with T-lineage ALL: 8% (n = 33)
MRD and outcome did not emerge in some analyses. Thus, vs. 62% (n = 13; P = 0001), and MRD remained the only
in a study using PCR amplification of immunoglobulin genes predictor in a multivariate analysis including other known
using either consensus primers to detect the presence of prognostic factors (Giebel et al, 2010).
clonal rearrangements or ASO PCR, MRD was a prognostic
factor overall but was not predictive of relapse among the 19
Acute myeloid leukaemia
patients who received allogeneic HSCT in first remission
(Mortuza et al, 2002). Lack of prognostic power was also In a study of paediatric AML, levels of the WT1 gene were
noted in a subsequent study from the same group, this time measured 2 weeks prior to allogeneic HSCT in peripheral
also including ASO RQ-PCR to detect MRD: among 36 blood of 36 patients who were in morphological remission
patients with B-lineage ALL receiving allogeneic HSCT in (Jacobsohn et al, 2009). Using a cut-off level to define MRD
first remission, 13 were MRD positive and 2 relapsed as positivity based on the highest level of WT1 determined in
compared to 4 of the 23 patients with MRD negative or most healthy individuals by RT-PCR (after normalizing the
<001%; 5-year EFS was 52% vs. 50% (Patel et al, 2010). A values to those obtained in the WT1-positive cell line K562),
multitude of factors may have caused the discrepancy 11 patients were considered to be MRD-positive: seven
between these and other studies, including differences in relapsed and two died of transplant-related mortality while
cohort composition, pre-transplant chemotherapy and condi- none of the 25 MRD-negative patients relapsed (although
tioning, donor source and graft-versus-leukaemia effect post- three did not engraft and eight died of transplant-related
transplant. The collective results of correlative studies, mortality); 5-year EFS was 68% vs. 18% (P = 0007), and
however, would indicate that MRD levels prior to transplant MRD was the only significant prognostic factor in a multi-
does exert a considerable influence on the success of alloge- variate analysis (Jacobsohn et al, 2009). We used flow cytom-
neic HSCT. Recent data with large cohorts of patients under- etry to measure MRD before HSCT in 58 children and
going unrelated cord blood transplantation confirmed the adolescents with AML: 5-year cumulative incidence of relapse
prognostic power of pre-HSCT MRD. One study determined was 37% for the 25 with detectable MRD and 7% for the 33
MRD levels by either flow cytometry or molecular methods MRD-negative patients (P = 0005); OS rates were 47% and
in 170 children with ALL (72 in first remission, 98 in second 68%, respectively (P = 005; Leung et al, 2011).
or higher remission) and the 4-year LFS was 29% for MRD In the context of autologous HSCT, it is clear that AML
positive (n = 74) and 54% for MRD-negative patients cells present in the inoculum can contribute to relapse (Bren-
(n = 96; P = 0006; Ruggeri et al, 2012). Another study used ner et al, 1993) and studies have shown significant correla-
flow cytometry to study MRD in 86 children and adult ALL tions between levels of MRD as measured by flow cytometry
patients: patients with detectable MRD before transplant or PCR amplification of WT1 prior to autologous HSCT
(n = 10) had a 2-year relapse rate of 30% and 3-year LFS of with risk of relapse (Feller et al, 2005; Osborne et al, 2005).
30% as compared to 16% and 55% for the MRD-negative In an early study using flow cytometry, 12 patients had
patients (n = 76; P = 002; Bachanova et al, 2012). MRD higher than 0035% (the best discriminative threshold
Perhaps not unexpectedly, autologous HSCT is associated according to these investigators) prior to autologous HSCT
with a very poor outcome when the graft is MRD-positive and all relapsed at a median of 7 months post-transplant
(Mizuta et al, 1999). Among the 13 adult patients who (Table III). By contrast, among the 19 with lower levels of
received autologous HSCT studied by Mortuza et al (2002), MRD, five patients relapsed (median time post-transplant, of
six of the seven with MRD-positive harvests subsequently 11 months) and 14 remained in remission with a median
relapsed whereas none of the six who received an MRD- follow-up of 56 months (P < 0001); MRD status was the
negative graft did. In the subsequent study by the same strongest prognostic factor in a multivariate analysis (Vend-
group, MRD positivity prior to autologous HSCT was signif- itti et al, 2003). Subsequently, this group of investigators
icantly associated with a lower rate of LFS at 5-years: 25% extended their studies to a total of 77 patients who under-
vs. 77% for MRD-negative patients (P = 001); of the nine went autologous or allogeneic HSCT: 5-year LFS was 22%
harvest samples studied, four were positive and three of the for the 42 who had MRD >0035% before transplant and
patients who received them relapsed within 9 months from 55% for the 35 who had lower MRD (Maurillo et al, 2008).
transplant while none of the five who received MRD-negative The prognostic importance of MRD was clearly demon-
grafts relapsed after a median follow-up of 84 years (Patel strated in a recent study of 99 patients receiving allogeneic
et al, 2010). A study of the European Study Group for Adult HSCT in first remission: the 24 with detectable MRD by flow
ALL analysed data from 103 recipients of autologous HSCT cytometry had a 2-year OS of 30% as compared to 77% for

ª 2013 John Wiley & Sons Ltd 151


British Journal of Haematology, 2013, 162, 147–161
Review

Table III. Prognostic significance of minimal residual disease prior to haematopoietic stem cell transplantation in acute myeloid leukaemia.

Outcome

Type of No. of MRD MRD


Study HSCT patients Age group Method Estimate Neg Pos* P

Venditti et al (2003) Auto 31 Adults Flow cytometry Relapses/patients studied 5/19 12/12 <0001†
Maurillo et al (2008) Allo/Auto 77 Adult Flow cytometry 5-year LFS 55% 22% 0004
Jacobsohn et al (2009) Allo 36 Paediatric RT-PCR WT1 1-year incidence of relapse 0% 76% <0001
5-year EFS 68% 18% 0007
Leung et al (2011) Allo 58 Paediatric Flow cytometry 5-year incidence of relapse 7% 37% 0005
5-year OS 68% 47% 005
Walter et al (2011) Allo 99 Both Flow cytometry 2-year incidence of relapse 18% 65% <0001
2-year OS 77% 30% <0001

HSCT, haematopoietic stem cell transplantation; MRD, minimal residual disease; allo, allogeneic; auto, autologous; RT-PCR, reverse transcrip-
tion-polymerase chain reaction; LFS, leukaemia-free survival; EFS, event-free survival; OS, overall survival.
*Definition varies among studies.
†Calculated by Fisher’s exact test based on the data reported in the article.

the 75 MRD-negative patients; 2-year rates of relapse were a median follow-up of 43 months (P < 00001; Knechtli
65% and 18%, respectively; MRD remained a significant pre- et al, 1998b). The authors suggested that technical reasons,
dictor of outcome after adjusting for known prognostic vari- such as poor quality sample, clonal evolution and infrequent
ables, and when analyses were restricted to patients who were sampling could explain the negative findings in 3 of the 4
in cytogenetic remission (Walter et al, 2011). patients; the fourth had an isolated testicular relapse
65 months after HSCT. Similar predictive findings were
reported by Uzunel et al (2003) who used ASO-PCR to
Prognostic significance of MRD after HSCT
monitor MRD in 23 children and nine adults with ALL after
HSCT: eight of the nine MRD-positive patients relapsed
Acute lymphoblastic leukaemia
between 05 and 30 months from the MRD finding, in con-
The signal detected by an MRD assay might reveal, directly trast to six relapses among the 23 MRD-negative patients,
or indirectly, the presence of leukaemia stem-cells or simply with MRD remaining as the sole significant predictor in a
originate from end-stage cells lacking self-renewing potential. multivariate analysis. The study of Spinelli et al (2007),
If the former is true, one would expect that detection of which used RQ-PCR, found that the cumulative incidence of
MRD post-HSCT will be followed by overt relapse in the relapse post-transplant was 80% for the 14 patients who were
majority of cases unless further therapy is initiated or graft MRD-positive on day +100 versus 7% for the 17 MRD-nega-
anti-leukaemia mechanisms prevail. Indeed, most studies tive patients (P = 00006).
have shown a strong correlation between MRD post-HSCT Other investigators used flow cytometry to monitor MRD
and relapse (Table IV). post-HSCT (Sanchez et al, 2002). A recent study applied this
An early study used consensus primers and PCR amplifi- method as well as RT(RQ)-PCR amplification of BCR-ABL1
cation to detect clonal rearrangements of immunoglobulin to monitor 139 patients and found that those who were
genes in 20 patients with ALL, 17 of whom had been trans- MRD positive post-transplant had a 2-year EFS of 54% com-
planted with relapse or refractory disease. Among the 13 pared to 80% for MRD-negative patients (P < 0001; Zhao
MRD-positive patients in the first 100 d after transplant, 11 et al, 2012). Relapse could be predicted by the MRD status
relapsed and the other two died; there was one relapse in 22 of 38 patients, and the time from the first MRD posi-
among the seven patients who were MRD-negative, while tive result to overt relapse was between 1 and 3 months.
two died and the remaining four were alive and well at the
time of the report (Radich et al, 1995). Using consensus
Acute myeloid leukaemia
primer-based PCR to target immunoglobulin and TCR genes,
Knechtli et al (1998b) monitored MRD in children with ALL A few studies reported small cohorts of patients with genetic
who received allogeneic HSCT. Among the 68 evaluable subtypes of AML monitored by PCR amplification of fusion
patients with B- or T-lineage ALL, MRD was detected in 36, transcripts (Elmaagacli et al, 1998; Bacher et al, 2009), while
either as a consistent positive finding or emerging after an others were performed using WT1 as a PCR target (Elmaaga-
MRD negative finding: relapse occurred in 28 of these cli et al, 2000; Ogawa et al, 2003; Candoni et al, 2011). From
patients, at 15–11 months (median, 3 months) from the first the results of these studies, however, it is difficult to deter-
MRD-positive sample. By contrast, only four of the 32 who mine whether MRD monitoring by these methods is predic-
were consistently MRD-negative subsequently relapsed, with tive of relapse and, if so, whether it would provide timely

152 ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 147–161
Review

Table IV. Prognostic significance of minimal residual disease after haematopoietic stem cell transplantation (HSCT).

Outcome

Type No. Age MRD MRD


Study of HSCT of patients group Method Estimate Neg Pos* P

Acute lymphoblastic leukaemia


Radich et al (1995) Allo 20 Adult PCR IG Relapses/patients studied 1/7 11/13 0004†
Knechtli et al (1998b) Allo 68 Paediatric PCR IG/TCR Relapses/patients studied 4/32 28/36 <0001†
Uzunel et al (2003) Allo 32 Both ASO PCR IG/TCR Relapses/patients studied 6/23 8/9 0004†
Spinelli et al (2007) Allo 25 Adult ASO RQ-PCR 3-year incidence of relapse 7% 80% <0001
IG/TCR or RQ-PCR
fusion transcripts
Zhao et al (2012) Allo 139 Both Flow cytometry or 2-year incidence of relapse 8% 54% <0001
RT(RQ)-PCR 2-year EFS 80% 54% <0001
BCR-ABL1
Acute myeloid leukaemia
Elmaagacli et al (2000) Allo 38 Adult RT-PCR WT1 Relapses/patients studied 5/24 7/14 NS†
Kwon et al (2012) Allo 21 Adult RT(RQ)-PCR WT1 Relapses/patients studied 0/11 9/10 <0001†
Diez-Campelo et al (2009) Allo 41 Adult Flow cytometry 4-year EFS 74% 17% 001
4-year OS 73% 25% 0002

HSCT, haematopoietic stem cell transplantation; MRD, minimal residual disease; allo, allogeneic; auto, autologous; PCR, polymerase chain reac-
tion; IG, immunoglobulin genes; TCR, T-cell receptor genes; ASO, allele-specific oligonucleotides; RQ, real time; RT, reverse transcription, EFS,
event-free survival; OS, overall survival; NS, not significant.
*Definition varies among studies.
†Calculated by Fisher’s exact test based on the data reported in the article.

clinical information and/or superior information to chime- chemotherapy or undergo HSCT. In this regard, studies of
rism studies. To this end, in a recent analysis of 21 patients, MRD have helped identify patients who still harbour high
the 11 who had low and stable WT1 levels by RT(RQ)-PCR levels of disease below the detection capabilities of morpho-
post-HSCT also had complete chimerism and remained in logical analysis and have a high risk of relapse if treated with
complete remission with a median follow-up of 27 months, standard chemotherapy. For example, in our Total XIII study
whereas nine of the 10 patients with WT1 overexpression where MRD was not used to guide treatment, nine children
relapsed (P < 0001), with the WT1 increase preceding chi- with ALL had MRD levels  1% at the end of remission
merism changes (Kwon et al, 2012; Table IV). Other investi- induction therapy (day 42) and a 5-year cumulative inci-
gators used flow cytometry to monitor MRD post-HSCT. dence of relapse of 72%: only two of nine patients in this
Diez-Campelo et al (2009) studied MRD in 41 patients with group were alive and in remission at 25 and 45 years after
AML or myelodysplastic syndrome and found that MRD diagnosis, and one of them had undergone allogeneic HSCT
 01% on day 100 post-HSCT provided prognostic informa- because of high-risk presenting features (Coustan-Smith et al,
tion: 4-year EFS was 17% as compared to 74% for patients 2000). In the subsequent Total XV study, all patients with
with lower MRD (P = 001); OS was 73% vs. 25% MRD  1% at the end of remission induction therapy
(P = 0002); MRD was an independent predictor of outcome (n = 23, 46% of the 498 enrolled in the trial) were eligible
in a multivariate analysis. Of note, Miyazaki et al (2012) for allogeneic HSCT after receiving reintensification therapy:
compared results of MRD detection post-allogeneic HSCT by 12 of the 23 patients were alive and well at the time of the
flow cytometry, RT(RQ)-PCR amplification of WT1 or of report (Pui et al, 2009). In a cohort of 31 adult patients with
fusion transcripts in a group of 31 patients with AML and 10 ALL and detectable MRD at the end of post-remission che-
with ALL: they found that flow cytometry data predicted out- motherapy, the 5-year LFS was 80% with allogeneic HSCT,
come while molecular testing did not. These results recall our 53% with autologous HSCT, and 0% with chemotherapy
recent experience comparing flow cytometry and PCR ampli- (P = 0003; Laane et al, 2006). In the German Multicentre
fication of fusion transcripts to monitor MRD in childhood Study Group for Adult ALL, the probability of continuous
AML (Inaba et al, 2012a). complete remission at 5 years for patients with Ph-negative
ALL who failed to achieve MRD negative status was signifi-
cantly higher for those who received allogeneic HSCT in first
MRD to guide pre- and post-transplant
remission: 66% vs. 12% for those who received chemother-
management strategies
apy alone (P < 00001; Gokbuget et al, 2012).
For patients with acute leukaemia who achieve remission, Children and adolescents with AML and persistent MRD
a key question is whether they should proceed with after remission induction chemotherapy are also at a high

ª 2013 John Wiley & Sons Ltd 153


British Journal of Haematology, 2013, 162, 147–161
Review

risk of relapse (Coustan-Smith et al, 2003; van der Velden remission for the duration of the imatinib course, although
et al, 2010). There is a paucity of studies comparing the rela- three relapsed after imatinib discontinuation (Wassmann
tive benefits of chemotherapy versus HSCT in these patients et al, 2005). Pfeifer et al (2012) recently compared the merits
but a protocol in which persistent MRD (  01%) after three of prophylactic (n = 26) versus MRD-triggered imatinib
courses of therapy was an indication for transplant in first (n = 29) post-HSCT. Prophylactic imatinib reduced the
remission yielded cure rates among the highest reported to occurrence of molecular relapse but 5-year overall survival
date, suggesting a benefit from this strategy (Rubnitz et al, for both groups of patients was high (80% vs. 75%). The
2010a). In the study by Maurillo et al (2008) with adult authors noted that patients with early molecular recurrence
AML patients who had MRD >0035% after remission induc- and/or higher levels of BCR-ABL1 transcripts appeared to
tion therapy, 60 relapses occurred among the 74 patients have limited benefit from imatinib: six of 19 eventually
who were treated with either chemotherapy alone or autolo- relapsed and three were switched to dasatinib after imatinib
gous HSCT as compared to 6 relapses (or failures because of failed to induce MRD-negativity (Pfeifer et al, 2012). Finally,
treatment-related mortality) among the 14 patients who Platzbecker et al (2012) recently reported a study in which
received allogeneic HSCT. azacitidine was given to 20 patients with AML or myelodys-
Immunomodulatory strategies post-HSCT may be consid- plastic syndrome and a decrease in donor chimerism to
ered for patients with high-levels of MRD before transplant <80% in peripheral blood CD34+ cells; 16 patients
but whether a graft-versus-leukaemia effect can have a signif- responded with an increase or stabilization of donor chime-
icant clinical impact in ALL is unclear. Lankester et al (2010) rism and appeared to have delayed occurrence of relapse.
stratified 48 children with ALL according their MRD level:
18 had MRD pre-HSCT of  001% and were eligible for
Practical issues
early ciclosporin tapering followed by up to four donor
lymphocyte infusions (DLI). There was no evidence of Minimal residual disease assays have been considerably
benefit for this approach and the EFS for this group was only refined over the last two decades. Current methods for PCR
19%, although relapses appeared to occur later than expected amplification of antigen-receptor genes are based on primers
or presented at extramedullary sites (Lankester et al, 2010). complementary to the leukaemia-specific junctional regions
There is data suggesting that for patients with emerging (Bruggemann et al, 2010). A new development in this area is
mixed chimerism post-HSCT, reduction of immunosuppres- the use of consensus primers to universally amplify all rear-
sion to increase the graft-versus-leukaemia effect or the initi- ranged immunoglobulin and TCR gene segments present in a
ation of cellular therapy might be beneficial (Formankova leukaemic clone followed by high-throughput sequencing.
et al, 2003; Bader et al, 2004; Zeiser et al, 2005; Rettinger This approach can identify all clonal gene rearrangements at
et al, 2011; Buckley et al, 2012). For example, in the study diagnosis (or relapse) and monitor their fluctuations during
reported by Bader et al (2004), 16 of the 31 patients with treatment (Boyd et al, 2009; Faham et al, 2012; Wu et al,
increasing mixed chimerism who received such forms of 2012). In experiments comparing this assay with flow cytom-
immunotherapy reverted to complete chimerism, and 13 etry and ASO-PCR, we found it to be capable of detecting
were alive and disease-free at the last follow-up (median, MRD as well as the other methods but to have a higher sen-
866 d), while 11 of the 15 patients who were not treated sitivity: it can potentially detect residual disease at levels
relapsed. Early studies reported a few patients with ALL in below 1 leukaemic cell in 106 leucocytes if sufficient DNA is
whom MRD post-HSCT remained at low levels or disap- available (Faham et al, 2012). Flow cytometric methods have
peared in association with GvHD or DLI (Kanamori et al, also improved with the development of more sophisticated
1997; Knechtli et al, 1998b; Miglino et al, 2002; Bradfield instruments and the discovery of new markers to distinguish
et al, 2004; Dominietto et al, 2007) but the evidence for such leukaemic from normal cells (Coustan-Smith et al, 2011).
an effect seems more convincing in AML (Zeiser et al, 2005; Finally, the application of single nucleotide polymorphism
Rettinger et al, 2011). A recent study reported 105 patients arrays and whole-genome sequencing has identified many
with AML who were MRD-positive post-HSCT; 49 received previously unknown abnormalities in leukaemia, which could
low-dose interleukin 2 (IL2), and 56 received DLI with or expand the pool of potential targets for molecular analysis of
without IL2. The latter had a significantly lower cumulative MRD, particularly in AML (Ley et al, 2010; Ding et al,
risk of relapse and higher LFS, which was comparable to that 2012). Thus, genes mutated at diagnosis might be used to
of MRD-negative patients (Yan et al, 2012). track leukaemic cells during follow-up but their suitability
Besides immunomodulatory strategies, other types of for this purpose remains largely untested.
intervention may reduce the adverse impact of MRD post- Figure 1 summarizes the overall application of MRD to
HSCT. One study focused on patients with Ph-positive ALL, manage patients with paediatric acute leukaemia at St. Jude
using RT-PCR targeting BCR-ABL1 transcripts to monitor Children’s Research Hospital. For patients with ALL, MRD is
MRD and determine the effects of imatinib post-transplant currently measured primarily with flow cytometry, reserving
(Wassmann et al, 2005). Among 27 patients, MRD became ASO RQ-PCR amplification of antigen-receptor genes for
undetectable in 14 after 09–37 months and all remained in patients with no suitable MRD markers or whose MRD

154 ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 147–161
Review

Fig 1. Minimal residual disease risk-adapted


approaches before and after HSCT for acute
leukaemia at St. Jude Children’s Research Hos-
pital. MRD, minimal residual disease; HSCT,
haematopoietic stem cell transplantation; IG,
immunoglobulin genes; TCR, T-cell receptor
genes; GvHD, graft-versus-host disease; DLI,
donor lymphocyte infusion; NK cell natural
killer cell.

results are ambiguous. MRD  1% at the end of remission can track MRD. The requirement for the sequence of the
induction, or persisting MRD post-remission is an indication leukaemia-specific junctional regions to either amplify (by
for HSCT; chemotherapy pre-HSCT is given with the aim of ASO PCR) or recognize (after deep sequencing) leukaemic
reducing MRD levels as much as possible. MRD in patients clones in follow-up samples precludes MRD studies by these
with AML is monitored by flow cytometry and by RQ-PCR approaches if this material is not available. In principle, one
targeting fusion transcripts, although the results of the for- could perform a clonality analysis using consensus primers
mer are given priority (Inaba et al, 2012b). Patients with but this approach would have limited sensitivity (Langerak
residual disease  5% by flow cytometry after Induction 1 or et al, 2012). Flow cytometric studies of MRD are also best
MRD  01% after Induction 2 are candidates for HSCT. performed when the immunophenotypic abnormalities
Patients with MRD  1% at this point receive additional expressed by the leukaemic cells are known, so that tailor-
chemotherapy or natural killer (NK) cell therapy before made antibody panels can be used to track them in remis-
HSCT. Those with lower levels of MRD may also receive sion samples. Alternatively, broad antibody panels designed
additional chemotherapy if the donor is not ready. to detect all known abnormalities can be applied; the chances
Minimal residual disease tests are technically complex and of success for this approach have been increased by the
require highly skilled staff and expensive reagents. Their wealth of new markers and fluorochromes that can now be
systematic use to guide clinical decisions, however, should not used. In the case of an MRD-negative result, however, the
only benefit patients but ultimately save costs, particularly in possibility remains that the leukaemic clone lacks any immu-
the context of HSCT (Goulden et al, 2001). The increasing nophenotypic abnormality and it remains, therefore, unde-
use of MRD in the clinic may tempt laboratories without tectable.
specific expertise in this area to process samples and release The optimal frequency at which MRD measurements
results. This should be avoided at all costs, as the should be performed remains unclear. MRD prior to HSCT
consequences of an incorrect result, e.g., a false-positive result is typically measured at the end of each block of preceding
which indicates transplant in a patient who, in fact, is MRD- chemotherapy; for MRD measurements post-HSCT, the
negative, could be disastrous. In this regard, centres proficient frequency varies. In fact, the optimal schedule may depend
in performing MRD testing can generally offer training as well on the leukaemia subtype. To this end, Ommen et al (2010)
as distance consultation and quality control. It has been diffi- calculated the kinetics of relapse in patients with AML based
cult to draft widely accepted guidelines for MRD testing that on the time relationship between MRD findings and overt
would meet expertise level and quality standards, and be disease recurrence. They found that one test in peripheral
within the cost limits of most centres. Meeting this objective blood every 6 months would be sufficient to detect 90% of
is important to facilitate the interpretation of MRD relapses at a median preceding time of 60 d in AML patients
significance across different studies and implement a more with CBFB-MYH11 AML, but bone marrow testing at more
standardized clinical application of MRD testing. frequent intervals would be necessary in other subtypes, such
A problem that may be encountered by transplant centres as those with RUNX1-RUNXT1 or NPM1 mutations with or
is the lack of available diagnostic (or relapse) material to without FLT3-internal tandem duplications (Ommen et al,
identify the molecular or immunophenotypic signatures that 2010). Our approach is to measure MRD monthly during

ª 2013 John Wiley & Sons Ltd 155


British Journal of Haematology, 2013, 162, 147–161
Review

the first 3 months post-HSCT, followed by measurements at to reduce MRD levels pre-HSCT, or perhaps replace HSCT
6 and 12 months, but additional measurement are performed altogether.
in the case of positive MRD findings or incomplete donor While DLI can be effective, it also has a high risk of
chimerism. GvHD (Kolb et al, 1995). There is considerable evidence that
NK cell alloreactivity can suppress relapse post-HSCT with-
out GvHD (Ruggeri et al, 2002; Giebel et al, 2003; Leung
Conclusions
et al, 2004; Cooley et al, 2010), which has led to the practice
It has been estimated that less than 1% of the published can- of selecting donors with a NK receptor profile that predicts
cer biomarkers enter clinical practice (Kern, 2012). MRD the strongest alloreactive potential. There are also data
clearly belongs to this select group; it is the most useful indicating that allogeneic NK cell infusions can have an anti-
prognostic parameter overall in acute leukaemia. Because leukaemia effect, particularly in AML (Miller et al, 2005;
HSCT is effective but also carries the risk of considerable Rubnitz et al, 2010b). NK cells can be effectively expanded
toxicity, it should be reserved for patients who are unlikely and activated (Fujisaki et al, 2009; Lapteva et al, 2012; Ru-
to be cured with a less intensive and toxic treatment. Thus, jkijyanont et al, 2013), and also redirected against different
modern treatment protocols rely on MRD to identify such leukaemia cell types using clinically applicable methods (Imai
patients. As discussed in this article, data suggest that opting et al, 2005; Shimasaki et al, 2012). To this end, NK cells
for HSCT may improve the outcome for these very-high risk expressing a chimeric receptor that recognizes the CD19
patients (Laane et al, 2006; Maurillo et al, 2008; Pui et al, molecule and delivering activation signals through CD3f and
2009; Rubnitz et al, 2010a; Leung et al, 2011; Buckley et al, 4-1BB markedly enhanced NK cell killing of CD19+ ALL and
2012; Gokbuget et al, 2012), but additional analyses are lymphoma cells, even in autologous setting (Imai et al, 2005;
required. Shimasaki et al, 2012). More recently, we found that a recep-
Minimal residual disease levels prior to HSCT are directly tor containing NKG2D (a NK activating receptor) and the
related to risk of relapse post-HSCT, particularly in ALL signalling molecules CD3f and DAP10 significantly improved
(Krejci et al, 2003; Bader et al, 2009; Leung et al, 2012). It NK cell activity against a variety of cancer cell types (Chang
should be noted, however, that high levels of MRD pre- et al, 2013). Antibodies targeting markers expressed by
transplant do not completely preclude success (Schneider leukaemic cells (e.g., CD19, CD33) could be used to trigger
et al, 2001; Bader et al, 2009; Leung et al, 2012). Moreover, antibody-dependent cytotoxicity, while antibodies against NK
we found that the adverse prognostic impact of any given inhibitory receptors could further augment NK cell activity
level of MRD was lower in children with ALL or AML trans- (Chan et al, 2012). We expect that MRD-directed NK cell
planted in recent years (2000–2007) as compared to earlier therapy will increasingly be used to reduce leukaemia burden
cohorts (Leung et al, 2012). For example, in our study of before HSCT or eradicate MRD post-HSCT.
patients with persistent MRD at <5% at the time of HSCT In summary, MRD has had a major impact on many criti-
treated in the more recent era, the 5-year OS rate was 67% cal facets of the clinical management of patients with acute
for patients with AML and 49% for ALL (Leung et al, 2012). leukaemia, including HSCT. MRD monitoring helps refining
Whether treatment intensification prior to HSCT in efforts the eligibility criteria for HSCT, and can guide treatment
to reduce MRD levels diminishes the risk of relapse post- choices before and after HSCT. As new targeted agents and
HSCT or simply adds toxicity with marginal benefits remains cell-based therapies become available, MRD should provide
to be determined (Goulden & Steward, 2002; Buckley et al, an essential tool for selecting the best candidates for these
2012). To this end, targeted therapies without cross-resis- therapies and measuring their effects.
tance or overlapping toxicity may play a crucial role, as
suggested by the experience with blinatumomab, indicating
Acknowledgements
that patients who achieved MRD-negativity with this agent
had an excellent outcome post-allogeneic HSCT (Handgret- This work was supported in part by the National Medical
inger et al, 2011; Topp et al, 2011). Recent data demonstrat- Research Council of Singapore grant 1299/2011, the Viva
ing clinical responses in patients receiving autologous T cells Foundation for Children with Cancer, the National Institutes
genetically modified to express anti-CD19 chimeric receptors of Health Cancer Center Support (CORE) grant P30
(Porter et al, 2011) provide further indication of the anti- CA021765, the Assisi Foundation of Memphis, and the
leukaemic potential of immune cells, and suggest new ways American Lebanese Syrian Associated Charities (ALSAC).

References transplantation: recommendations from a work- Bachanova, V., Burke, M.J., Yohe, S., Cao, Q.,
shop at the 2001 Tandem Meetings of the Inter- Sandhu, K., Singleton, T.P., Brunstein, C.G.,
Antin, J.H., Childs, R., Filipovich, A.H., Giralt, S., national Bone Marrow Transplant Registry and Wagner, J.E., Verneris, M.R. & Weisdorf, D.J.
Mackinnon, S., Spitzer, T. & Weisdorf, D. (2001) the American Society of Blood and Marrow (2012) Unrelated cord blood transplantation in
Establishment of complete and mixed donor Transplantation. Biology of Blood and Marrow adult and pediatric acute lymphoblastic
chimerism after allogeneic lymphohematopoietic Transplantation, 7, 473–485. leukemia: effect of minimal residual disease on

156 ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 147–161
Review

relapse and survival. Biology of Blood and Mar- Baruchel, A., Bassan, R., Benoit, Y., Biondi, A., Coustan-Smith, E., Sancho, J., Hancock, M.L.,
row Transplantation, 18, 963–968. Cave, H., Dombret, H., Fielding, A.K., Foa, R., Boyett, J.M., Behm, F.G., Raimondi, S.C., Sandl-
Bacher, U., Kern, W., Schoch, C., Schnittger, S., Gokbuget, N., Goldstone, A.H., Goulden, N., und, J.T., Rivera, G.K., Rubnitz, J.E., Ribeiro,
Hiddemann, W. & Haferlach, T. (2006) Evalua- Henze, G., Hoelzer, D., Janka-Schaub, G.E., R.C., Pui, C.H. & Campana, D. (2000) Clinical
tion of complete disease remission in acute mye- Macintyre, E.A., Pieters, R., Rambaldi, A., Riber- importance of minimal residual disease in child-
loid leukemia: a prospective study based on a, J.M., Schmiegelow, K., Spinelli, O., Stary, J., hood acute lymphoblastic leukemia. Blood, 96,
cytomorphology, interphase fluorescence in situ von Stackelberg, A., Kneba, M., Schrappe, M. & 2691–2696.
hybridization, and immunophenotyping during van Dongen, J.J. (2010) Standardized MRD Coustan-Smith, E., Ribeiro, R.C., Rubnitz, J.E.,
follow-up in patients with acute myeloid leuke- quantification in European ALL trials: proceed- Razzouk, B.I., Pui, C.H., Pounds, S., Andrean-
mia. Cancer, 106, 839–847. ings of the Second International Symposium on sky, M., Behm, F.G., Raimondi, S.C., Shurtleff,
Bacher, U., Badbaran, A., Fehse, B., Zabelina, T., MRD assessment in Kiel, Germany, 18-20 Sep- S.A., Downing, J.R. & Campana, D. (2003) Clin-
Zander, A.R. & Kroger, N. (2009) Quantitative tember 2008. Leukemia, 24, 521–535. ical significance of residual disease during treat-
monitoring of NPM1 mutations provides a valid Buccisano, F., Maurillo, L., Del Principe, M.I., Del ment in childhood acute myeloid leukemia.
minimal residual disease parameter following Poeta, G., Sconocchia, G., Lo-Coco, F., Arcese, British Journal of Haematology, 123, 243–252.
allogeneic stem cell transplantation. Experimen- W., Amadori, S. & Venditti, A. (2012) Prognos- Coustan-Smith, E., Song, G., Clark, C., Key, L.,
tal Hematology, 37, 135–142. tic and therapeutic implications of minimal Liu, P., Mehrpooya, M., Stow, P., Su, X., Shurt-
Bader, P., Hancock, J., Kreyenberg, H., Goulden, residual disease detection in acute myeloid leu- leff, S., Pui, C.H., Downing, J.R. & Campana, D.
N.J., Niethammer, D., Oakhill, A., Steward, C.G., kemia. Blood, 119, 332–341. (2011) New markers for minimal residual dis-
Handgretinger, R., Beck, J.F. & Klingebiel, T. Buckley, S.A., Appelbaum, F.R. & Walter, R.B. ease detection in acute lymphoblastic leukemia.
(2002) Minimal residual disease (MRD) status (2012) Prognostic and therapeutic implications Blood, 117, 6267–6276.
prior to allogeneic stem cell transplantation is a of minimal residual disease at the time of trans- Diez-Campelo, M., Perez-Simon, J.A., Perez, J.,
powerful predictor for post-transplant outcome plantation in acute leukemia. Bone Marrow Alcoceba, M., Richtmon, J., Vidriales, B. & San
in children with ALL. Leukemia, 16, 1668–1672. Transplantation, doi: 10.1038/bmt.2012.139 Miguel, J. (2009) Minimal residual disease mon-
Bader, P., Kreyenberg, H., Hoelle, W., Dueckers, Campana, D. (2012) Should minimal residual dis- itoring after allogeneic transplantation may help
G., Handgretinger, R., Lang, P., Kremens, B., ease monitoring in acute lymphoblastic leukemia to individualize post-transplant therapeutic
Dilloo, D., Sykora, K.W., Schrappe, M., Niemey- be standard of care? Current Hematologic Malig- strategies in acute myeloid malignancies. Ameri-
er, C., Von Stackelberg, A., Gruhn, B., Henze, nancy Reports, 7, 170–177. can Journal of Hematology, 84, 149–152.
G., Greil, J., Niethammer, D., Dietz, K., Beck, Candoni, A., Toffoletti, E., Gallina, R., Simeone, Ding, L., Ley, T.J., Larson, D.E., Miller, C.A.,
J.F. & Klingebiel, T. (2004) Increasing mixed E., Chiozzotto, M., Volpetti, S. & Fanin, R. Koboldt, D.C., Welch, J.S., Ritchey, J.K., Young,
chimerism is an important prognostic factor for (2011) Monitoring of minimal residual disease M.A., Lamprecht, T., McLellan, M.D., McMich-
unfavorable outcome in children with acute by quantitative WT1 gene expression following ael, J.F., Wallis, J.W., Lu, C., Shen, D., Harris,
lymphoblastic leukemia after allogeneic stem-cell reduced intensity conditioning allogeneic stem C.C., Dooling, D.J., Fulton, R.S., Fulton, L.L.,
transplantation: possible role for pre-emptive cell transplantation in acute myeloid leukemia. Chen, K., Schmidt, H., Kalicki-Veizer, J., Mag-
immunotherapy? Journal of Clinical Oncology, Clinical Transplantation, 25, 308–316. rini, V.J., Cook, L., McGrath, S.D., Vickery,
22, 1696–1705. Chan, W.K., Kung Sutherland, M., Li, Y., Zalevsky, T.L., Wendl, M.C., Heath, S., Watson, M.A.,
Bader, P., Kreyenberg, H., Henze, G.H., Eckert, C., J., Schell, S. & Leung, W. (2012) Antibody- Link, D.C., Tomasson, M.H., Shannon, W.D.,
Reising, M., Willasch, A., Barth, A., Borkhardt, dependent cell-mediated cytotoxicity overcomes Payton, J.E., Kulkarni, S., Westervelt, P., Walter,
A., Peters, C., Handgretinger, R., Sykora, K.W., NK cell resistance in MLL-rearranged leukemia M.J., Graubert, T.A., Mardis, E.R., Wilson, R.K.
Holter, W., Kabisch, H., Klingebiel, T. & von expressing inhibitory KIR ligands but not acti- & DiPersio, J.F. (2012) Clonal evolution in
Stackelberg, S.A. (2009) Prognostic value of vating ligands. Clinical Cancer Research, 18, relapsed acute myeloid leukaemia revealed by
minimal residual disease quantification before 6296–6305. whole-genome sequencing. Nature, 481, 506–
allogeneic stem-cell transplantation in relapsed Chang, Y.H., Connolly, J., Shimasaki, N., Mimura, 510.
childhood acute lymphoblastic leukemia: the K., Kono, K. & Campana, D. (2013) A chimeric Dombret, H., Gabert, J., Boiron, J.M., Rigal-Huguet,
ALL-REZ BFM Study Group. Journal of Clinical receptor with Nkg2d specificity enhances natural F., Blaise, D., Thomas, X., Delannoy, A., Buzyn,
Oncology, 27, 377–384. killer cell activation and killing of tumor cells. A., Bilhou-Nabera, C., Cayuela, J.M., Fenaux, P.,
Boyd, S.D., Marshall, E.L., Merker, J.D., Maniar, Cancer Research, 73, 1777–1786. Bourhis, J.H., Fegueux, N., Charrin, C., Boucheix,
J.M., Zhang, L.N., Sahaf, B., Jones, C.D., Simen, Chen, Y., Cortes, J., Estrov, Z., Faderl, S., Qiao, C., Lheritier, V., Esperou, H., MacIntyre, E.,
B.B., Hanczaruk, B., Nguyen, K.D., Nadeau, W., Abruzzo, L., Garcia-Manero, G., Pierce, S., Vernant, J.P. & Fiere, D. (2002) Outcome of
K.C., Egholm, M., Miklos, D.B., Zehnder, J.L. & Huang, X., Kebriaei, P., Kadia, T., De Lima, M., treatment in adults with Philadelphia chromo-
Fire, A.Z. (2009) Measurement and clinical Kantarjian, H. & Ravandi, F. (2011) Persistence some-positive acute lymphoblastic leukemia–
monitoring of human lymphocyte clonality by of cytogenetic abnormalities at complete remis- results of the prospective multicenter LALA-94
massively parallel VDJ pyrosequencing. Science sion after induction in patients with acute mye- trial. Blood, 100, 2357–2366.
Translational Medicine, 1, 12ra23 loid leukemia: prognostic significance and the Dominietto, A., Pozzi, S., Miglino, M., Albarracin,
Bradfield, S.M., Radich, J.P. & Loken, M.R. (2004) potential role of allogeneic stem-cell transplanta- F., Piaggio, G., Bertolotti, F., Grasso, R., Zupo, S.,
Graft-versus-leukemia effect in acute lympho- tion. Journal of Clinical Oncology, 29, 2507– Raiola, A.M., Gobbi, M., Frassoni, F. &
blastic leukemia: the importance of tumor 2513. Bacigalupo, A. (2007) Donor lymphocyte
burden and early detection. Leukemia, 18, 1156– Cooley, S., Weisdorf, D.J., Guethlein, L.A., Klein, infusions for the treatment of minimal residual
1158. J.P., Wang, T., Le, C.T., Marsh, S.G., Gera- disease in acute leukemia. Blood, 109, 5063–5064.
Brenner, M.K., Rill, D.R., Moen, R.C., Krance, ghty, D., Spellman, S., Haagenson, M.D., Lad- Elmaagacli, A.H., Beelen, D.W., Kroll, M., Trzen-
R.A., Mirro, J. Jr, Anderson, W.F. & Ihle, J.N. ner, M., Trachtenberg, E., Parham, P. & sky, S., Stein, C. & Schaefer, U.W. (1998) Detec-
(1993) Gene-marking to trace origin of relapse Miller, J.S. (2010) Donor selection for natural tion of CBFbeta/MYH11 fusion transcripts in
after autologous bone-marrow transplantation. killer cell receptor genes leads to superior sur- patients with inv(16) acute myeloid leukemia
Lancet, 341, 85–86. vival after unrelated transplantation for acute after allogeneic bone marrow or peripheral
Bruggemann, M., Schrauder, A., Raff, T., Pfeifer, myelogenous leukemia. Blood, 116, 2411– blood progenitor cell transplantation. Bone
H., Dworzak, M., Ottmann, O.G., Asnafi, V., 2419. Marrow Transplantation, 21, 159–166.

ª 2013 John Wiley & Sons Ltd 157


British Journal of Haematology, 2013, 162, 147–161
Review

Elmaagacli, A.H., Beelen, D.W., Trenschel, R. & are candidates for stem cell transplantation and Knechtli, C.J.C., Goulden, N.J., Hancock, J.P.,
Schaefer, U.W. (2000) The detection of wt-1 targeted therapies. Blood, 120, 1868–1876. Grandage, V.L.G., Harris, E.L., Garland, R.J.,
transcripts is not associated with an increased Goulden, N. & Steward, C. (2002) Clinical rele- Jones, C.G., Rowbottom, A.W., Hunt, L.P.,
leukemic relapse rate in patients with acute vance of MRD in children undergoing allogeneic Green, A.F., Clarke, E., Lankester, A.W., Cor-
leukemia after allogeneic bone marrow or stem cell transplantation for ALL. Best Practice nish, J.M., Pamphilon, D.H., Steward, C.G. &
peripheral blood stem cell transplantation. Bone & Research. Clinical Haematology, 15, 59–70. Oakhill, A. (1998a) Minimal residual disease sta-
Marrow Transplantation, 25, 91–96. Goulden, N., Oakhill, A. & Steward, C. (2001) tus before allogeneic bone marrow transplanta-
Elorza, I., Palacio, C., Dapena, J.L., Gallur, L., de Practical application of minimal residual disease tion is an important determinant of successful
Toledo, J.S. & de Heredia, C.D. (2010) Relation- assessment in childhood acute lymphoblastic outcome for children and adolescents with acute
ship between minimal residual disease measured leukaemia annotation. British Journal of Haema- lymphoblastic leukemia. Blood, 92, 4072–4079.
by multiparametric flow cytometry prior to allo- tology, 112, 275–281. Knechtli, C.J., Goulden, N.J., Hancock, J.P., Harris,
geneic hematopoietic stem cell transplantation Handgretinger, R., Zugmaier, G., Henze, G., E.L., Garland, R.J., Jones, C.G., Grandage, V.L.,
and outcome in children with acute lymphoblas- Kreyenberg, H., Lang, P. & von Stackelberg, Rowbottom, A.W., Green, A.F., Clarke, E.,
tic leukemia. Haematologica, 95, 936–941. A. (2011) Complete remission after blinatumo- Lankester, A.W., Potter, M.N., Cornish, J.M.,
Faham, M., Zheng, J., Moorhead, M., Carlton, mab-induced donor T-cell activation in three Pamphilon, D.H., Steward, C.G. & Oakhill, A.
V.E., Stow, P., Coustan-Smith, E., Pui, C.H. & pediatric patients with post-transplant relapsed (1998b) Minimal residual disease status as a pre-
Campana, D. (2012) Deep-sequencing approach acute lymphoblastic leukemia. Leukemia, 25, dictor of relapse after allogeneic bone marrow
for minimal residual disease detection in acute 181–184. transplantation for children with acute lympho-
lymphoblastic leukemia. Blood, 120, 5173–5180. Harris, J. & Freireich, E.J. (1970) In vitro growth blastic leukaemia. British Journal of Haematology,
Feller, N., Jansen-van der Weide, M.C., van der of myeloid colonies from bone marrow of 102, 860–871.
Pol, M.A., Westra, G.A., Ossenkoppele, G.J. & patients with acute leukemia in remission. Blood, Kolb, H.J., Schattenberg, A., Goldman, J.M., Her-
Schuurhuis, G.J. (2005) Purging of peripheral 35, 61–63. tenstein, B., Jacobsen, N., Arcese, W., Ljungman,
blood stem cell transplants in AML: a predictive Imai, C., Iwamoto, S. & Campana, D. (2005) P., Ferrant, A., Verdonck, L. & Niederwieser, D.
model based on minimal residual disease Genetic modification of primary natural killer (1995) Graft-versus-leukemia effect of donor
burden. Experimental Hematology, 33, 120–130. cells overcomes inhibitory signals and induces lymphocyte transfusions in marrow grafted
Formankova, R., Sedlacek, P., Krskova, L., Rihova, specific killing of leukemic cells. Blood, 106, patients. European Group for Blood and Mar-
H., Sramkova, L. & Star, J. (2003) Chimerism- 376–383. row Transplantation Working Party Chronic
directed adoptive immunotherapy in prevention Inaba, H., Coustan-Smith, E., Cao, X., Pounds, Leukemia. Blood, 86, 2041–2050.
and treatment of post-transplant relapse of S.B., Shurtleff, S.A., Wang, K.Y., Raimondi, S.C., Krejci, O., van der Velden, V., Bader, P., Kreyen-
leukemia in childhood. Haematologica, 88, 117– Onciu, M., Jacobsen, J., Ribeiro, R.C., Dahl, berg, H., Goulden, N., Hancock, J., Schilham,
118. G.V., Bowman, W.P., Taub, J.W., Degar, B., M.W., Lankester, A., Revesz, T., Klingebiel, T. &
Freireich, E.J., Cork, A., Stass, S.A., McCredie, Leung, W., Downing, J.R., Pui, C.H., Rubnitz, van Dongen, J.J. (2003) Level of minimal resid-
K.B., Keating, M.J., Estey, E.H., Kantarjian, J.E. & Campana, D. (2012a) Comparative analy- ual disease prior to haematopoietic stem cell
H.M. & Trujillo, J.M. (1992) Cytogenetics for sis of different approaches to measure treatment transplantation predicts prognosis in paediatric
detection of minimal residual disease in acute response in acute myeloid leukemia. Journal of patients with acute lymphoblastic leukaemia: a
myeloblastic leukemia. Leukemia, 6, 500–506. Clinical Oncology, 30, 3625–3632. report of the Pre-BMT MRD Study Group. Bone
Fujisaki, H., Kakuda, H., Shimasaki, N., Imai, C., Inaba, H., Bhojwani, D., Pauley, J.L., Pei, D., Marrow Transplantation, 32, 849–851.
Ma, J., Lockey, T., Eldridge, P., Leung, W.H. & Cheng, C., Metzger, M.L., Howard, S.C., Rub- Kwon, M., Martinez-Laperche, C., Infante, M., Car-
Campana, D. (2009) Expansion of highly cyto- nitz, J.E., Sandlund, J.T., Ribeiro, R.C., Leung, retero, F., Balsalobre, P., Serrano, D., Gayoso, J.,
toxic human natural killer cells for cancer cell W., Campana, D., Pui, C.H. & Jeha, S. (2012b) Perez-Corral, A., Anguita, J., Diez-Martin, J.L. &
therapy. Cancer Research, 69, 4010–4017. Combination chemotherapy with clofarabine, Buno, I. (2012) Evaluation of minimal residual
Giebel, S., Locatelli, F., Lamparelli, T., Velardi, A., cyclophosphamide, and etoposide in children disease by real-time quantitative PCR of Wilms’
Davies, S., Frumento, G., Maccario, R., Bonetti, with refractory or relapsed haematological tumor 1 expression in patients with acute
F., Wojnar, J., Martinetti, M., Frassoni, F., malignancies. British Journal of Haematology, myelogenous leukemia after allogeneic stem cell
Giorgiani, G., Bacigalupo, A. & Holowiecki, J. 156, 275–279. transplantation: correlation with flow cytometry
(2003) Survival advantage with KIR ligand Jacobsohn, D.A., Tse, W.T., Chaleff, S., Rade- and chimerism. Biology of Blood and Marrow
incompatibility in hematopoietic stem cell trans- maker, A., Duerst, R., Olszewski, M., Huang, Transplantation, 18, 1235–1242.
plantation from unrelated donors. Blood, 102, W., Chou, P.M. & Kletzel, M. (2009) High WT1 Laane, E., Derolf, A.R., Bjorklund, E., Mazur, J.,
814–819. gene expression before haematopoietic stem cell Everaus, H., Soderhall, S., Bjorkholm, M. &
Giebel, S., Stella-Holowiecka, B., Krawczyk-Kulis, transplant in children with acute myeloid leu- Porwit-MacDonald, A. (2006) The effect of
M., Gokbuget, N., Hoelzer, D., Doubek, M., kaemia predicts poor event-free survival. British allogeneic stem cell transplantation on outcome
Mayer, J., Piatkowska-Jakubas, B., Skotnicki, Journal of Haematology, 146, 669–674. in younger acute myeloid leukemia patients with
A.B., Dombret, H., Ribera, J.M., Piccaluga, P.P., Kanamori, H., Sasaki, S., Yamazaki, E., Ueda, S., minimal residual disease detected by flow
Czerw, T., Kyrcz-Krzemien, S. & Holowiecki, J. Hattori, M., Fukawa, H., Tamura, T., Harano, cytometry at the end of post-remission chemo-
(2010) Status of minimal residual disease deter- H., Matsuzaki, M., Ogawa, K., Mohri, H. & Ok- therapy. Haematologica, 91, 833–836.
mines outcome of autologous hematopoietic ubo, T. (1997) Eradication of minimal residual Langerak, A.W., Groenen, P.J., Bruggemann, M.,
SCT in adult ALL. Bone Marrow Transplanta- disease during graft-versus-host reaction Beldjord, K., Bellan, C., Bonello, L., Boone, E.,
tion, 45, 1095–1101. induced by abrupt discontinuation of immuno- Carter, G.I., Catherwood, M., Davi, F., Delfau-
Gokbuget, N., Kneba, M., Raff, T., Trautmann, H., suppression following bone marrow transplanta- Larue, M.H., Diss, T., Evans, P.A., Gameiro, P.,
Bartram, C.R., Arnold, R., Fietkau, R., Freund, tion in a patient with Ph1-ALL. Transplant Garcia Sanz, R., Gonzalez, D., Grand, D.,
M., Ganser, A., Ludwig, W.D., Maschmeyer, G., International, 10, 328–330. Hakansson, A., Hummel, M., Liu, H., Lombardia,
Rieder, H., Schwartz, S., Serve, H., Thiel, E., Kern, S.E. (2012) Why your new cancer biomarker L., Macintyre, E.A., Milner, B.J., Montes-Moreno,
Bruggemann, M. & Hoelzer, D. (2012) Adult may never work: recurrent patterns and remark- S., Schuuring, E., Spaargaren, M., Hodges, E. &
patients with acute lymphoblastic leukemia and able diversity in biomarker failures. Cancer van Dongen, J.J. (2012) EuroClonality/BIOMED-
molecular failure display a poor prognosis and Research, 72, 6097–6101. 2 guidelines for interpretation and reporting of

158 ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 147–161
Review

Ig/TCR clonality testing in suspected lymphopro- DiPersio, J.F., Mardis, E.R. & Wilson, R.K. tic value of reclassification of leukemia accord-
liferations. Leukemia, 26, 2159–2171. (2010) DNMT3A mutations in acute myeloid ing to in vitro growth characteristics. Blood, 44,
Lankester, A.C., Bierings, M.B., Van Wering, E.R., leukemia. New England Journal of Medicine, 363, 1–18.
Wijkhuijs, A.J., de Weger, R.A., Wijnen, J.T., 2424–2433. Mortuza, F.Y., Papaioannou, M., Moreira, I.M.,
Vossen, J.M., Versluys, B., Egeler, R.M., van Tol, Mathe, G., Schwarzenberg, L., Mery, A.M., Cattan, Coyle, L.A., Gameiro, P., Gandini, D., Prentice,
M.J., Putter, H., Revesz, T., van Dongen, J.J., A., Schneider, M., Amiel, J.L., Schlumberger, H.G., Goldstone, A., Hoffbrand, A.V. & Foroni,
van der Velden, V.H. & Schilham, M.W. (2010) J.R., Poisson, J. & Wajcner, G. (1966) Extensive L. (2002) Minimal residual disease tests provide
Preemptive alloimmune intervention in high- histological and cytological survey of patients an independent predictor of clinical outcome in
risk pediatric acute lymphoblastic leukemia with acute leukaemia in “complete remission”. adult acute lymphoblastic leukemia. Journal of
patients guided by minimal residual disease level British Medical Journal, 5488, 640–642. Clinical Oncology, 20, 1094–1104.
before stem cell transplantation. Leukemia, 24, Maurillo, L., Buccisano, F., Del Principe, M.I., Del, Ogawa, H., Tamaki, H., Ikegame, K., Soma, T.,
1462–1469. P.G., Spagnoli, A., Panetta, P., Ammatuna, E., Kawakami, M., Tsuboi, A., Kim, E.H., Hosen,
Lapteva, N., Durett, A.G., Sun, J., Rollins, L.A., Neri, B., Ottaviani, L., Sarlo, C., Venditti, D., N., Murakami, M., Fujioka, T., Masuda, T.,
Huye, L.L., Fang, J., Dandekar, V., Mei, Z., Jack- Quaresima, M., Cerretti, R., Rizzo, M., De Fab- Taniguchi, Y., Nishida, S., Oji, Y., Oka, Y. &
son, K., Vera, J., Ando, J., Ngo, M.C., Coustan- ritiis, F.P., Lo Coco, F., Arcese, W., Amadori, S. Sugiyama, H. (2003) The usefulness of monitor-
Smith, E., Campana, D., Szmania, S., Garg, T., & Venditti, A. (2008) Toward optimization of ing WT1 gene transcripts for the prediction and
Moreno-Bost, A., Vanrhee, F., Gee, A.P. & postremission therapy for residual disease-posi- management of relapse following allogeneic stem
Rooney, C.M. (2012) Large-scale ex vivo expan- tive patients with acute myeloid leukemia. Jour- cell transplantation in acute type leukemia.
sion and characterization of natural killer cells nal of Clinical Oncology, 26, 4944–4951. Blood, 101, 1698–1704.
for clinical applications. Cytotherapy, 14, 1131– Mertelsmann, R., Moore, M.A., Broxmeyer, H.E., Ommen, H.B., Schnittger, S., Jovanovic, J.V.,
1143. Cirrincione, C. & Clarkson, B. (1981) Diagnostic Ommen, I.B., Hasle, H., Ostergaard, M.,
Lee, S., Kim, D.W., Cho, B.S., Yoon, J.H., Shin, and prognostic significance of the CFU-c assay Grimwade, D. & Hokland, P. (2010) Strikingly
S.H., Yahng, S.A., Lee, S.E., Eom, K.S., Kim, in acute nonlymphoblastic leukemia. Cancer different molecular relapse kinetics in NPM1c,
Y.J., Chung, N.G., Kim, H.J., Min, C.K., Lee, Research, 41, 4844–4848. PML-RARA, RUNX1-RUNX1T1, and CBFB-
J.W., Min, W.S. & Park, C.W. (2012) Impact of Miglino, M., Berisso, G., Grasso, R., Canepa, L., MYH11 acute myeloid leukemias. Blood, 115,
minimal residual disease kinetics during imati- Clavio, M., Pierri, I., Pietrasanta, D., Gatto, S., 198–205.
nib-based treatment on transplantation outcome Varaldo, R., Ballerini, F., Verdiani, S., Casarino, Osborne, D., Frost, L., Tobal, K. & Liu Yin, J.A.
in Philadelphia chromosome-positive acute lym- L., DeStefano, F., Sessarego, M., Dominietto, A., (2005) Elevated levels of WT1 transcripts in
phoblastic leukemia. Leukemia, 26, 2367–2374. Raiola, A.M., Bregante, S., di Grazia, C., Gobbi, bone marrow harvests are associated with a high
Leung, W., Iyengar, R., Turner, V., Lang, P., Bad- M. & Bacigalupo, A. (2002) Allogeneic bone relapse risk in patients autografted for acute
er, P., Conn, P., Niethammer, D. & Handgretin- marrow transplantation (BMT) for adults with myeloid leukaemia. Bone Marrow Transplanta-
ger, R. (2004) Determinants of antileukemia acute lymphoblastic leukemia (ALL): predictive tion, 36, 67–70.
effects of allogeneic NK cells. Journal of Immu- role of minimal residual disease monitoring on Paganin, M., Zecca, M., Fabbri, G., Polato, K., Bi-
nology, 172, 644–650. relapse. Bone Marrow Transplantation, 30, 579– ondi, A., Rizzari, C., Locatelli, F. & Basso, G.
Leung, W., Campana, D., Yang, J., Pei, D., Cou- 585. (2008) Minimal residual disease is an important
stan-Smith, E., Gan, K., Rubnitz, J.E., Sandlund, Miller, J.S., Soignier, Y., Panoskaltsis-Mortari, A., predictive factor of outcome in children with
J.T., Ribeiro, R.C., Srinivasan, A., Hartford, C., McNearney, S.A., Yun, G.H., Fautsch, S.K., relapsed ‘high-risk’ acute lymphoblastic leuke-
Triplett, B.M., Dallas, M., Pillai, A., Handgretin- McKenna, D., Le, C., Defor, T.E., Burns, L.J., mia. Leukemia, 22, 2193–2200.
ger, R., Laver, J.H. & Pui, C.H. (2011) High Orchard, P.J., Blazar, B.R., Wagner, J.E., Slung- Patel, B., Rai, L., Buck, G., Richards, S.M., Mortu-
success rate of hematopoietic cell transplantation aard, A., Weisdorf, D.J., Okazaki, I.J. & McGl- za, Y., Mitchell, W., Gerrard, G., Moorman,
regardless of donor source in children with very ave, P.B. (2005) Successful adoptive transfer and A.V., Duke, V., Hoffbrand, A.V., Fielding, A.K.,
high-risk leukemia. Blood, 118, 223–230. in vivo expansion of human haploidentical NK Goldstone, A.H. & Foroni, L. (2010) Minimal
Leung, W., Pui, C.H., Coustan-Smith, E., Yang, J., cells in cancer patients. Blood, 105, 3051–3057. residual disease is a significant predictor of
Pei, D., Gan, K., Srinivasan, A., Hartford, C., Miyazaki, T., Fujita, H., Fujimaki, K., Hosoyama, treatment failure in non T-lineage adult acute
Triplett, B.M., Dallas, M., Pillai, A., Shook, D., T., Watanabe, R., Tachibana, T., Fujita, A., Mat- lymphoblastic leukaemia: final results of the
Rubnitz, J.E., Sandlund, J.T., Jeha, S., Inaba, H., sumoto, K., Tanaka, M., Koharazawa, H., Tagu- international trial UKALL XII/ECOG2993. Brit-
Ribeiro, R.C., Handgretinger, R., Laver, J.H. & chi, J., Tomita, N., Sakai, R., Fujisawa, S., ish Journal of Haematology, 148, 80–89.
Campana, D. (2012) Detectable minimal resid- Kanamori, H. & Ishigatsubo, Y. (2012) Clinical Pfeifer, H., Wassmann, B., Bethge, W., Dengler, J.,
ual disease before hematopoietic cell transplan- significance of minimal residual disease detected Bornhauser, M., Stadler, M., Beelen, D., Vucinic,
tation is prognostic but does not preclude cure by multidimensional flow cytometry: serial V., Burmeister, T., Stelljes, M., Faul, C., Dreger,
for children with very-high-risk leukemia. Blood, monitoring after allogeneic stem cell transplan- P., Kiani, A., Schafer-Eckart, K., Schwerdtfeger,
120, 468–472. tation for acute leukemia. Leukemia Research, R., Lange, E., Kubuschok, B., Horst, H.A.,
Ley, T.J., Ding, L., Walter, M.J., McLellan, M.D., 36, 998–1003. Gramatzki, M., Bruck, P., Serve, H., Hoelzer, D.,
Lamprecht, T., Larson, D.E., Kandoth, C., Pay- Mizuta, S., Ito, Y., Kohno, A., Kiyoi, H., Miyam- Gokbuget, N. & Ottmann, O.G. (2012)
ton, J.E., Baty, J., Welch, J., Harris, C.C., Lichti, ura, K., Tanimoto, M., Takamatsu, J., Naoe, T., Randomized comparison of prophylactic and
C.F., Townsend, R.R., Fulton, R.S., Dooling, Morishima, Y., Ueda, R. & Saito, H. (1999) minimal residual disease-triggered imatinib after
D.J., Koboldt, D.C., Schmidt, H., Zhang, Q., Accurate quantitation of residual tumor burden allogeneic stem cell transplantation for BCR-
Osborne, J.R., Lin, L., O’Laughlin, M., McMich- at bone marrow harvest predicts timing of sub- ABL1 positive acute lymphoblastic leukemia.
ael, J.F., Delehaunty, K.D., McGrath, S.D., Ful- sequent relapse in patients with common ALL Leukemia, doi: 10.1038/leu.2012.352.
ton, L.A., Magrini, V.J., Vickery, T.L., Hundal, treated by autologous bone marrow transplanta- Platzbecker, U., Wermke, M., Radke, J., Oelschlae-
J., Cook, L.L., Conyers, J.J., Swift, G.W., Reed, tion. Nagoya BMT Group.. Bone Marrow Trans- gel, U., Seltmann, F., Kiani, A., Klut, I.M.,
J.P., Alldredge, P.A., Wylie, T., Walker, J., Kal- plantation, 24, 777–784. Knoth, H., Rollig, C., Schetelig, J., Mohr, B.,
icki, J., Watson, M.A., Heath, S., Shannon, Moore, M.A., Spitzer, G., Williams, N., Metcalf, D. Graehlert, X., Ehninger, G., Bornhauser, M. &
W.D., Varghese, N., Nagarajan, R., Westervelt, & Buckley, J. (1974) Agar culture studies in 127 Thiede, C. (2012) Azacitidine for treatment of
P., Tomasson, M.H., Link, D.C., Graubert, T.A., cases of untreated acute leukemia: the prognos- imminent relapse in MDS or AML patients after

ª 2013 John Wiley & Sons Ltd 159


British Journal of Haematology, 2013, 162, 147–161
Review

allogeneic HSCT: results of the RELAZA trial. A.M., Leung, W. & Vong, Q. (2013) Ex vivo acti- G. & Bargou, R.C. (2011) Targeted therapy with
Leukemia, 26, 381–389. vation of CD56+ immune cells that eradicate neu- the T-cell-engaging antibody blinatumomab of
Porter, D.L., Levine, B.L., Kalos, M., Bagg, A. & roblastoma. Cancer Research, 73, 2608–2618. chemotherapy-refractory minimal residual dis-
June, C.H. (2011) Chimeric antigen receptor- Sanchez, J., Serrano, J., Gomez, P., Martinez, F., ease in B-lineage acute lymphoblastic leukemia
modified T cells in chronic lymphoid leukemia. Martin, C., Madero, L., Herrera, C., Garcia, patients results in high response rate and pro-
New England Journal of Medicine, 365, 725–733. J.M., Casano, J. & Torres, A. (2002) Clinical longed leukemia-free survival. Journal of Clinical
Pui, C.H., Campana, D., Pei, D., Bowman, W.P., value of immunological monitoring of minimal Oncology, 29, 2493–2498.
Sandlund, J.T., Kaste, S.C., Ribeiro, R.C., residual disease in acute lymphoblastic leukae- Uzunel, M., Jaksch, M., Mattsson, J. & Ringden,
Rubnitz, J.E., Raimondi, S.C., Onciu, M., Cou- mia after allogeneic transplantation. British Jour- O. (2003) Minimal residual disease detection
stan-Smith, E., Kun, L.E., Jeha, S., Cheng, C., nal of Haematology, 116, 686–694. after allogeneic stem cell transplantation is
Howard, S.C., Simmons, V., Bayles, A., Metzger, Sanchez-Garcia, J., Serrano, J., Serrano-Lopez, J., correlated to relapse in patients with acute
M.L., Boyett, J.M., Leung, W., Handgretinger, Gomez-Garcia, P., Martinez, F., Garcia-Castel- lymphoblastic leukaemia. British Journal of
R., Downing, J.R., Evans, W.E. & Relling, M.V. lano, J.M., Rojas, R., Martin, C., Rodriguez- Haematology, 122, 788–794.
(2009) Treating childhood acute lymphoblastic Villa, A., Molina-Hurtado, J.R., Alvarez, M.A., van der Velden, V., Joosten, S.A., Willemse, M.J.,
leukemia without cranial irradiation. New Eng- Casano, J. & Torres-Gomez, A. (2012) Quantifi- Van Wering, E.R., Lankester, A.W., van Dongen,
land Journal of Medicine, 360, 2730–2741. cation of minimal residual disease levels by flow J.J. & Hoogerbrugge, P.M. (2001) Real-time
Radich, J., Ladne, P. & Gooley, T. (1995) Polymer- cytometry at time of transplant predicts out- quantitative PCR for detection of minimal resid-
ase chain reaction-based detection of minimal come after myeloablative allogeneic transplanta- ual disease before allogeneic stem cell transplan-
residual disease in acute lymphoblastic leukemia tion in ALL. Bone Marrow Transplantation, 48, tation predicts outcome in children with acute
predicts relapse after allogeneic BMT. Biology of 396–402. lymphoblastic leukemia. Leukemia, 15, 1485–
Blood and Marrow Transplantation, 1, 24–31. Schneider, M., Hettinger, K., Matthes-Martin, S., 1487.
Rettinger, E., Willasch, A.M., Kreyenberg, H., Konrad, M., Peters, C., Gadner, H. & Panzer- van der Velden, V.H., Sluijs-Geling, A., Gibson,
Borkhardt, A., Holter, W., Kremens, B., Strahm, Grumayer, E.R. (2001) Influence of transplanta- B.E., te Marvelde, J.G., Hoogeveen, P.G., Hop,
B., Woessmann, W., Mauz-Koerholz, C., Gruhn, tion regimen on prognostic significance of W.C., Wheatley, K., Bierings, M.B., Schuurhuis,
B., Burdach, S., Albert, M.H., Schlegel, P.G., high-level minimal residual disease before G.J., de Graaf, S.S., Van Wering, E.R. & van
Klingebiel, T. & Bader, P. (2011) Preemptive allogeneic stem cell transplantation in children Dongen, J.J. (2010) Clinical significance of flow-
immunotherapy in childhood acute myeloid leu- with ALL. Bone Marrow Transplantation, 28, cytometric minimal residual disease detection in
kemia for patients showing evidence of mixed 1087–1089. pediatric acute myeloid leukemia patients trea-
chimerism after allogeneic stem cell transplanta- Shimasaki, N., Fujisaki, H., Cho, D., Masselli, M., ted according to the DCOG ANLL97/MRC
tion. Blood, 118, 5681–5688. Lockey, T., Eldridge, P., Leung, W.H. & Cam- AML12 protocol. Leukemia, 24, 1599–1606.
Rubnitz, J.E., Inaba, H., Dahl, G.V., Ribeiro, R.C., pana, D. (2012) A clinically adaptable method Venditti, A., Maurillo, L., Buccisano, F., Del Poeta,
Bowman, W.P., Taub, J.W., Pounds, S., Raz- to enhance the cytotoxicity of natural killer cells G., Mazzone, C., Tamburini, A., Del Principe,
zouk, B.I., Lacayo, N., Cao, X., Meshinchi, S., against B-cell malignancies. Cytotherapy, 14, M.I., Consalvo, M.I., De Fabritiis, P., Cudillo,
Degar, B., Airewele, G., Raimondi, S.C., Onciu, 830–840. L., Picardi, A., Franchi, A., Lo-Coco, F. & Ama-
M., Coustan-Smith, E., Downing, J.R., Leung, Shook, D., Coustan-Smith, E., Ribeiro, R.C., Rub- dori, S. (2003) Pretransplant minimal residual
W.H., Pui, C.H. & Campana, D. (2010a) Mini- nitz, J.E. & Campana, D. (2009) Minimal resid- disease level predicts clinical outcome in patients
mal residual disease-directed therapy for child- ual disease quantitation in acute myeloid with acute myeloid leukemia receiving high-dose
hood acute myeloid leukemia: results of the leukemia. Clinical Lymphoma & Myeloma, 9 chemotherapy and autologous stem cell trans-
AML02 multicenter trial. The Lancet Oncology, (Suppl. 3), S281–S285. plantation. Leukemia, 17, 2178–2182.
11, 543–552. Spinelli, O., Peruta, B., Tosi, M., Guerini, V., Salvi, Walter, R.B., Gooley, T.A., Wood, B.L., Milano, F.,
Rubnitz, J.E., Inaba, H., Ribeiro, R.C., Pounds, S., A., Zanotti, M.C., Oldani, E., Grassi, A., Inter- Fang, M., Sorror, M.L., Estey, E.H., Salter, A.I.,
Rooney, B., Bell, T., Pui, C.H. & Leung, W. mesoli, T., Mico, C., Rossi, G., Fabris, P., Lamb- Lansverk, E., Chien, J.W., Gopal, A.K., Appel-
(2010b) NKAML: a pilot study to determine the ertenghi-Deliliers, G., Angelucci, E., Barbui, T., baum, F.R. & Pagel, J.M. (2011) Impact of
safety and feasibility of haploidentical natural Bassan, R. & Rambaldi, A. (2007) Clearance of pretransplantation minimal residual disease, as
killer cell transplantation in childhood acute minimal residual disease after allogeneic stem detected by multiparametric flow cytometry, on
myeloid leukemia. Journal of Clinical Oncology, cell transplantation and the prediction of the outcome of myeloablative hematopoietic cell
28, 955–959. clinical outcome of adult patients with high-risk transplantation for acute myeloid leukemia.
Ruggeri, L., Capanni, M., Urbani, E., Perruccio, K., acute lymphoblastic leukemia. Haematologica, Journal of Clinical Oncology, 29, 1190–1197.
Shlomchik, W.D., Tosti, A., Posati, S., Rogaia, 92, 612–618. Wassmann, B., Pfeifer, H., Stadler, M., Bornhauser,
D., Frassoni, F., Aversa, F., Martelli, M.F. & Sramkova, L., Muzikova, K., Fronkova, E., Krejci, M., Bug, G., Scheuring, U.J., Bruck, P., Stelljes,
Velardi, A. (2002) Effectiveness of donor natural O., Sedlacek, P., Formankova, R., Mejstrikova, M., Schwerdtfeger, R., Basara, N., Perz, J.,
killer cell alloreactivity in mismatched hemato- E., Stary, J. & Trka, J. (2007) Detectable mini- Bunjes, D., Ledderose, G., Mahlberg, R.,
poietic transplants. Science, 295, 2097–2100. mal residual disease before allogeneic hemato- Binckebanck, A., Gschaidmeier, H., Hoelzer, D.
Ruggeri, A., Michel, G., Dalle, J.H., Caniglia, M., poietic stem cell transplantation predicts & Ottmann, O.G. (2005) Early molecular
Locatelli, F., Campos, A., de Heredia, C.D., extremely poor prognosis in children with acute response to posttransplantation imatinib deter-
Mohty, M., Hurtado, J.M., Bierings, M., Bitten- lymphoblastic leukemia. Pediatric Blood & Can- mines outcome in MRD+ Philadelphia-positive
court, H., Mauad, M., Purtill, D., Cunha, R., cer, 48, 93–100. acute lymphoblastic leukemia (Ph+ ALL). Blood,
Kabbara, N., Gluckman, E., Labopin, M., Peters, Topp, M.S., Kufer, P., Gokbuget, N., Goebeler, M., 106, 458–463.
C. & Rocha, V. (2012) Impact of pretransplant Klinger, M., Neumann, S., Horst, H.A., Raff, T., Wu, D., Sherwood, A., Fromm, J.R., Winter, S.S.,
minimal residual disease after cord blood trans- Viardot, A., Schmid, M., Stelljes, M., Schaich, Dunsmore, K.P., Loh, M.L., Greisman, H.A.,
plantation for childhood acute lymphoblastic M., Degenhard, E., Kohne-Volland, R., Brugge- Sabath, D.E., Wood, B.L. & Robins, H. (2012)
leukemia in remission: an Eurocord, PDWP- mann, M., Ottmann, O., Pfeifer, H., Burmeister, High-throughput sequencing detects minimal
EBMT analysis. Leukemia, 26, 2455–2461. T., Nagorsen, D., Schmidt, M., Lutterbuese, R., residual disease in acute T lymphoblastic
Rujkijyanont, P., Chan, W.K., Eldridge, P.W., Loc- Reinhardt, C., Baeuerle, P.A., Kneba, M., Einsel- leukemia. Science Translational Medicine, 4,
key, T., Holladay, M., Rooney, B., Davidoff, e, H., Riethmuller, G., Hoelzer, D., Zugmaier, 134ra63.

160 ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 162, 147–161
Review

Yan, C.H., Liu, D.H., Liu, K.Y., Xu, L.P., Liu, Y.R., Zeiser, R., Spyridonidis, A., Wasch, R., Ihorst, G., Zhao, X.S., Liu, Y.R., Zhu, H.H., Xu, L.P.,
Chen, H., Han, W., Wang, Y., Qin, Y.Z. & Grullich, C., Bertz, H. & Finke, J. (2005) Evalua- Liu, D.H., Liu, K.Y. & Huang, X.J. (2012)
Huang, X.J. (2012) Risk stratification-directed tion of immunomodulatory treatment based on Monitoring MRD with flow cytometry: an
donor lymphocyte infusion could reduce relapse conventional and lineage-specific chimerism effective method to predict relapse for ALL
of standard-risk acute leukemia patients after analysis in patients with myeloid malignancies patients after allogeneic hematopoietic stem cell
allogeneic hematopoietic stem cell transplanta- after myeloablative allogeneic hematopoietic cell transplantation. Annals of Hematology, 91, 183–
tion. Blood, 119, 3256–3262. transplantation. Leukemia, 19, 814–821. 192.

ª 2013 John Wiley & Sons Ltd 161


British Journal of Haematology, 2013, 162, 147–161

You might also like