Vous êtes sur la page 1sur 9

state of the art review

How I determine if and when to recommend stopping tyrosine


kinase inhibitor treatment for chronic myeloid leukaemia

David M. Ross1,2,3 and Timothy P. Hughes1,2,4


1
Haematology Directorate, SA Pathology, 2School of Medicine, University of Adelaide, Adelaide, 3Flinders University and Medical
Centre, Bedford Park, and 4Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia

concept acknowledged the impossibility of proving cure of


Summary
CML, whilst also recognizing the enormous clinical success
Treatment-free remission (TFR) has recently emerged as a of a treatment (at the time, imatinib) that enables a patient
goal of treatment in chronic myeloid leukaemia (CML). to enjoy long-term freedom from symptoms and signs of the
Molecular remission is sustained in around 30% of imatinib- disease, and a normal or near-normal life expectancy, with
treated patients who stop treatment after 2 years with unde- minimal toxicity. Treatment-free remission (TFR) takes this
tectable minimal residual disease (UMRD) by conventional one step further in that patients can enjoy the benefits of dis-
real-time reverse transcription polymerase chain reaction. An ease control without the need for ongoing treatment.
additional 2030% of patients will lose UMRD, but remain Although ABL1 tyrosine kinase inhibitors (TKIs) are well-tol-
in stable major molecular remission off treatment. Most erated, most patients experience lingering mild toxicity that
patients with molecular recurrence have a significant increase can impair quality of life. Furthermore, the newer and more
in BCR-ABL1 within the first 6 months off treatment, but potent TKIs, such as nilotinib and dasatinib, may be associ-
there are also rare late relapses. As re-treatment with imati- ated with the emergence of severe adverse events, such as
nib restores control, a trial of TFR is safe so long as careful peripheral vascular events, myocardial ischaemia, pleural
molecular monitoring is provided to enable prompt re-treat- effusions and pulmonary hypertension. Lastly, the cost of
ment. The minimum eligibility criteria for a trial of TFR are lifelong TKI therapy is substantial. As CML patients now
not yet defined, but the available data support a MRD level have a near-normal life expectancy, the provision of TKI
of around a molecular response of 45 log for at least treatment for an average of 25 years is likely to cost around
2 years. Factors associated with a higher probability of TFR USD$500 000 per patient, at current prices. If patients are
include low risk Sokal score, prior interferon treatment, able to stop treatment after 510 years the potential cost sav-
longer total duration of imatinib treatment and higher num- ing is enormous. For all of these reasons there is considerable
bers of natural killer cells at the time of imatinib discontinu- interest among CML patients and their clinicians in the
ation. Preliminary data suggest that the rate of TFR in achievement of TFR.
patients treated with more potent tyrosine kinase inhibitors The first treatment to enable patients to achieve TFR was
will probably be higher. The biology that underlies TFR is an allogeneic stem cell transplantation. Patients who achieve
area of active investigation. molecular remission post-allograft, without graft-versus-host
disease and without the need for long-term immunosuppres-
Keywords: chronic myeloid leukaemia, tyrosine kinase inhib- sion, have an excellent quality of life and a very low risk of
itors, minimal residual disease, BCR-ABL1, treatment-free relapse (Radich et al, 1995; Mughal et al, 2001). Nevertheless,
remission. even this clinical state is not always equivalent to cure,
because very late relapses of CML have been observed (Nor-
kin et al, 2011). The success of allografting, and rescue of
relapse by donor lymphocyte infusion, highlights the poten-
What is treatment-free remission?
tial importance of immunological surveillance in the long-
Some years ago the late Professor John Goldman introduced term control of CML.
the concept of operational cure in chronic phase chronic Prior to the advent of imatinib the best non-transplant
myeloid leukaemia (CML) (Goldman & Gordon, 2006). This therapy for CML was interferon-a (IFN) with or without cyt-
arabine (Guilhot et al, 1997). Only around 20% of IFN-trea-
ted patients achieved a complete cytogenetic response, and a
Correspondence: Dr David M. Ross, Department of Haematology, minority of these patients also achieved undetectable mini-
Flinders University and Medical Centre, Bedford Park, SA 5042, mal residual disease (UMRD) using sensitive real-time
Australia. E-mail: david.ross@health.sa.gov.au reverse transcription polymerase chain reaction (RQ-PCR)

2014 John Wiley & Sons Ltd First published online 23 April 2014
British Journal of Haematology, 2014, 166, 311 doi:10.1111/bjh.12892
Review

for BCR-ABL1. Nevertheless, it was shown that TFR (defined significant rise in BCR-ABL1 level (later defined as 10-fold).
in that study as stable complete cytogenetic response) was TFR was sustained in 41% of patients at 2 years, with most
sustained in the majority of these highly-selected patients molecular recurrence occurring in the first 6 months.
after IFN treatment, with a median follow-up of 3 years The Australasian Leukaemia and Lymphoma Group
(Mahon et al, 2002). The mechanism of action of IFN is (ALLG) conducted the TWISTER study from August 2006 to
complex and poorly understood, but it is known that IFN August 2011 and enrolled 40 patients (Ross et al, 2013). As
has immunological effects, lending further circumstantial evi- in STIM, the patients were evenly divided between IM-only
dence to the idea that the host immune status might be and IFN-IM. Eligibility criteria and monitoring were very
important in TFR. IFN treatment is associated with the similar to STIM, but the molecular recurrence definition was
emergence of cytotoxic T lymphocytes (CTLs) that are reac- more stringent, with any two consecutive positive RQ-PCR
tive against myeloid-associated antigens, and CTLs are asso- results triggering imatinib re-treatment, even if there was no
ciated with TFR after IFN treatment (Molldrem et al, 2000; rise in BCR-ABL1 from the first to the second test. The esti-
Burchert et al, 2003; Kanodia et al, 2010). The combination mated rate of TFR at 2 years was 47%.
of IFN and imatinib may have additive or synergistic effects A French follow-up study, According to STIM (A-STIM)
(Preudhomme et al, 2010). enrolled an additional 80 similar patients, completing accrual
In 2007 the first report of TFR after imatinib cessation in June 2012 (Rousselot et al, 2014). A novel element of this
raised the possibility that prior IFN exposure was associated study was that it tested the safety of less stringent molecular
with a higher probability of TFR, with neither of the two response inclusion criteria (occasional low level BCR-ABL1
patients treated with imatinib de novo (IM-only) remaining was allowed in the preceding 2 years) and a less stringent
in UMRD after the drug was stopped, versus six out of 10 molecular recurrence definition, loss of major molecular
patients treated initially with IFN, then switched to imatinib response (MMR), i.e. BCR-ABL1 >01%. This enabled the
when it became available (IFN-IM) (Rousselot et al, 2007). TFR rate to be described for a similar population using
The first case of successful TFR after imatinib treatment recurrence definitions of varying stringency. Using the STIM
alone was reported the following year (Verma et al, 2008) definition, the rate of TFR at 2 years was 46%, while using
Subsequently, four prospective clinical trials have reported the new definition (loss of MMR) the rate of TFR at 2 years
TFR in 3050% of patients stopping imatinib after a period was 64%.
of stable molecular remission (Mahon et al, 2010, 2013; Ross The most recent study from the French CML Intergroup,
et al, 2010, 2013; Rousselot et al, 2014). These trials form the STIM2, has not yet been published, but was recently pre-
basis of the currently available clinical data; the key features sented at the American Society of Hematology annual meet-
of the studies are outlined here and summarized in Table I. ing (Mahon et al, 2013). This study is important because in
Following on from their pilot study in 200405, the the current era the overwhelming majority of CML patients
French CML Intergroup conducted the STIM (STop IMati- are treated only with TKIs. STIM2 enrolled 124 IM-only
nib) study from July 2007 to December 2009 and enrolled patients between April 2011 and June 2013. Using the origi-
100 patients (Mahon et al, 2010). The key eligibility criteria nal STIM definition of molecular recurrence, 61% of patients
were UMRD for a minimum period of 2 years with an esti- were in TFR at the time of the interim analysis (median fol-
mated RQ-PCR limit of detection (sensitivity) of 5 log. low-up 12 months). Importantly, 33% of patients in TFR
Around half of the patients received IFN-IM, while the showed fluctuating low levels of BCR-ABL1 below 01%. It is
remainder received IM-only. RQ-PCR monitoring was per- not clear why the proportion of patients in STIM2 with loss
formed monthly for the first year. Molecular recurrence, the of UMRD, but stable MMR is higher than in the previous
trigger for resumption of imatinib treatment, was defined as studies, but taking this into account, the rate of TFR defined
two consecutive samples with detectable BCR-ABL1 and a by the stringent UMRD criterion is similar to that seen in

Table I. Summary of prospective trials of imatinib discontinuation.

Rate of TFR according to different recurrence definitions*

% IM- Median Median duration Median duration Loss of UMRD 10-fold Loss of
Study N only age (years) of imatinib (months) of UMRD (months) (TWISTER) rise (STIM) MMR (A-STIM)

STIM 100 51 62 50 36 (2485) NR 41 NR


TWISTER 40 48 61 70 36 (2482) 47 NR NR
A-STIM 80 46 55 79 41 (2496) NR 44 61
STIM2 124 100 61 NR NR 34 61 NR

IM, imatinib; UMRD, undetectable minimal residual disease; TFR, treatment-free remission; MMR, major molecular response; NR, not reported.
*Rates of TFR are not directly comparable between studies as estimates may refer to different time points. STIM2 is in progress and only interim
results are available.
As intermittent low positive results were allowed this is not strictly a duration of UMRD.

4 2014 John Wiley & Sons Ltd


British Journal of Haematology, 2014, 166, 311
Review

IM-only patients in TWISTER, and in a small retrospective prerequisite for TFR. A-STIM enrolled patients who had an
series from Korea, at around 30% (Yhim et al, 2012). occasional low level positive RQ-PCR test during the qualify-
ing period, and this slight relaxation of the MRD inclusion
criterion did not have a measurable effect on the TFR rate.
Which patients are eligible for a trial of TFR?
Several studies currently underway have used MR4.5 (which
We estimate that around 40% of patients treated with imati- includes both UMRD and patients with low level detectable
nib de novo will eventually be eligible for a trial of TKI dis- BCR-ABL1) as an inclusion criterion that is more easily
continuation, based on the criteria used in STIM and standardized, and the results of this next generation of TFR
TWISTER (Branford et al, 2013). Therefore, approximately studies will soon be available.
1015% of all patients starting imatinib for CML can expect, The studies outlined above have generally included only
at some time in the future, to be able to remain off therapy patients in chronic phase, with no history of BCR-ABL1
with UMRD. These percentages may change over time as the kinase domain mutations. STIM allowed the enrolment of
eligibility criteria for TKI discontinuation and the criteria for patients with accelerated phase CML, although the number
resumption of TKI therapy are refined. of such patients was not reported. A series of three patients
When the first TKI discontinuation studies were planned who stopped dasatinib in UMRD after a history of imatinib
there were no data on the safety of this approach. In order intolerance or failure (including BCR-ABL1 kinase domain
to select the patients with the most favourable prognosis the mutations) was reported (Ross et al, 2011). Two of the three
first studies of TFR included only patients with UMRD patients remained in UMRD off treatment, while the third
[approximately equivalent to a molecular response of 45 log patient resumed dasatinib after molecular recurrence, and
(MR 45) or BCR-ABL1 0003%] sustained for at least regained UMRD. Until further data are available we would
2 years. In this population (a mixture of IM-only and IFN- not recommend stopping TKIs in patients with a history of
IM) the rate of stable UMRD off treatment was approxi- advanced phase disease or kinase domain mutations, unless
mately 40%. in the context of a clinical trial.
The apparently dichotomous outcome of either early Patient age may be a factor in deciding whether or not to
recurrence or durable TFR raised the question of whether or stop TKI. Older patients may be less concerned about the
not the leukaemic clone was eradicated in some patients. potential for late toxicity from treatment, including effects
Given that the level of BCR-ABL1 may continue to fall on on fertility or teratogenicity. Younger patients may experi-
TKI treatment after achieving UMRD, the actual level of ence a greater loss of quality of life from TKI toxicity (Effic-
MRD in these patients could be anywhere from just below ace et al, 2011) and may have a greater immediate incentive
0003% to zero. Is the difference between the 40% in TFR to stop treatment. The age range of patients in TFR studies
and the 60% with molecular recurrence simply a product of reported to date is 2784 years, and age has not emerged as
the depth of MRD at the time of stopping? Two lines of evi- a factor influencing molecular recurrence (Mahon et al,
dence suggest that it is not. Kinetic studies have shown a 2010, 2013; Ross et al, 2013; Rousselot et al, 2014). It should
slow, progressive depletion of MRD during continued imati- be noted, however, that advanced age increases the Sokal risk
nib treatment (Michor et al, 2005; Roeder et al, 2006), so score (Sokal et al, 1984), which may influence recurrence risk
one would expect that a longer duration of imatinib after the (see below).
first achievement of UMRD would be associated with a lower
risk of molecular recurrence, but that has not proven to be
How should patients be monitored off TKI
the case. Secondly, highly sensitive BCR-ABL1 DNA PCR
therapy?
with a limit of detection of around 1-log below that of con-
ventional RQ-PCR detected MRD in the majority of patients The availability of a sensitive RQ-PCR assay with a rapid
in the TWISTER study, even in those patients who have turn-around time is essential for patients to safely undergo a
remained in TFR for several years (Ross et al, 2010, 2013). trial of TFR. Monitoring with karyotyping or fluorescence in
Using RQ-PCR on sorted cell populations, Rousselot et al situ hybridization is not sufficient, because most clinicians
(2014) showed that the residual BCR-ABL1 signal in deep would re-start TKI at loss of MMR. If an insensitive assay is
molecular response arises from the granulocyte lineage. used the UMRD threshold may actually be little better than
Therefore, the persistence of BCR-ABL1 positivity in patients MMR, and the risk of molecular recurrence and adverse out-
with TFR cannot be explained simply by persistence of long- comes may well be higher in patients who stop with a higher
lived lymphocytes, which was one hypothesis raised by these disease burden. Considerable effort has been expended
original findings. around the world to standardize reporting of RQ-PCR detec-
UMRD is essentially an arbitrary line in the sand. The tion limits, and to make available good quality and highly
commonly used sensitivity requirement of 45-log represents sensitive RQ-PCR tests.
an MRD threshold that is based on the technical limitations Most patients who experience molecular recurrence after
of RQ-PCR technology, not based on empirical data. TKI withdrawal do so within the first 6 months (Mahon
This raises the question of what level of MRD really is a et al, 2010; Ross et al, 2013). The median time to detectable

2014 John Wiley & Sons Ltd 5


British Journal of Haematology, 2014, 166, 311
Review

BCR-ABL1 is 34 months, and almost all patients relapsing The overwhelming majority of patients regain UMRD, typi-
early do so with exponentially rising BCR-ABL1 levels, so cally within 6 months. In a single case a patient who had
that without additional treatment loss of MMR would occur molecular recurrence after ceasing imatinib and subsequently
within 12 months of losing UMRD (Branford et al, 2012). regained MMR on imatinib abruptly developed blast crisis
Therefore, we recommend monitoring monthly for 9 months later. That patient was in the IFN-IM cohort of
12 months with a sensitive RQ-PCR assay. Less frequent A-STIM and started imatinib 7 years after her original diag-
monitoring (e.g. every 23 months) may be sufficient there- nosis of CML (Rousselot et al, 2014).
after, but any positive result should trigger repeat RQ-PCR A small number of patients in STIM and TWISTER
the following month to confirm the abnormality and to stopped imatinib for a second time after regaining UMRD,
assess the kinetics of BCR-ABL1. The latest molecular recur- and remaining on treatment in UMRD for at least
rence reported to date was at 42 months in a Japanese retro- 12 months (Legros et al, 2012; Ross et al, 2013). Interest-
spective survey of imatinib discontinuation (Takahashi et al, ingly, a second molecular recurrence was not inevitable, and
2012). The latest relapse in a prospective clinical trial was in around 25% of patients remained off treatment without loss
the TWISTER study at 27 months: that patient had two con- of MMR.
secutive BCR-ABL1 values of 0002% and the following
month again had UMRD before imatinib was re-started
What factors predict durable TFR?
(Ross et al, 2013). In A-STIM, the latest loss of MMR was at
17 months (Rousselot et al, 2014). Several themes are emerging from the data available so far
Molecular recurrence of CML after TKI withdrawal has when trying to predict TFR. Receiving more than 50 months
not been associated with kinase domain mutations. In the of imatinib therapy before the trial of TFR was one of the
TWISTER study, kinase domain mutation analysis was per- strongest predictors of relapse risk (hazard ratio 0421)
formed in all patients with detectable BCR-ABL1 and no (Mahon et al, 2010). The Sokal risk score calculated at diag-
mutations were detected (Ross et al, 2013). This is perhaps nosis has been consistently identified as prognostic (Mahon
not surprising, given that BCR-ABL1 mutant clones do not et al, 2010; Yhim et al, 2012; Ross et al, 2013). Among Sokal
have a proliferative advantage over wild-type BCR-ABL1 low risk patients, 5060% sustained TFR, whereas in high
unless the relevant TKI is present. risk patients the rate of TFR was around 1020%, suggesting
that the effect of adverse disease biology at diagnosis is not
completely overcome in TKI-responsive patients, even in this
What is failure of TFR, and how should it be
highly selected fraction of patients with the best possible
managed?
response to treatment.
The concept of failure, like the concept of an optimal The prognostic value of the initial and subsequent fall in
response, is moveable. Failure of TFR after IFN treatment the level of BCR-ABL1 in response to imatinib treatment has
was loss of complete cytogenetic response, in A-STIM it was not been examined in great detail. STIM, A-STIM and
loss of MMR, and in TWISTER it was loss of UMRD. TWISTER all enrolled a mixture of patients treated with IM-
Whether or not the less stringent recurrence definitions will only and IFN-IM, resulting in heterogeneous response kinet-
result in TFR in the longer term remains to be seen. In the ics. In the relatively small number of IM-only patients whose
shorter term it will be interesting to hear patients perspec- BCR-ABL1 kinetics could be examined there was no link
tives on whether this represents a therapeutic success. After between the time taken to achieve molecular response targets
spending years striving for a deep molecular response, and and the later probability of TFR (Mahon et al, 2010; Ross
eventual UMRD, our patients may be somewhat confused by et al, 2013; Rousselot et al, 2014).
our acceptance of MMR without therapy. Patient education A trend seen in all three studies that included IFN-IM
and counselling are an important part of the clinicians role patients was a higher rate of TFR in that cohort than in IM-
in TFR. only patients: around 50% vs. around 30%. There was a
The average doubling time of BCR-ABL1 for patients off trend toward higher TFR rate in patients with a longer dura-
TKI therapy is around 10 d, equating to an increase of tion of IFN exposure (Ross et al, 2013). A beneficial effect of
around 1-log every month (Branford et al, 2012). This dou- prior IFN treatment was also observed in a retrospective
bling time is observed in most patients who fail a trial of study from Japan (Takahashi et al, 2012). There are various
TFR. In patients with exponentially rising BCR-ABL1 one hypotheses that might account for a beneficial effect of IFN,
can expect loss of MMR to occur approximately 2 months including its pleiotropic immunological effects and stem cell
after BCR-ABL1 is first detected. We would recommend depletion (Essers et al, 2009). However, there is an inherent
re-starting TKI in any patient who shows a short doubling selection bias in the IFN-IM cohort as, historically, only a
time, rather than waiting for loss of MMR. minority of patients remained on IFN for long enough to
All of the TKI discontinuation studies to date have used benefit from switching to IM when it first became available,
molecular recurrence as a trigger to re-start treatment, and and biological low risk patients are likely to be over-repre-
in all cases patients have remained responsive to treatment. sented in this cohort. Burchert et al (2010) reported a

6 2014 John Wiley & Sons Ltd


British Journal of Haematology, 2014, 166, 311
Review

number of patients treated with the combination of imatinib that DNA PCR is much less susceptible to false positive
and pegylated IFN, following which imatinib was stopped results. There is no doubt that increased sensitivity can be
and IFN continued as maintenance treatment. Whilst this achieved by replicate PCR testing (for both RNA and DNA
is a slightly different clinical setting it demonstrated a pro- targets), and an important development in this regard is dig-
gressive reduction in MRD level after the withdrawal of ital PCR (dPCR). In dPCR the reaction mixture is parti-
TKI, associated with the emergence of CTLs against CML- tioned into multiple tiny volumes (either chambers in a
associated antigens. microfluidic system or droplets in an emulsion). In an MRD
We started the TWISTER study with the hypothesis that setting, where the number of targets is small, each dPCR
the depth of MRD at the time of stopping imatinib would be reaction will contain a maximum of one copy of the target,
the major determinant of relapse risk. Consequently, we so every result is positive or negative (hence, digital). Stud-
developed a highly sensitive (6-log below baseline) patient- ies in CML or Ph-positive ALL have shown the feasibility of
specific assay for BCR-ABL1 DNA, with the aim of stratifying this approach for detecting low levels of BCR-ABL1, and at
patients according to depth of response and risk of relapse least one study is examining its utility in the TFR setting
(Ross et al, 2010). Unexpectedly, we found that the majority (Goh et al, 2011; Mori et al, 2013; Iacobucci et al, 2014).
of patients had detectable BCR-ABL1 DNA at the time of
stopping TKI. Consequently, the presence or absence of
What are the key biological questions in the
MRD was not informative with regard to subsequent molec-
field of TFR?
ular recurrence, although it did enable the detection of
molecular recurrence 12 months earlier than RQ-PCR. We consider TFR to be a distinct biological state that can be
Technological advances in PCR continuously re-define achieved in a subset of patients with deep molecular
what can be achieved in MRD analysis. The difficulty of response. The pathobiology of TFR remains to be defined,
detecting BCR-ABL1 breakpoints is no longer a major obsta- but the two most promising fields of investigation are the
cle with several methods now available (based on multiplex properties of the CML clone, and the nature of the host
PCR or massively parallel paired-ends sequencing) (Bartley immune response. Figure 1 summarizes strategies that could
et al, 2010; Score et al, 2010; Alikian et al, 2013). For a long be used to maximize the numbers of CML patients who
time there has been speculation that CML cells in MRD may eventually achieve TFR.
be quiescent and have reduced expression of BCR-ABL1, and The main evidence that we have at present in relation to
this could give a potential advantage to DNA PCR (Kumari how the properties of the CML clone influence the likelihood
et al, 2012). However, there are conflicting data on this of TFR is the association with Sokal score. Sokal, and other
topic: BCR-ABL1 mRNA is enriched in the granulocyte com- clinical risk scores, must have a biological basis. Gene expres-
partment of CML patients with UMRD (Rousselot et al, sion studies have shown alterations in a number of genes to be
2014), and primitive stem cells isolated in vitro still express associated with Sokal risk score (Schmidt et al, 2001; Flamant
BCR-ABL1 transcripts (Copland et al, 2006). In our opinion, et al, 2010), but the biological basis of high-risk disease has
the major advantage of BCR-ABL1 DNA is its specificity, so not yet been characterized in detail. The study of CML

3. New strategies
Failure of for second attempt
TFR at TFR

More potent TKIs


Combination of TKI
with drugs targetting
1. Induction of 2. Consolidation immune responses or
molecular of molecular CML stem cells
remission remission

More potent TKIs Increased


duration of TKI
Combination of
prior to stopping
TKI with other Successful 3. Long-term
drugs (e.g. IFN) Development of TFR molecular
targetting immune biomarkers for monitoring
responses or CML recurrence risk
stem cells

Fig 1. Possible strategies to maximize the number of patients in TFR. CML, chronic myeloid leukaemia; TFR, treatment-free remission;
TKI, tyrosine kinase inhibitor; IFN, interferon-a.

2014 John Wiley & Sons Ltd 7


British Journal of Haematology, 2014, 166, 311
Review

disease-specific properties in TFR patients is hampered by two lost MR4.5, but only one patient lost MMR (Benjamini et al,
practical problems. Firstly, the median time from diagnosis to 2013).
TKI discontinuation is usually >5 years, so cryopreserved cells What is the minimum duration of MR4.5 (or UMRD) before
from diagnosis are typically not available for study; and sec- attempting TFR? The retrospective series from Japan reported
ondly, the number of persistent leukaemic cells at the time of on the outcome of 43 patients who stopped imatinib with
stopping TKI treatment is too small to permit isolation of suf- UMRD and remained off treatment for at least 6 months
ficient numbers for study. Clinical trials in which patients are (Takahashi et al, 2012). The median duration of UMRD prior
treated de novo, and then have the option of proceeding to to imatinib cessation in patients who sustained TFR was
TFR will represent the best opportunity to provide detailed 325 months vs. 6 months in patients with a molecular recur-
information on how the properties of the original CML clone rence. The estimated rate of TFR at 2 years was almost 80% in
influence TFR outcome. those with at least 2 years of UMRD prior to TKI discontinua-
At present the only reported potential biomarkers for suc- tion versus 30% if the period of UMRD was shorter. These
cessful TFR are related to immunological status. Immunolog- figures are not directly comparable with those from the pro-
ical anergy is a feature of CML, affecting both T cells and spective studies, because the requirement for patients to
natural killer (NK) cell activity (Chen et al, 2008, 2012; remain off TKI for at least 6 months introduces a bias against
Mumprecht et al, 2009). One study found increased numbers patients with early molecular recurrence. A report from the
of IFN-c-producing NK cells and decreased numbers of nave MD Anderson Cancer Center found that patients with UMRD
T cells (CD8+ CD62L+) in patients with TFR after imatinib for more than 5 years at the time of TKI discontinuation had a
when compared with patients with UMRD remaining on higher rate of TFR (Benjamini et al, 2013).
imatinib treatment (Ohyashiki et al, 2012). As patients in What duration of BCR-ABL1 monitoring is required in
TFR were not studied before imatinib was withdrawn it is CML patients in TFR? It should be emphasized that the
unknown whether this phenotype has any prognostic value, number of patients with long-term follow-up remains very
but follow-up studies are warranted. Rea et al (2013) pre- small, and there is still (at least theoretically) a risk of molec-
sented the results of immunological studies in a subset of 51 ular recurrence even after five or more years. Although most
patients enrolled in the STIM study. Average NK cell num- molecular recurrence occurs in the first 6 months, the num-
bers in the peripheral blood at the time of stopping imatinib ber of instances of loss of UMRD in the second and third
were higher in patients who sustained TFR than in those years is not trivial. The frequency of molecular monitoring
with molecular recurrence. The difference was primarily due may be reduced after the second year of TFR, but we would
to the differentiated CD56-dim compartment that has the recommend indefinite RQ-PCR testing at least every
greatest cytotoxic activity, and accounts for most of the NK 3 months. This is important both to establish the long-term
cells in the blood of normal individuals. Interestingly, there durability of TFR, and to safeguard each individual patient
was no increase in NK cell numbers when patients were against the delayed diagnosis of relapse. In our practice the
taken off imatinib, arguing that this immunological defect is cost of an RQ-PCR test is equivalent to around 1 week of
not simply a toxicity of the TKI, but is related in some way imatinib treatment, so RQ-PCR monitoring of TFR remains
to the disease or to constitutional factors. There was consid- highly cost-effective.
erable overlap between NK cell numbers in TFR patients and Perhaps the most pressing question is whether there is a
in patients destined for recurrence. More detailed immuno- difference in the rate of TFR after dasatinib or nilotinib treat-
logical profiling might help to improve risk stratification. ment. Early response data from the pivotal up-front studies
indicate that these more potent TKIs will increase the pro-
portion of patients who meet the eligibility criteria for a trial
What are the key unanswered clinical
of TFR (Kantarjian et al, 2012; Larson et al, 2012). The
questions in the field of TFR?
ENESTcmr study also showed that switching patients from
Many important clinical questions remain regarding the selec- imatinib to nilotinib could increase the rate of MR4.5 (Leber
tion of patients for a trial of TFR. What is the optimum level of et al, 2013). However, the achievement of deep molecular
MRD at which a trial of TFR should be entertained? Is there a response is necessary for TFR, but not sufficient, and it
continuous gradient with higher TFR rate as the level of MRD remains to be seen what rate of TFR is achieved in those
falls, or is there a critical threshold below which there is no fur- patients. Stopping studies in patients treated with nilotinib
ther improvement in the probability of TFR? In A-STIM the first-line or after imatinib (ENESTfreedom, ENESTop) and
inclusion criteria were relaxed to allow patients with intermit- dasatinib (DASFREE) are underway, but the results from
tent low-level positive RQ-PCR tests during the 2 years prior these large international studies are not yet available. A
to stopping imatinib. The rate of loss of UMRD (STIM defini- French study of stopping second-generation (2G) TKIs
tion) was higher in these patients, but there was no difference reported on the outcome of 34 patients with at least
in the rate of loss of MMR (Rousselot et al, 2014). A single 6 months of follow-up (Rea et al, 2012). TFR (defined as
centre study reported on the outcome of five patients who persistent MMR off treatment) was observed in 58% of
stopped TKI with MR4.5 but detectable MRD: four patients patients, or 44% using the stringent TWISTER criterion.

8 2014 John Wiley & Sons Ltd


British Journal of Haematology, 2014, 166, 311
Review

Around half of the patients switched from imatinib to a 2G known about prediction of molecular recurrence off therapy
TKI due to intolerance, and around half for suboptimal has been summarized. Among patients with stable MR4.5 or
response. The interim presentation of a Japanese dasatinib UMRD for at least 2 years there is no identified subgroup of
discontinuation study reported TFR (stable UMRD) in 44% patients with a TFR rate below 10%. Factors that may drive an
of 63 patients with UMRD after second-line dasatinib attempt at TKI discontinuation include: (i) patient preference;
(Tanaka et al, 2013). Based on these early data it seems that (ii) significant TKI toxicity; (iii) cost or (iv) planned preg-
the rate of TFR after 2G TKIs may be similar to the rate seen nancy. In a patient who has a high likelihood of molecular
after imatinib or slightly better, but if more patients achieve recurrence (e.g. high risk Sokal) a trial of TFR may not be jus-
a deep molecular response, the absolute number of patients tified unless there is a strong motivation for the patient to dis-
who achieve TFR could be substantially higher than in continue therapy. Although the clinical risks associated with
IM-only patients. short-term molecular recurrence are minor, an unsuccessful
Is there a novel treatment that could be added to a TKI to attempt at TKI discontinuation uses additional monitoring
increase the probability of TFR in patients who already have a resources, and may cause unnecessary anxiety.
deep molecular response, either before stopping TKI or as a
second-line treatment after an initial failure of TKI? There
Concluding remarks
are several candidate drugs in this area: e.g. immunomodula-
tion with IFN or ipilimumab (Bashey et al, 2009), stem cell As TFR has become a possibility for many CML patients the
depletion with IFN or inhibitors of the Hedgehog pathway goal of treatment is expanding beyond simply preventing dis-
(Zhang et al, 2010), or targeting of alternative signalling ease progression to achieving the deepest possible molecular
pathways, such as Janus kinase (JAK) or phosphatidylinosi- response in order to enable the patient to be free of treat-
tide 3-kinase (PI3K) (Ding et al, 2013; Warsch et al, 2013). ment and free of disease. This trend has parallels in other
Given that patients with a deep molecular response on TKIs haematological neoplasms (e.g. limited stage Hodgkin lym-
generally have good quality of life and a normal life expec- phoma or paediatric acute lymphoblastic leukaemia) that
tancy the potential toxicity of any new therapy must be care- respond so well to treatment that recent clinical trials have
fully considered. focussed on the de-escalation of therapy to try to minimize
the late toxicities of treatment without compromising effi-
cacy. With this new aim come new risks: the single case of
Stopping TKIs outside the setting of a clinical
blast crisis after a trial of TFR reminds us that careful moni-
trial
toring is essential. At least for now, most attempts at TKI
Wherever possible, patients wishing to discontinue TKI treat- discontinuation should, if possible, be undertaken in the set-
ment should do so in the structured setting of a clinical trial. ting of a clinical trial. Just as it seemed that all the major
This provides a degree of protection to the patient and adds questions in CML treatment were close to being answered a
to the body of scientific knowledge available. In cases where whole new area of investigation has emerged.
no such opportunity exists, a patient may still wish to dis-
continue treatment. We consider that this can safely be
Author contributions
undertaken under the supervision of an experienced clini-
cian, so long as high quality RQ-PCR test results are avail- DMR and TPH reviewed the literature and wrote the paper.
able within a reasonable length of time. The patient must
understand the need for more intensive follow-up with
Disclosures
molecular testing, and be prepared to comply with the fol-
low-up plan. Inadequate follow-up testing could expose the DMR and TPH were the principal investigators in the TWIS-
patient to unnecessary risk. TER study. DMR has received research funding and honor-
For an individual patient the decision whether or not to dis- aria from Novartis Pharmaceuticals. TPH has received
continue TKI is dependent on the motivation to stop treat- research funding and honoraria from Novartis, Bristol-Myers
ment and the estimated risk of molecular recurrence. What is Squibb, and Ariad.

References Bartley, P.A., Martin-Harris, M.H., Budgen, B.J., CTLA4 blockade with ipilimumab to treat
Alikian, M., Ellery, P., Forbes, M., Gerrard, G., Ross, D.M. & Morley, A.A. (2010) Rapid isola- relapse of malignancy after allogeneic hemato-
Aitman, T., Kasperaviciute, D., Milojkovic, D., tion of translocation breakpoints in chronic poietic cell transplantation. Blood, 113, 1581
Marin, D., Goldman, J.M., Apperley, J.F., myeloid and acute promyelocytic leukaemia. 1588.
Foroni, L. & Reid, A. (2013) NGS-assisted British Journal of Haematology, 149, 231236. Benjamini, O., Kantarjian, H., Rios, M.B., Jabbour,
DNA-based digital qPCR facilitates stratification Bashey, A., Medina, B., Corringham, S., Pasek, M., E., OBrien, S., Jain, P., Cardenas-Turanzas, M.,
of CML patients in long-term molecular remis- Carrier, E., Vrooman, L., Lowy, I., Solomon, Faderl, S., Garcia-Manero, G., Ravandi, F.,
sion based on the presence of detectable BCR- S.R., Morris, L.E., Holland, H.K., Mason, J.R., Borthakur, G., Quintas-Cardama, A. & Cortes, J.
ABL1 DNA. Blood, 122, Abstract 4006. Alyea, E.P., Soiffer, R.J. & Ball, E.D. (2009) (2013) Patient-driven discontinuation of

2014 John Wiley & Sons Ltd 9


British Journal of Haematology, 2014, 166, 311
Review

tyrosine kinase inhibitors: single institution to, L., Ulisciani, S., Veneri, D., Sica, S., Rambal- sion levels in hematopoietic precursor cells
experience. Leukaemia & Lymphoma, doi: 10. di, A., Vignetti, M., Mandelli, F. & GIMEMA enable persistence of chronic myeloid leukemia
3109/10428194.2013.831092. (2011) Health-related quality of life in chronic under imatinib. Blood, 119, 530539.
Branford, S., Yeung, D.T., Prime, J.A., Choi, S.Y., myeloid leukemia patients receiving long-term Larson, R.A., Hochhaus, A., Hughes, T.P., Clark,
Bang, J.H., Park, J.E., Kim, D.W., Ross, D.M. & therapy with imatinib compared with the gen- R.E., Etienne, G., Kim, D.W., Flinn, I.W.,
Hughes, T.P. (2012) BCR-ABL1 doubling times eral population. Blood, 118, 45544560. Kurokawa, M., Moiraghi, B., Yu, R., Blakesley,
more reliably assess the dynamics of CML Essers, M.A., Offner, S., Blanco-Bose, W.E., Wai- R.E., Gallagher, N.J., Saglio, G. & Kantarjian,
relapse compared with the BCR-ABL1 fold rise: bler, Z., Kalinke, U., Duchosal, M.A. & Trumpp, H.M. (2012) Nilotinib vs imatinib in patients
implications for monitoring and management. A. (2009) IFNalpha activates dormant haemato- with newly diagnosed Philadelphia chromo-
Blood, 119, 42644271. poietic stem cells in vivo. Nature, 458, 904908. some-positive chronic myeloid leukemia in
Branford, S., Yeung, D.T., Ross, D.M., Prime, J.A., Flamant, S., Ritchie, W., Guilhot, J., Holst, J., Bon- chronic phase: ENESTnd 3-year follow-up. Leu-
Field, C.R., Altamura, H.K., Yeoman, A.L., net, M.L., Chomel, J.C., Guilhot, F., Turhan, kemia, 26, 21972203.
Georgievski, J., Jamison, B.A., Phillis, S., Sulli- A.G. & Rasko, J.E. (2010) Micro-RNA response Leber, B., Cervantes, F., Spector, N., Lipton, J.H.,
van, B., Briggs, N.E., Hertzberg, M., Seymour, to imatinib mesylate in patients with chronic Etienne, G., Pasquini, R., Clementino, N.C.,
J.F., Reynolds, J. & Hughes, T.P. (2013) Early myeloid leukemia. Haematologica, 95, 1325 Guerci-Bresler, A., Forrest, D., Schwarer, A.P.,
molecular response and female sex strongly pre- 1333. Acharya, S., Collins, L., Szczudlo, T. & Hughes,
dict stable undetectable BCR-ABL1, the criteria Goh, H.G., Lin, M., Fukushima, T., Saglio, G., T.P. (2013) Achievement and maintenance of
for imatinib discontinuation in patients with Kim, D., Choi, S.Y., Kim, S.H., Lee, J., Lee, Y.S., deeper molecular response by switching to nil-
CML. Blood, 121, 38183824. Oh, S.M. & Kim, D.W. (2011) Sensitive quanti- otinib in patients (pts) with chronic myeloid
Burchert, A., Wolfl, S., Schmidt, M., Brendel, C., tation of minimal residual disease in chronic leukemia in chronic phase (CML-CP) with
Denecke, B., Cai, D., Odyvanova, L., Lahaye, T., myeloid leukemia using nanofluidic digital poly- residual disease on long-term imatinib: ENES-
Muller, M.C., Berg, T., Gschaidmeier, H., Wit- merase chain reaction assay. Leukaemia & Lym- Tcmr 36-month follow-up. Blood (ASH Annual
tig, B., Hehlmann, R., Hochhaus, A. & Neu- phoma, 52, 896904. Meeting Abstracts), 120, 94.
bauer, A. (2003) Interferon-alpha, but not the Goldman, J. & Gordon, M. (2006) Why do Legros, L., Rousselot, P., Giraudier, S., Tulliez, M.,
ABL-kinase inhibitor imatinib (STI571), induces chronic myelogenous leukemia stem cells survive Huguet, F., Nicolini, F.E. & Mahon, F.X. (2012)
expression of myeloblastin and a specific T-cell allogeneic stem cell transplantation or imatinib: Second attempt to discontinue imatinib in CP-
response in chronic myeloid leukemia. Blood, does it really matter? Leukaemia & Lymphoma, CML patients with a second sustained complete
101, 259264. 47, 17. molecular response. Blood, 120, 19591960.
Burchert, A., Muller, M.C., Kostrewa, P., Erben, Guilhot, F., Chastang, C., Michallet, M., Guerci, Mahon, F.X., Delbrel, X., Cony-Makhoul, P., Fab-
P., Bostel, T., Liebler, S., Hehlmann, R., Neu- A., Harousseau, J.L., Maloisel, F., Bouabdallah, eres, C., Boiron, J.M., Barthe, C., Bilhou-Nabera,
bauer, A. & Hochhaus, A. (2010) Sustained R., Guyotat, D., Cheron, N., Nicolini, F., Abgr- C., Pigneux, A., Marit, G. & Reiffers, J. (2002)
molecular response with interferon alfa mainte- all, J.F. & Tanzer, J. (1997) Interferon alfa-2b Follow-up of complete cytogenetic remission in
nance after induction therapy with imatinib plus combined with cytarabine versus interferon patients with chronic myeloid leukemia after
interferon alfa in patients with chronic myeloid alone in chronic myelogenous leukemia. French cessation of interferon alfa. Journal of Clinical
leukemia. Journal of Clinical Oncology, 28, 1429 Chronic Myeloid Leukemia Study Group. New Oncology, 20, 214220.
1435. England Journal of Medicine, 337, 223229. Mahon, F.X., Rea, D., Guilhot, J., Guilhot, F.,
Chen, C.I., Maecker, H.T. & Lee, P.P. (2008) Iacobucci, I., Lonetti, A., Venturi, C., Ferrari, A., Huguet, F., Nicolini, F., Legros, L., Charbonnier,
Development and dynamics of robust T-cell Papayannidis, C., Ottaviani, E., Abbenante, A., Guerci, A., Varet, B., Etienne, G., Reiffers, J.
responses to CML under imatinib treatment. M.C., Paolini, S., Bresciani, P., Potenza, L., & Rousselot, P. (2010) Discontinuation of
Blood, 111, 53425349. Parisi, S., Cattina, F., Soverini, S., Russo, D., imatinib in patients with chronic myeloid
Chen, C.I., Koschmieder, S., Kerstiens, L., Schemi- Luppi, M. & Martinelli, G. (2014) Use of a high leukaemia who have maintained complete
onek, M., Altvater, B., Pscherer, S., Gerss, J., sensitive nanofluidic array for the detection molecular remission for at least 2 years: the pro-
Maecker, H.T., Berdel, W.E., Juergens, H., Lee, of rare copies of BCR-ABL1 transcript in spective, multicentre Stop Imatinib (STIM) trial.
P.P. & Rossig, C. (2012) NK cells are dysfunc- patients with Philadelphia-positive acute lym- Lancet Oncology, 11, 10291035.
tional in human chronic myelogenous leukemia phoblastic leukemia in complete response. Leu- Mahon, F.X., Nicolini, F.E., Noel, M.P., Escoffre,
before and on imatinib treatment and in BCR- kemia Research, doi: 10.1016/j.leukres.2014.02. M., Charbonnier, A., Rea, D., Dubruille, V.,
ABL-positive mice. Leukemia, 26, 465474. 005. Varet, B.R., Legros, L., Guerci, A., Etienne, G.,
Copland, M., Hamilton, A., Elrick, L.J., Baird, Kanodia, S., Wieder, E., Lu, S., Talpaz, M., Ala- Guilhot, F., Dulucq, S., Rousselot, P. & Guilhot,
J.W., Allan, E.K., Jordanides, N., Barow, M., trash, G., Clise-Dwyer, K. & Molldrem, J.J. J. (2013) Preliminary report of the STIM2 study:
Mountford, J.C. & Holyoake, T.L. (2006) Dasat- (2010) PR1-specific T cells are associated with a multicenter stop imatinib trial for chronic
inib (BMS-354825) targets an earlier progenitor unmaintained cytogenetic remission of chronic phase chronic myeloid leukemia de novo
population than imatinib in primary CML but myelogenous leukemia after interferon with- patients on imatinib. Blood (ASH Annual Meet-
does not eliminate the quiescent fraction. Blood, drawal. PLoS ONE, 5, e11770. ing Abstracts), 122, 654.
107, 45324539. Kantarjian, H.M., Shah, N.P., Cortes, J.E., Baccara- Michor, F., Hughes, T.P., Iwasa, Y., Branford, S.,
Ding, J., Romani, J., Zaborski, M., Macleod, R.A., ni, M., Agarwal, M.B., Undurraga, M.S., Wang, Shah, N.P., Sawyers, C.L. & Nowak, M.A.
Nagel, S., Drexler, H.G. & Quentmeier, H. J., Ipina, J.J., Kim, D.W., Ogura, M., Pavlovsky, (2005) Dynamics of chronic myeloid leukaemia.
(2013) Inhibition of PI3K/mTOR overcomes nil- C., Junghanss, C., Milone, J.H., Nicolini, F.E., Nature, 435, 12671270.
otinib resistance in BCR-ABL1 positive leukemia Robak, T., Van Droogenbroeck, J., Vellenga, E., Molldrem, J.J., Lee, P.P., Wang, C., Felio, K., Kan-
cells through translational down-regulation of Bradley-Garelik, M.B., Zhu, C. & Hochhaus, A. tarjian, H.M., Champlin, R.E. & Davis, M.M.
MDM2. PLoS ONE, 8, e83510. (2012) Dasatinib or imatinib in newly diagnosed (2000) Evidence that specific T lymphocytes
Efficace, F., Baccarani, M., Breccia, M., Alimena, chronic-phase chronic myeloid leukemia: 2-year may participate in the elimination of chronic
G., Rosti, G., Cottone, F., Deliliers, G.L., Barate, follow-up from a randomized phase 3 trial myelogenous leukemia. Nature Medicine, 6,
C., Rossi, A.R., Fioritoni, G., Luciano, L., Turri, (DASISION). Blood, 119, 11231129. 10181023.
D., Martino, B., Di Raimondo, F., Dabusti, M., Kumari, A., Brendel, C., Hochhaus, A., Neubauer, Mori, S., Vagge, E., le Coutre, P., Abruzzese, E.,
Bergamaschi, M., Leoni, P., Simula, M.P., Leva- A. & Burchert, A. (2012) Low BCR-ABL expres- Martino, B., Pungolino, E., Elena, C., Pierri, I.,

10 2014 John Wiley & Sons Ltd


British Journal of Haematology, 2014, 166, 311
Review

Assouline, S., DEmilio, A., Gozzini, A., Giraldo, BCR-ABL transcripts. Blood (ASH Annual Meet- Schmidt, M., Hochhaus, A., Nitsche, A., Hehl-
P., Stagno, F., Nagler, A., Iurlo, A., Rizzo, C., ing Abstracts), 120, 916. mann, R. & Neubauer, A. (2001) Expression of
Fossati, A., Pogliani, E.M., Kim, D.W., Pirola, Rea, D., Dulphy, N., Henry, G., Guilhot, J., Guil- nuclear transcription factor interferon consensus
A., Piazza, R. & Gambacorti-Passerini, C. (2013) hot, F., Nicolini, F.E., Legros, L., Rousselot, P., sequence binding protein in chronic myeloid
Validation of digital-PCR analysis through pro- Mahon, F.X. & Toubert, A. (2013) Low natural leukemia correlates with pretreatment risk fea-
grammed imatinib interruption in Q-RT-PCR killer (NK) cell counts and functionality are tures and cytogenetic response to interferon-
negative chronic myeloid leukemia patients. associated with molecular relapse after imatinib alpha. Blood, 97, 36483650.
Blood (ASH Annual Meeting Abstracts), 122, discontinuation in patients (pts) with chronic Score, J., Calasanz, M.J., Ottman, O., Pane, F.,
4040. phase (CP)-chronic myeloid leukemia (CML) Yeh, R.F., Sobrinho-Simoes, M.A., Kreil, S.,
Mughal, T.I., Yong, A., Szydlo, R.M., Dazzi, F., with undetectable BCR-ABL transcripts for at Ward, D., Hidalgo-Curtis, C., Melo, J.V., Wie-
Olavarria, E., van Rhee, F., Kaeda, J., Cross, least 2 years: preliminary results from immuno- mels, J., Nadel, B., Cross, N.C. & Grand, F.H.
N.C., Craddock, C., Kanfer, E., Apperley, J. & stim, On Behalf Of STIM Investigators. Blood (2010) Analysis of genomic breakpoints in p190
Goldman, J.M. (2001) Molecular studies in (ASH Annual Meeting Abstracts), 122, 856. and p210 BCR-ABL indicate distinct mecha-
patients with chronic myeloid leukaemia in Roeder, I., Horn, M., Glauche, I., Hochhaus, A., nisms of formation. Leukemia, 24, 17421750.
remission 5 years after allogeneic stem cell Mueller, M.C. & Loeffler, M. (2006) Dynamic Sokal, J.E., Cox, E.B., Baccarani, M., Tura, S., Gomez,
transplant define the risk of subsequent modeling of imatinib-treated chronic myeloid G.A., Robertson, J.E., Tso, C.Y., Braun, T.J., Clark-
relapse. British Journal of Haematology, 115, leukemia: functional insights and clinical impli- son, B.D., Cervantes, F., Rozman, C. and The
569574. cations. Nature Medicine, 12, 11811184. Italian Cooperative CML Study Group. (1984)
Mumprecht, S., Schurch, C., Schwaller, J., Solent- Ross, D.M., Branford, S., Seymour, J.F., Schwarer, Prognostic discrimination in good-risk chronic
haler, M. & Ochsenbein, A.F. (2009) Pro- A.P., Arthur, C., Bartley, P.A., Slader, C., Field, granulocytic leukemia. Blood, 63, 789799.
grammed death 1 signaling on chronic myeloid C., Dang, P., Filshie, R.J., Mills, A.K., Grigg, Takahashi, N., Kyo, T., Maeda, Y., Sugihara, T.,
leukemia-specific T cells results in T-cell exhaus- A.P., Melo, J.V. & Hughes, T.P. (2010) Patients Usuki, K., Kawaguchi, T., Usui, N., Okamoto,
tion and disease progression. Blood, 114, 1528 with chronic myeloid leukemia who maintain a S., Ohe, Y., Ohtake, S., Kitamura, K., Yamamot-
1536. complete molecular response after stopping o, M., Teshima, H., Motoji, T., Tamaki, T., Saw-
Norkin, M., Uberti, J.P. & Schiffer, C.A. (2011) imatinib treatment have evidence of persistent ada, K. & Ohyashiki, K. (2012) Discontinuation
Very late recurrences of leukemia: why does leu- leukemia by DNA PCR. Leukemia, 24, 1719 of imatinib in Japanese patients with chronic
kemia awake after many years of dormancy? 1724. myeloid leukemia. Haematologica, 97, 903906.
Leukemia Research, 35, 139144. Ross, D.M., Bartley, P.A., Goyne, J., Morley, A.A., Tanaka, H., Imagawa, J., Okada, M., Nakamae, H.,
Ohyashiki, K., Katagiri, S., Tauchi, T., Ohyashiki, Seymour, J.F. & Grigg, A.P. (2011) Durable Hino, M., Murai, K., Ishida, Y., Kumagai, T.,
J.H., Maeda, Y., Matsumura, I. & Kyo, T. complete molecular remission of chronic mye- Sato, S., Ohashi, K., Sakamaki, H., Wakita, H.,
(2012) Increased natural killer cells and loid leukemia following dasatinib cessation, Uoshima, N., Nakagawa, Y., Minami, Y., Oga-
decreased CD3(+)CD8(+)CD62L(+) T cells in despite adverse disease features. Haematologica, sawara, M., Takeoka, T., Akasaka, H., Morita,
CML patients who sustained complete molecular 96, 17201722. S., Sakamoto, J. & Kimura, S. (2013) Discontin-
remission after discontinuation of imatinib. Ross, D.M., Branford, S., Seymour, J.F., Schwarer, uation of dasatinib in patients with CML who
British Journal of Haematology, 157, 254256. A.P., Arthur, C., Yeung, D.T., Dang, P., Goyne, have maintained complete molecular response
Preudhomme, C., Guilhot, J., Nicolini, F.E., J.M., Slader, C., Filshie, R.J., Mills, A.K., Melo, for at least one year: results from a prospective
Guerci-Bresler, A., Rigal-Huguet, F., Maloisel, J.V., White, D.L., Grigg, A.P. & Hughes, T.P. discontinuation (DADI) trial. Blood (ASH
F., Coiteux, V., Gardembas, M., Berthou, C., (2013) Safety and efficacy of imatinib cessation Annual Meeting Abstracts), 122, 3998.
Vekhoff, A., Rea, D., Jourdan, E., Allard, C., for CML patients with stable undetectable mini- Verma, D., Kantarjian, H., Jain, N. & Cortes, J.
Delmer, A., Rousselot, P., Legros, L., Berger, M., mal residual disease: results from the TWISTER (2008) Sustained complete molecular response
Corm, S., Etienne, G., Roche-Lestienne, C., study. Blood, 122, 515522. after imatinib discontinuation in a patient with
Eclache, V., Mahon, F.X. & Guilhot, F. (2010) Rousselot, P., Huguet, F., Rea, D., Legros, L., chronic myeloid leukemia not previously
Imatinib plus peginterferon alfa-2a in chronic Cayuela, J.M., Maarek, O., Blanchet, O., Marit, exposed to interferon alpha. Leukaemia & Lym-
myeloid leukemia. New England Journal of Medi- G., Gluckman, E., Reiffers, J., Gardembas, M. & phoma, 49, 13991402.
cine, 363, 25112521. Mahon, F.X. (2007) Imatinib mesylate discon- Warsch, W., Walz, C. & Sexl, V. (2013) JAK of all
Radich, J.P., Gehly, G., Gooley, T., Bryant, E., tinuation in patients with chronic myelogenous trades: JAK2-STAT5 as novel therapeutic targets
Clift, R.A., Collins, S., Edmands, S., Kirk, J., leukemia in complete molecular remission for in BCR-ABL1+ chronic myeloid leukemia.
Lee, A., Kessler, P., Schoch, G., Buckner, C.D., more than 2 years. Blood, 109, 5860. Blood, 122, 21672175.
Sullivan, K.M., Appelbaum, F.R. & Thomas, Rousselot, P., Charbonnier, A., Cony-Makhoul, P., Yhim, H.Y., Lee, N.R., Song, E.K., Yim, C.Y., Jeon,
E.D. (1995) Polymerase chain reaction detection Agape, P., Nicolini, F.E., Varet, B., Gardembas, S.Y., Shin, S., Kim, J.A., Kim, H.S., Cho, E.H. &
of the BCR-ABL fusion transcript after alloge- M., Etienne, G., Rea, D., Roy, L., Escoffre-Barbe, Kwak, J.Y. (2012) Imatinib mesylate discontinu-
neic marrow transplantation for chronic mye- M., Guerci-Bresler, A., Tulliez, M., Prost, S., ation in patients with chronic myeloid leukemia
loid leukemia: results and implications in 346 Spentchian, M., Cayuela, J.M., Reiffers, J., Cho- who have received front-line imatinib mesylate
patients. Blood, 85, 26322638. mel, J.C., Turhan, A., Guilhot, J., Guilhot, F. & therapy and achieved complete molecular
Rea, D., Rousselot, P., Guilhot, F., Tulliez, M., Mahon, F.X. (2014) Loss of major molecular response. Leukemia Research, 36, 689693.
Nicolini, F., Guerci-Bresler, A., Legros, L., response as a trigger for restarting tyrosine Zhang, B., Irvine, D., Ho, Y.W., Buonamici, S., Man-
Gardembas, M., Giraudier, S., Guillerm, G. & kinase inhibitor therapy in patients with ley, P., Holyoake, T.L., Copland, M. & Bhatia, R.
Mahon, F. (2012) Discontinuation of second chronic-phase chronic myelogenous leukemia (2010) Inhibition of chronic myeloid leukemia
generation (2G) tyrosine kinase inhibitors (TKI) who have stopped imatinib after durable unde- stem cells by the combination of the hedgehog
in chronic phase (CP)-chronic myeloid leukemia tectable disease. Journal of Clinical Oncology, 32, pathway inhibitor LDE225 with nilotinib. Blood
(CML) patients with stable undetectable 424430. (ASH Annual Meeting Abstracts), 116, 514.

2014 John Wiley & Sons Ltd 11


British Journal of Haematology, 2014, 166, 311

Vous aimerez peut-être aussi