The introduction of tyrosine kinase inhibitors (TKIs), a treatment of chronic myelogenous leukemia (CML), has largely replaced curative strategies based on allogeneic stem cell transplantation (SCT). Nevertheless, SCT still remains an option for accelerated/blastic-phase and selected chronic-phase CML. Transplant outcomes can be optimized by peritransplant TKIs, conditioning regimen, BCR-ABL monitoring, and relapse management. Controversies exist in transplant timing, pediatric CML, alternative donors, and economics. SCT continues to serve as a platform of “operational cure” for CML with TKIs and immunotherapies.

Stem cell transplantation (SCT) to treat chronic myelogenous leukemia (CML) was pioneered by Buckner et al1,2  and subsequently by Goldman et al,3  with the aim of treating accelerated-phase (AP) and blastic-phase (BP) CML with myeloablative radiation and an autologous chronic-phase (CP) bone marrow transplant to “set the clock back” to a more benign disease state.1-3  In most patients, this approach failed to control the leukemia but set the stage for allogeneic (allo)-SCT with syngeneic graft introduced by Fefer et al,4  and subsequently for bone marrow graft from HLA-matched siblings reported in 1982 by 3 groups.5-7  Outcomes in CP-CML patients were particularly promising, but it was soon realized that leukemic relapse was more frequent in T cell–depleted recipients and in those who did not develop graft-versus-host disease (GVHD).8,9  These experiences supported the evidence in humans that alloreactive T cells exerted a powerful graft-versus-leukemia (GVL) effect, which was directly confirmed by Kolb et al,10  who showed that donor lymphocyte infusions (DLIs) could achieve stable second remissions in CML patients relapsing after SCT.10-13  Outcomes for CP-CML patients continued to improve through the 1990s with general improvements in transplant management. The most recent reports indicate overall survival (OS) rates of over 85% for CP-CML patients receiving a matched-donor transplant.8  A European Group for Blood and Marrow Transplantation scoring system permitted the prediction of outcome based on disease status, donor status, and age.14 

The decisive demonstration by Druker et al15,16  that the tyrosine kinase inhibitor (TKI) imatinib was safe and highly effective at controlling CP-CML heralded a rapid decline in SCT for CP-CML, documented by the databases of both the Center for International Blood and Marrow Transplant Research (CIBMTR) and the European Group for Blood and Marrow Transplantation. Effective TKI therapy supplanted SCT because it represented a safer, low-technology alternative (no immediate drug-related mortality). Nevertheless, SCT is still preferred for patients in more advanced-phase CML and selected cases of CP-CML.8,17-19  Not all patients tolerate TKIs, some patients progress despite second- or third-line TKIs, and some develop TKI-resistant mutations. Although ponatinib and omacetaxine have some activities against the case with T351I mutation,20,21  SCT, which can achieve prolonged progression-free survival (PFS) in such mutations, may be preferable. Lastly, CML leukemia stem cells (LSCs) are not dependent on BCR-ABL signaling for survival,22  and the quiescent LSC population is not eliminated by TKIs.23  The generally accepted current indications for allo-SCT in CML are listed in Table 1.

The German CML Study Group24  reported 3-year respective OS rates after allo-SCT in selected high-risk CP, imatinib-failure CP, and AP/BP patients of 88%, 94%, and 59%, with only 8% transplant-related mortality (TRM), and concluded that allo-SCT is a favorable second-line option following first TKI failure. In a prospective study in AP-CML (not including BP), Jiang et al25  demonstrated an advantage for allo-SCT over imatinib (6-year OS, 83.3% vs 51.4%; PFS, 71.8% vs 39.2%). In imatinib-resistant CML (including 40% BCR-ABL1 mutations), SCT had a response rate of 91%, a 2-year OS rate of 63%, and a PFS rate of 49%.26  Lastly, the latest CIBMTR analysis of allo-SCT for CML in the TKI era reported 3-year OS rates of 36% in second CP, 43% in AP, and 14% in BP.27  These data support a continuing role of allo-SCT as a salvage treatment of CML.

Pretransplant TKI therapy

A CIBMTR analysis found that pretransplant TKI therapy also improved posttransplant survival in CP28  but not in advanced disease.27  In another study, major or complete cytogenetic response to TKI therapy before allo-SCT was associated with better posttransplant outcome.29  The choice of pretransplant TKI for advanced-phase disease is not well standardized, but dasatinib and nilotinib were at least safely administered before allo-SCT without increased TRM.30  Thus, it is appropriate to use TKIs to reduce the disease burden before allo-SCT for AP and BP-CML.

Intensity of conditioning

The powerful GVL effect in CML has prompted several studies to explore reduced-intensity conditioning (RIC) following the assumption that disease control in CP depends mainly on the rapid establishment of donor lymphoid engraftment. The limits of intensity reduction are now defined: we found that the entirely nonmyeloablative combination of fludarabine and cyclophosphamide followed by an HLA-matched sibling peripheral blood stem cell allograft achieved full, sustained molecular remission, but in only 2 patients. Although there was no treatment-related mortality, 4 other recipients required repeated DLIs or full-intensity conditioning SCT to achieve sustained molecular cure.31  In contrast, reduced-intensity SCT using combinations of fludarabine, anti-lymphocyte globulin, and busulfan were effective.32  The need to reduce conditioning regimens for older and debilitated patients has prompted a number of investigators to use RIC (usually by reducing doses of busulfan and fludarabine) to minimize regimen toxicity. It is unfortunate that these approaches show no superiority over full myeloablative transplants.33,34  Mortality in these patients can exceed 30% because of GVHD, failure of residual disease control, and older age of the recipient.34  In a study in which the outcomes of 28 patients receiving RIC conditioning were compared to those of 56 recipients of myeloablative SCT matched for disease severity and stage,35  the probabilities of 5- and 10-year leukemia-free survival and OS were similar. However, RIC recipients had higher rates of relapse, whereas myeloablative recipients had higher mortality. A multicenter CIBMTR analysis compared RIC regimens with even less intensive nonmyeloablative regimens (given largely to older recipients). Among other risk factors, when compared to nonmyeloablative regimens, RIC regimens were associated with a threefold lower risk of relapse and an almost twofold higher disease-free survival in a multivariate analysis.36  Thus, there is no advantage for nonmyeloablative regimens. RIC regimens may have a place for older recipients but, for other patients and those with advanced CML, an intensive conditioning offers the best chance to control disease.

GVHD prophylaxis

GVHD is strongly linked to the GVL effect in CML37 ; thus, the choice of GVHD prophylaxis can play a critical role in transplant outcome. The role of T-cell depletion in favoring residual disease and leukemic recurrence has long been known in CML. Nevertheless, T-cell depletion has its advocates for CML SCT because it favors GVHD-free survival, and disease recurrence can be controlled with DLIs.38  Transplant regimens using the monoclonal antibody alemtuzumab are similarly associated with effective GVHD control, but also with prolonged immunosuppression.39  These regimens are associated with higher relapse rates but can be controlled with DLIs and TKIs, which can be useful adjuvants, even in patients previously resistant to these agents.

Posttransplant BCR-ABL monitoring

Because GVL is slow to develop, detection of BCR-ABL in the first few months after SCT has no adverse prognostic significance.40  However, the pattern of BCR-ABL (persistently negative, fluctuating, or persistently positive) after 6 months posttransplant predicts relapse risk.41  A recent study, however, showed that very low levels of persistent disease (ABL/BCR ratio <0.1%) occurring up to 10 years posttransplant had less implication for relapse: of 52 patients with occasional low levels of BCR/ABL detection posttransplant, 6 relapsed but 35 ultimately became polymerase chain reaction negative.40  In the RIC allo-SCT, BCR-ABL transcript may be detectable much longer, and preemptive DLIs may be needed to achieve sustained molecular remission without use of TKIs.42  Continued regular long-term monitoring of BCR-ABL posttransplant is needed to anticipate the occasional late relapsing patient; however, both the optimal frequency of monitoring and the threshold of BCR-ABL transcripts for preemptive therapy with TKIs or DLIs need to be established in the contexts of conditioning regimen and graft manipulation.

Posttransplant prophylactic TKI therapy

Given the high proportion of high-risk CML patients now selected for SCT, the role of prophylactic TKI therapy to prevent relapse has been extensively explored. Several reports suggest that early posttransplant TKIs (including second-generation TKIs) are safe to administer effectively in CP-CML but are less effective in advanced CML.43,44  Furthermore, the administration of TKIs with DLIs appears to be safe and does not risk GVHD. In 2 retrospective analyses, posttransplant TKI therapy was associated with a lower incidence of extensive chronic GVHD45  (hypothesized as the effects of TKI on platelet-derived growth factor receptor pathways).44  Although the use of posttransplant TKI therapy is widespread, prospective studies are needed to explore the best TKI dose, treatment duration, and coadministration with DLIs.

Posttransplant relapse

Relapse of CML can occur as late as the second decade after allo-SCT.46  Relapsed CML has been treated with DLIs, TKIs, chemotherapy, and second allo-SCT.47-53  Molecular relapse is frequent after T cell–depleted transplantation but can be salvaged. We reported that combinations of TKI therapy and DLIs can achieve a 5-year postrelapse survival rate of 62%, and some patients became TKI-free, suggesting a persistent GVL effect.53 

Transplant timing

Opinions differ as to whether patients failing second-line TKI should receive a third-line agent or be considered for SCT. The decision to transplant is favored by the presence of unfavorable BCR-ABL kinase-domain mutations. Data from the CIBMTR showed that diagnosis-to-transplant intervals >12 months were associated with worse OS and PFS in AP-CML patients, irrespective of pretransplant TKI therapy.27  AP-CML patients are a heterogeneous population, and further risk stratification is needed to identify the high-risk group suitable for early allo-SCT. Whatever the ultimate treatment choice, it is reasonable to perform HLA typing and a donor search after first-line TKI failure for AP-CML. For BP-CML, prompt referral to a transplant center is critical, along with the immediate initiation of TKI therapy and induction chemotherapy.

Treatment of children with CML

There is unease about consigning children developing CML to a lifetime of TKI therapy. Several groups report imatinib-induced growth delay, especially in prepubertal children, due to altered bone metabolism and growth hormone suppression.54-56  The International BFM Study Group and other experts recommended that guidelines for children with CML should follow those for adults.57,58  However, there are still concerns for lifelong safety and quality of life when using TKIs in pediatric CML. Fatigue, limited physical activity, and emotional problems are the major limitations in TKI use in CP-CML, especially in young and female patients.59  The shortcomings of long-term TKI use must be balanced against the potential complications of chronic GVHD and long-term immunosuppression, which are major factors associated with reduced health-related quality of life.60,61  A recent French study62  reported that 37% of children fail to achieve <10% BCR-ABL1 transcripts at 3 months and have shorter PFS, suggesting less-favorable kinetics of disease or TKI efficacy in pediatric CML. This observation opens the question for the indication of allo-SCT, especially when an HLA-identical sibling is available, because outcome of SCT in children is generally more favorable than in adults. More prospective data are urgently needed to define the best treatment approach for pediatric CML. Meanwhile, treatment decisions must be made on a case-by-case basis.

Pregnancy in CML

Imatinib and other TKIs are known to be associated with a higher risk of fetal malformation in women with CML,63,64  and the interruption of TKI therapy during pregnancy was associated with poor outcomes in CML.65  However, infertility is a major late event after allo-SCT,66,67  and the best strategy for fertility preservation is still in development. Both RIC (especially avoiding total body irradiation) and cryopreservation of sperm, oocyte, or gonadal tissue resulted in successful pregnancy in posttransplant survivors with CML.68,69  Fertility preservation should be discussed before allo-SCT with both pediatric and adult recipients who desire to bear children.

Identical-twin donors

The first successful SCT in CML using a healthy donor was from an identical twin.4  A CIBMTR analysis demonstrated that although the relapse rate of 40% in CML patients receiving syngeneic SCT was higher than the 7% observed in HLA-matched sibling recipients (as predicted by the lack of an allogeneic GVL effect), the long-term PFS rate of 59% was equivalent to the 61% seen in HLA-identical sibling recipients.70  Furthermore, patients receiving a larger marrow-cell dose from their twin had a significantly lower relapse rate, suggesting protective effects of some graft component against relapse.71  Given the favorable outcome for SCT in syngeneic transplants and the absence of severe GVHD, the opportunity to use an identical-twin donor should not be overlooked.

Alternative donors

A recent study from China using unrelated cord blood transplantation in AP/BP-CML showed 5-year OS and PFS rates of 62.5% and 50%, respectively, equivalent to the outcomes of their HLA-matched sibling cohort despite a higher TRM.72  Preliminary data using haploidentical SCT with postgraft cyclophosphamide have demonstrated the safety (0% TRM) and efficacy (60% OS and PFS) of this approach.73  These results should encourage the use of alternative graft sources for CML patients requiring allo-SCT who lack a fully matched related or unrelated donor.

Health economics

The high cost of lifetime TKI therapy can be simply unaffordable.74  SCT can be a less-expensive alternative to TKIs in some countries. A prospective study from Mexico75  not only showed comparable PFS rates for TKI therapy and SCT but also demonstrated that transplants were less expensive. An analysis from Sweden76  comparing pre-TKI (SCT-predominant) and post-TKI periods estimated the incremental cost-effectiveness ratio as €52 700 per quality-adjusted life-year gained. The incremental cost-effectiveness ratio was predicted to fall to only €22 700 per quality-adjusted life-year after the patent expiry of imatinib with an 80% cost reduction. Cost-effectiveness data should be interpreted in the context of health care systems in each country, and treatment choices for CML may be modified according to the local imperatives of patient care.

In the concept of cure in CML, Goldman and others preferred the term “operational cure”: prolonged survival in molecular remission without therapy.77,78  The challenge is to find a strategy to avoid lifelong dependency on TKI therapy,79  which in addition to its cumulative expense, may have unrecognized adverse effects. CML persistence, despite control of the leukemia at the molecular level of detection, relates to the inability of current therapy to target quiescent LSCs.80  Although neither TKI nor transplant strategies always succeed to eradicate the quiescent LSCs, using allo-SCT after deep remission is induced by TKI could achieve operational cure. The third option for operational cure is immunotherapy evolving from GVL effects observed in SCT. Many immunotherapeutic approaches are under investigation; clinical trials of vaccination with leukemia-associated antigens (LAAs) such as BCR-ABL, WT1, and PR1 showed variable immunologic and clinical responses in CML patients.81-85  Adoptive T-cell immunotherapy using multi-LAA–specific T cells86,87  and chimeric antigen receptor–modified T cells88  demonstrated antileukemic activities either preclinically or clinically in a phase I study. Novel LAAs (aurora A kinase89  and BMI-190 ) or surface molecules (IL-1RAP91  and CD2692 ) specific to CML LSCs have recently been discovered and would be applicable for future immunotherapy. Given that the safety of SCT is continually evolving,93  it is important to maintain an open mind about the application of SCT as a platform for future targeted therapy and immunotherapy in CML.

The authors dedicate this article to the memory of John Michael Goldman who inspired us with his insightful observations on the definition and nature of cure of CML.

This study was supported by the National Institutes of Health Intramural Research Program at the National Heart, Lung, and Blood Institute.

Contribution: A.J.B. and S.I. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: A. John Barrett, Hematology Branch, NHLBI, NIH, Building 10, Room 7C103, 9000 Rockville Pike, Bethesda, MD 20892; e-mail: barrettj@nhlbi.nih.gov.

1
Buckner
 
CD
Clift
 
RA
Fefer
 
A
Neiman
 
PE
Storb
 
R
Thomas
 
ED
Treatment of blastic transformation of chronic granulocytic leukemia by high dose cyclophosphamide, total body irradiation and infusion of cryopreserved autologous marrow.
Exp Hematol
1974
, vol. 
2
 
3
(pg. 
138
-
146
)
2
Buckner
 
CD
Stewart
 
P
Clift
 
RA
, et al. 
Treatment of blastic transformation of chronic granulocytic leukemia by chemotherapy, total body irradiation and infusion of cryopreserved autologous marrow.
Exp Hematol
1978
, vol. 
6
 
1
(pg. 
96
-
109
)
3
Goldman
 
JM
Catovsky
 
D
Hows
 
J
Spiers
 
AS
Galton
 
DA
Cryopreserved peripheral blood cells functioning as autografts in patients with chronic granulocytic leukaemia in transformation.
BMJ
1979
, vol. 
1
 
6174
(pg. 
1310
-
1313
)
4
Fefer
 
A
Cheever
 
MA
Thomas
 
ED
, et al. 
Disappearance of Ph1-positive cells in four patients with chronic granulocytic leukemia after chemotherapy, irradiation and marrow transplantation from an identical twin.
N Engl J Med
1979
, vol. 
300
 
7
(pg. 
333
-
337
)
5
Clift
 
RA
Buckner
 
CD
Thomas
 
ED
, et al. 
Treatment of chronic granulocytic leukaemia in chronic phase by allogeneic marrow transplantation.
Lancet
1982
, vol. 
320
 
8299
(pg. 
621
-
623
)
6
Goldman
 
JM
Baughan
 
AS
McCarthy
 
DM
, et al. 
Marrow transplantation for patients in the chronic phase of chronic granulocytic leukaemia.
Lancet
1982
, vol. 
320
 
8299
(pg. 
623
-
625
)
7
McGlave
 
PB
Arthur
 
DC
Kim
 
TH
Ramsay
 
NK
Hurd
 
DD
Kersey
 
J
Successful allogeneic bone-marrow transplantation for patients in the accelerated phase of chronic granulocytic leukaemia.
Lancet
1982
, vol. 
320
 
8299
(pg. 
625
-
627
)
8
Pavlu
 
J
Szydlo
 
RM
Goldman
 
JM
Apperley
 
JF
Three decades of transplantation for chronic myeloid leukemia: what have we learned?
Blood
2011
, vol. 
117
 
3
(pg. 
755
-
763
)
9
Horowitz
 
MM
Gale
 
RP
Sondel
 
PM
, et al. 
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
1990
, vol. 
75
 
3
(pg. 
555
-
562
)
10
Kolb
 
HJ
Mittermüller
 
J
Clemm
 
C
, et al. 
Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients.
Blood
1990
, vol. 
76
 
12
(pg. 
2462
-
2465
)
11
van Rhee
 
F
Lin
 
F
Cullis
 
JO
, et al. 
Relapse of chronic myeloid leukemia after allogeneic bone marrow transplant: the case for giving donor leukocyte transfusions before the onset of hematologic relapse.
Blood
1994
, vol. 
83
 
11
(pg. 
3377
-
3383
)
12
Mackinnon
 
S
Papadopoulos
 
EB
Carabasi
 
MH
, et al. 
Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease.
Blood
1995
, vol. 
86
 
4
(pg. 
1261
-
1268
)
13
Adekola
 
K
Popat
 
U
Ciurea
 
SO
An update on allogeneic hematopoietic progenitor cell transplantation for myeloproliferative neoplasms in the era of tyrosine kinase inhibitors.
Bone Marrow Transplant
2014
, vol. 
49
 
11
(pg. 
1352
-
1359
)
14
Gratwohl
 
A
The EBMT risk score.
Bone Marrow Transplant
2012
, vol. 
47
 
6
(pg. 
749
-
756
)
15
Druker
 
BJ
Guilhot
 
F
O’Brien
 
SG
, et al. 
IRIS Investigators
Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia.
N Engl J Med
2006
, vol. 
355
 
23
(pg. 
2408
-
2417
)
16
Druker
 
BJ
Talpaz
 
M
Resta
 
DJ
, et al. 
Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.
N Engl J Med
2001
, vol. 
344
 
14
(pg. 
1031
-
1037
)
17
Baccarani
 
M
Deininger
 
MW
Rosti
 
G
, et al. 
European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013.
Blood
2013
, vol. 
122
 
6
(pg. 
872
-
884
)
18
Jabbour
 
E
Kantarjian
 
H
Chronic myeloid leukemia: 2014 update on diagnosis, monitoring, and management.
Am J Hematol
2014
, vol. 
89
 
5
(pg. 
547
-
556
)
19
O’Brien
 
S
Radich
 
JP
Abboud
 
CN
, et al. 
Ntational comprehensive cancer network
Chronic Myelogenous Leukemia, Version 1.2014.
J Natl Compr Canc Netw
2013
, vol. 
11
 
11
(pg. 
1327
-
1340
)
20
Cortes
 
JE
Kantarjian
 
H
Shah
 
NP
, et al. 
Ponatinib in refractory Philadelphia chromosome-positive leukemias.
N Engl J Med
2012
, vol. 
367
 
22
(pg. 
2075
-
2088
)
21
Cortes
 
J
Lipton
 
JH
Rea
 
D
, et al. 
Omacetaxine 202 Study Group
Phase 2 study of subcutaneous omacetaxine mepesuccinate after TKI failure in patients with chronic-phase CML with T315I mutation.
Blood
2012
, vol. 
120
 
13
(pg. 
2573
-
2580
)
22
Hamilton
 
A
Helgason
 
GV
Schemionek
 
M
, et al. 
Chronic myeloid leukemia stem cells are not dependent on Bcr-Abl kinase activity for their survival.
Blood
2012
, vol. 
119
 
6
(pg. 
1501
-
1510
)
23
Copland
 
M
Hamilton
 
A
Elrick
 
LJ
, et al. 
Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction.
Blood
2006
, vol. 
107
 
11
(pg. 
4532
-
4539
)
24
Saussele
 
S
Lauseker
 
M
Gratwohl
 
A
, et al. 
German CML Study Group
Allogeneic hematopoietic stem cell transplantation (allo SCT) for chronic myeloid leukemia in the imatinib era: evaluation of its impact within a subgroup of the randomized German CML Study IV.
Blood
2010
, vol. 
115
 
10
(pg. 
1880
-
1885
)
25
Jiang
 
Q
Xu
 
LP
Liu
 
DH
, et al. 
Imatinib mesylate versus allogeneic hematopoietic stem cell transplantation for patients with chronic myelogenous leukemia in the accelerated phase.
Blood
2011
, vol. 
117
 
11
(pg. 
3032
-
3040
)
26
Jabbour
 
E
Cortes
 
J
Santos
 
FP
, et al. 
Results of allogeneic hematopoietic stem cell transplantation for chronic myelogenous leukemia patients who failed tyrosine kinase inhibitors after developing BCR-ABL1 kinase domain mutations.
Blood
2011
, vol. 
117
 
13
(pg. 
3641
-
3647
)
27
Khoury
 
HJ
Kukreja
 
M
Goldman
 
JM
, et al. 
Prognostic factors for outcomes in allogeneic transplantation for CML in the imatinib era: a CIBMTR analysis.
Bone Marrow Transplant
2012
, vol. 
47
 
6
(pg. 
810
-
816
)
28
Lee
 
SJ
Kukreja
 
M
Wang
 
T
, et al. 
Impact of prior imatinib mesylate on the outcome of hematopoietic cell transplantation for chronic myeloid leukemia.
Blood
2008
, vol. 
112
 
8
(pg. 
3500
-
3507
)
29
Oehler
 
VG
Gooley
 
T
Snyder
 
DS
, et al. 
The effects of imatinib mesylate treatment before allogeneic transplantation for chronic myeloid leukemia.
Blood
2007
, vol. 
109
 
4
(pg. 
1782
-
1789
)
30
Jabbour
 
E
Cortes
 
J
Kantarjian
 
H
, et al. 
Novel tyrosine kinase inhibitor therapy before allogeneic stem cell transplantation in patients with chronic myeloid leukemia: no evidence for increased transplant-related toxicity.
Cancer
2007
, vol. 
110
 
2
(pg. 
340
-
344
)
31
Sloand
 
E
Childs
 
RW
Solomon
 
S
Greene
 
A
Young
 
NS
Barrett
 
AJ
The graft-versus-leukemia effect of nonmyeloablative stem cell allografts may not be sufficient to cure chronic myelogenous leukemia.
Bone Marrow Transplant
2003
, vol. 
32
 
9
(pg. 
897
-
901
)
32
Or
 
R
Shapira
 
MY
Resnick
 
I
, et al. 
Nonmyeloablative allogeneic stem cell transplantation for the treatment of chronic myeloid leukemia in first chronic phase.
Blood
2003
, vol. 
101
 
2
(pg. 
441
-
445
)
33
Das
 
M
Saikia
 
TK
Advani
 
SH
Parikh
 
PM
Tawde
 
S
Use of a reduced-intensity conditioning regimen for allogeneic transplantation in patients with chronic myeloid leukemia.
Bone Marrow Transplant
2003
, vol. 
32
 
2
(pg. 
125
-
129
)
34
Kebriaei
 
P
Detry
 
MA
Giralt
 
S
, et al. 
Long-term follow-up of allogeneic hematopoietic stem-cell transplantation with reduced-intensity conditioning for patients with chronic myeloid leukemia.
Blood
2007
, vol. 
110
 
9
(pg. 
3456
-
3462
)
35
Topcuoglu
 
P
Arat
 
M
Ozcan
 
M
, et al. 
Case-matched comparison with standard versus reduced intensity conditioning regimen in chronic myeloid leukemia patients.
Ann Hematol
2012
, vol. 
91
 
4
(pg. 
577
-
586
)
36
Warlick
 
E
Ahn
 
KW
Pedersen
 
TL
, et al. 
Reduced intensity conditioning is superior to nonmyeloablative conditioning for older chronic myelogenous leukemia patients undergoing hematopoietic cell transplant during the tyrosine kinase inhibitor era.
Blood
2012
, vol. 
119
 
17
(pg. 
4083
-
4090
)
37
Stern
 
M
de Wreede
 
LC
Brand
 
R
, et al. 
Sensitivity of hematological malignancies to graft-versus-host effects: an EBMT megafile analysis.
Leukemia
2014
, vol. 
28
 
11
(pg. 
2235
-
2240
)
38
Zuckerman
 
T
Katz
 
T
Haddad
 
N
, et al. 
Allogeneic stem cell transplantation for patients with chronic myeloid leukemia: risk stratified approach with a long-term follow-up.
Am J Hematol
2012
, vol. 
87
 
9
(pg. 
875
-
879
)
39
Poiré
 
X
Artz
 
A
Larson
 
RA
, et al. 
Allogeneic stem cell transplantation with alemtuzumab-based conditioning for patients with advanced chronic myelogenous leukemia.
Leuk Lymphoma
2009
, vol. 
50
 
1
(pg. 
85
-
91
)
40
Arpinati
 
M
Tolomelli
 
G
Bochicchio
 
MT
, et al. 
Molecular monitoring of BCR-ABL transcripts after allogeneic stem cell transplantation for chronic myeloid leukemia.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
5
(pg. 
735
-
740
)
41
Kaeda
 
J
O’Shea
 
D
Szydlo
 
RM
, et al. 
Serial measurement of BCR-ABL transcripts in the peripheral blood after allogeneic stem cell transplantation for chronic myeloid leukemia: an attempt to define patients who may not require further therapy.
Blood
2006
, vol. 
107
 
10
(pg. 
4171
-
4176
)
42
Heaney
 
NB
Copland
 
M
Stewart
 
K
, et al. 
Complete molecular responses are achieved after reduced intensity stem cell transplantation and donor lymphocyte infusion in chronic myeloid leukemia.
Blood
2008
, vol. 
111
 
10
(pg. 
5252
-
5255
)
43
Carpenter
 
PA
Snyder
 
DS
Flowers
 
ME
, et al. 
Prophylactic administration of imatinib after hematopoietic cell transplantation for high-risk Philadelphia chromosome-positive leukemia.
Blood
2007
, vol. 
109
 
7
(pg. 
2791
-
2793
)
44
Nakasone
 
H
Kanda
 
Y
Takasaki
 
H
, et al. 
Kanto Study Group for Cell Therapy
Prophylactic impact of imatinib administration after allogeneic stem cell transplantation on the incidence and severity of chronic graft versus host disease in patients with Philadelphia chromosome-positive leukemia.
Leukemia
2010
, vol. 
24
 
6
(pg. 
1236
-
1239
)
45
Luo
 
Y
Lai
 
XY
Tan
 
YM
, et al. 
Reduced-intensity allogeneic transplantation combined with imatinib mesylate for chronic myeloid leukemia in first chronic phase.
Leukemia
2009
, vol. 
23
 
6
(pg. 
1171
-
1174
)
46
Goldman
 
JM
Majhail
 
NS
Klein
 
JP
, et al. 
Relapse and late mortality in 5-year survivors of myeloablative allogeneic hematopoietic cell transplantation for chronic myeloid leukemia in first chronic phase.
J Clin Oncol
2010
, vol. 
28
 
11
(pg. 
1888
-
1895
)
47
Goldman
 
JM
Melo
 
JV
Chronic myeloid leukemia—advances in biology and new approaches to treatment.
N Engl J Med
2003
, vol. 
349
 
15
(pg. 
1451
-
1464
)
48
Ullmann
 
AJ
Hess
 
G
Kolbe
 
K
, et al. 
Current results on the use of imatinib mesylate in patients with relapsed Philadelphia chromosome positive leukemia after allogeneic or syngeneic hematopoietic stem cell transplantation.
Keio J Med
2003
, vol. 
52
 
3
(pg. 
182
-
188
)
49
Olavarria
 
E
Ottmann
 
OG
Deininger
 
M
, et al. 
Chronic Leukaemia Working Party of the European Group of Bone and Marrow Transplantation (EBMT)
Response to imatinib in patients who relapse after allogeneic stem cell transplantation for chronic myeloid leukemia.
Leukemia
2003
, vol. 
17
 
9
(pg. 
1707
-
1712
)
50
Kim
 
YJ
Kim
 
DW
Lee
 
S
, et al. 
Cytogenetic clonal evolution alone in CML relapse post-transplantation does not adversely affect response to imatinib mesylate treatment.
Bone Marrow Transplant
2004
, vol. 
33
 
2
(pg. 
237
-
242
)
51
DeAngelo
 
DJ
Hochberg
 
EP
Alyea
 
EP
, et al. 
Extended follow-up of patients treated with imatinib mesylate (gleevec) for chronic myelogenous leukemia relapse after allogeneic transplantation: durable cytogenetic remission and conversion to complete donor chimerism without graft-versus-host disease.
Clin Cancer Res
2004
, vol. 
10
 
15
(pg. 
5065
-
5071
)
52
Kantarjian
 
HM
O’Brien
 
S
Cortes
 
JE
, et al. 
Imatinib mesylate therapy for relapse after allogeneic stem cell transplantation for chronic myelogenous leukemia.
Blood
2002
, vol. 
100
 
5
(pg. 
1590
-
1595
)
53
Savani
 
BN
Montero
 
A
Kurlander
 
R
Childs
 
R
Hensel
 
N
Barrett
 
AJ
Imatinib synergizes with donor lymphocyte infusions to achieve rapid molecular remission of CML relapsing after allogeneic stem cell transplantation.
Bone Marrow Transplant
2005
, vol. 
36
 
11
(pg. 
1009
-
1015
)
54
Bansal
 
D
Shava
 
U
Varma
 
N
Trehan
 
A
Marwaha
 
RK
Imatinib has adverse effect on growth in children with chronic myeloid leukemia.
Pediatr Blood Cancer
2012
, vol. 
59
 
3
(pg. 
481
-
484
)
55
Giona
 
F
Mariani
 
S
Gnessi
 
L
, et al. 
Bone metabolism, growth rate and pubertal development in children with chronic myeloid leukemia treated with imatinib during puberty.
Haematologica
2013
, vol. 
98
 
3
(pg. 
e25
-
e27
)
56
Hobernicht
 
SL
Schweiger
 
B
Zeitler
 
P
Wang
 
M
Hunger
 
SP
Acquired growth hormone deficiency in a girl with chronic myelogenous leukemia treated with tyrosine kinase inhibitor therapy.
Pediatr Blood Cancer
2011
, vol. 
56
 
4
(pg. 
671
-
673
)
57
de la Fuente
 
J
Baruchel
 
A
Biondi
 
A
, et al. 
International BFM Group (iBFM) Study Group Chronic Myeloid Leukaemia Committee
Managing children with chronic myeloid leukaemia (CML): recommendations for the management of CML in children and young people up to the age of 18 years.
Br J Haematol
2014
, vol. 
167
 
1
(pg. 
33
-
47
)
58
Andolina
 
JR
Neudorf
 
SM
Corey
 
SJ
How I treat childhood CML.
Blood
2012
, vol. 
119
 
8
(pg. 
1821
-
1830
)
59
Efficace
 
F
Baccarani
 
M
Breccia
 
M
, et al. 
GIMEMA
Health-related quality of life in chronic myeloid leukemia patients receiving long-term therapy with imatinib compared with the general population.
Blood
2011
, vol. 
118
 
17
(pg. 
4554
-
4560
)
60
Bevans
 
MF
Mitchell
 
SA
Barrett
 
JA
, et al. 
Symptom distress predicts long-term health and well-being in allogeneic stem cell transplantation survivors.
Biol Blood Marrow Transplant
2014
, vol. 
20
 
3
(pg. 
387
-
395
)
61
Bevans
 
M
 
Health-related quality of life following allogeneic hematopoietic stem cell transplantation. Hematology Am Soc Hematol Educ Program. 2010;2010:248-254
62
Millot
 
F
Guilhot
 
J
Baruchel
 
A
, et al. 
Impact of early molecular response in children with chronic myeloid leukemia treated in the French Glivec phase 4 study.
Blood
2014
, vol. 
124
 
15
(pg. 
2408
-
2410
)
63
Apperley
 
J
Issues of imatinib and pregnancy outcome.
J Natl Compr Canc Netw
2009
, vol. 
7
 
10
(pg. 
1050
-
1058
)
64
Pye
 
SM
Cortes
 
J
Ault
 
P
, et al. 
The effects of imatinib on pregnancy outcome.
Blood
2008
, vol. 
111
 
12
(pg. 
5505
-
5508
)
65
Kuwabara
 
A
Babb
 
A
Ibrahim
 
A
, et al. 
Poor outcome after reintroduction of imatinib in patients with chronic myeloid leukemia who interrupt therapy on account of pregnancy without having achieved an optimal response.
Blood
2010
, vol. 
116
 
6
(pg. 
1014
-
1016
)
66
Borgmann-Staudt
 
A
Rendtorff
 
R
Reinmuth
 
S
, et al. 
Fertility after allogeneic haematopoietic stem cell transplantation in childhood and adolescence.
Bone Marrow Transplant
2012
, vol. 
47
 
2
(pg. 
271
-
276
)
67
Loren
 
AW
Chow
 
E
Jacobsohn
 
DA
, et al. 
Pregnancy after hematopoietic cell transplantation: a report from the late effects working committee of the Center for International Blood and Marrow Transplant Research (CIBMTR).
Biol Blood Marrow Transplant
2011
, vol. 
17
 
2
(pg. 
157
-
166
)
68
Wu
 
KN
Luo
 
Y
Liu
 
LZ
, et al. 
Twin pregnancy and childbirth after reduced-intensity conditioning allogeneic haematopoietic stem cell transplantation combined with imatinib mesylate for chronic myeloid leukaemia: case report and literature review.
J Int Med Res
2012
, vol. 
40
 
6
(pg. 
2409
-
2415
)
69
Kim
 
MK
Lee
 
DR
Han
 
JE
, et al. 
Live birth with vitrified-warmed oocytes of a chronic myeloid leukemia patient nine years after allogenic bone marrow transplantation.
J Assist Reprod Genet
2011
, vol. 
28
 
12
(pg. 
1167
-
1170
)
70
Gale
 
RP
Horowitz
 
MM
Ash
 
RC
, et al. 
Identical-twin bone marrow transplants for leukemia.
Ann Intern Med
1994
, vol. 
120
 
8
(pg. 
646
-
652
)
71
Barrett
 
AJ
Ringdén
 
O
Zhang
 
MJ
, et al. 
Effect of nucleated marrow cell dose on relapse and survival in identical twin bone marrow transplants for leukemia.
Blood
2000
, vol. 
95
 
11
(pg. 
3323
-
3327
)
72
Zheng
 
C
Tang
 
B
Yao
 
W
, et al. 
Comparison of unrelated cord blood transplantation and HLA-matched sibling hematopoietic stem cell transplantation for patients with chronic myeloid leukemia in advanced stage.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
12
(pg. 
1708
-
1712
)
73
Adekola
 
K
di Stasi
 
A
Ferro
 
R
, et al. 
Safety and efficacy of haploidentical stem cell transplantation for advanced chronic myeloid leukemia.
Biol Blood Marrow Transplant
2014
, vol. 
20
 
2
(pg. 
S213
-
S214
)
74
Experts in Chronic Myeloid Leukemia
The price of drugs for chronic myeloid leukemia (CML) is a reflection of the unsustainable prices of cancer drugs: from the perspective of a large group of CML experts.
Blood
2013
, vol. 
121
 
22
(pg. 
4439
-
4442
)
75
Ruiz-Argüelles
 
GJ
Tarin-Arzaga
 
LC
Gonzalez-Carrillo
 
ML
, et al. 
Therapeutic choices in patients with Ph-positive CML living in Mexico in the tyrosine kinase inhibitor era: SCT or TKIs?
Bone Marrow Transplant
2008
, vol. 
42
 
1
(pg. 
23
-
28
)
76
Ohm
 
L
Lundqvist
 
A
Dickman
 
P
, et al. 
Real-world cost-effectiveness in chronic myeloid leukemia: the price of success during four decades of development from non-targeted treatment to imatinib [published online ahead of print October 21, 2014].
Leuk Lymphoma
77
Goldman
 
JM
The significance of BCR-ABL transcripts after allogeneic stem cell transplantation for chronic myeloid leukemia.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
5
(pg. 
679
-
680
)
78
Cortes
 
J
Goldman
 
JM
Hughes
 
T
Current issues in chronic myeloid leukemia: monitoring, resistance, and functional cure.
J Natl Compr Canc Netw
2012
, vol. 
10
 
suppl 3
(pg. 
S1
-
S13
)
79
Mahon
 
FX
Réa
 
D
Guilhot
 
J
, et al. 
Intergroupe Français des Leucémies Myéloïdes Chroniques
Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial.
Lancet Oncol
2010
, vol. 
11
 
11
(pg. 
1029
-
1035
)
80
Goldman
 
J
Gordon
 
M
Why do chronic myelogenous leukemia stem cells survive allogeneic stem cell transplantation or imatinib: does it really matter?
Leuk Lymphoma
2006
, vol. 
47
 
1
(pg. 
1
-
7
)
81
Rojas
 
JM
Knight
 
K
Wang
 
L
Clark
 
RE
Clinical evaluation of BCR-ABL peptide immunisation in chronic myeloid leukaemia: results of the EPIC study.
Leukemia
2007
, vol. 
21
 
11
(pg. 
2287
-
2295
)
82
Rezvani
 
K
Yong
 
AS
Mielke
 
S
, et al. 
Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies.
Blood
2008
, vol. 
111
 
1
(pg. 
236
-
242
)
83
Rezvani
 
K
 
Posttransplantation vaccination: concepts today and on the horizon. Hematology Am Soc Hematol Educ Program. 2011;2011:299-304
84
Pinilla-Ibarz
 
J
Cathcart
 
K
Korontsvit
 
T
, et al. 
Vaccination of patients with chronic myelogenous leukemia with bcr-abl oncogene breakpoint fusion peptides generates specific immune responses.
Blood
2000
, vol. 
95
 
5
(pg. 
1781
-
1787
)
85
Bocchia
 
M
Defina
 
M
Aprile
 
L
, et al. 
Complete molecular response in CML after p210 BCR-ABL1-derived peptide vaccination.
Nat Rev Clin Oncol
2010
, vol. 
7
 
10
(pg. 
600
-
603
)
86
Weber
 
G
Gerdemann
 
U
Caruana
 
I
, et al. 
Generation of multi-leukemia antigen-specific T cells to enhance the graft-versus-leukemia effect after allogeneic stem cell transplant.
Leukemia
2013
, vol. 
27
 
7
(pg. 
1538
-
1547
)
87
Bornhäuser
 
M
Thiede
 
C
Platzbecker
 
U
, et al. 
Prophylactic transfer of BCR-ABL-, PR1-, and WT1-reactive donor T cells after T cell-depleted allogeneic hematopoietic cell transplantation in patients with chronic myeloid leukemia.
Blood
2011
, vol. 
117
 
26
(pg. 
7174
-
7184
)
88
Gill
 
S
Tasian
 
SK
Ruella
 
M
, et al. 
Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells.
Blood
2014
, vol. 
123
 
15
(pg. 
2343
-
2354
)
89
Ochi
 
T
Fujiwara
 
H
Suemori
 
K
, et al. 
Aurora-A kinase: a novel target of cellular immunotherapy for leukemia.
Blood
2009
, vol. 
113
 
1
(pg. 
66
-
74
)
90
Yong
 
AS
Stephens
 
N
Weber
 
G
, et al. 
Improved outcome following allogeneic stem cell transplantation in chronic myeloid leukemia is associated with higher expression of BMI-1 and immune responses to BMI-1 protein.
Leukemia
2011
, vol. 
25
 
4
(pg. 
629
-
637
)
91
Järås
 
M
Johnels
 
P
Hansen
 
N
, et al. 
Isolation and killing of candidate chronic myeloid leukemia stem cells by antibody targeting of IL-1 receptor accessory protein.
Proc Natl Acad Sci USA
2010
, vol. 
107
 
37
(pg. 
16280
-
16285
)
92
Herrmann
 
H
Sadovnik
 
I
Cerny-Reiterer
 
S
, et al. 
Dipeptidylpeptidase IV (CD26) defines leukemic stem cells (LSC) in chronic myeloid leukemia.
Blood
2014
, vol. 
123
 
25
(pg. 
3951
-
3962
)
93
Gooley
 
TA
Chien
 
JW
Pergam
 
SA
, et al. 
Reduced mortality after allogeneic hematopoietic-cell transplantation.
N Engl J Med
2010
, vol. 
363
 
22
(pg. 
2091
-
2101
)
Sign in via your Institution