You are on page 1of 9

Biol Blood Marrow Transplant 21 (2015) 412e420

Biology of Blood and


Marrow Transplantation
journal homepage: www.bbmt.org

Allogeneic Hematopoietic Cell Transplantation for


Myelodysplastic Syndromes: Lingering Uncertainties and
Emerging Possibilities
Sudipto Mukherjee 1, *, Dominic Boccaccio 2, Mikkael A. Sekeres 1, Edward Copelan 3
1
Leukemia Program, Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
2
Medical Humanities Department, Davidson College, Davidson, North Carolina
3
Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina

Article history: a b s t r a c t
Received 5 February 2014 The landscape of transplantation in myelodysplastic syndrome (MDS) has evolved rapidly in the last decade,
Accepted 25 July 2014 driven mostly by advances in patient selection through better risk stratification, increasing age of allogeneic
recipients, introduction of reduced-intensity conditioning regimens, increased availability of unrelated do-
Key Words: nors, new donor sources, and improvements in transplant technology and supportive care. Despite these
MDS advances, several issues, mostly centering on approaches to improve post-transplant survival while mini-
HCT mizing transplant-related mortality, continue to present significant challenges. Advances in understanding
Mutations
the molecular pathogenesis of MDS have made it feasible to construct clinically useful risk models that
Donor
integrate prognostic genes with conventional risk parameters for better selection of patients likely to benefit
Relapse
from hematopoietic cell transplantation. Simultaneous research efforts in several areas, including comor-
bidity assessment, novel preparative regimens, optimal pretransplant cytoreductive strategy, and post-
transplantation therapies, are expected to improve long-term disease-free survival and quality of life.
Ó 2015 American Society for Blood and Marrow Transplantation.

INTRODUCTION of pretransplant cytoreductive therapy, and the optimal


Myelodysplastic syndromes (MDS) is a heterogeneous timing of transplantation.
collection of clonal hematopoietic stem cell disorders char-
acterized by ineffective hematopoiesis leading to peripheral
cytopenias and related complications and a variable risk of EPIDEMIOLOGY OF TRANSPLANTATION IN MDS: A TALE
progression to acute myeloid leukemia (AML). According to OF UNMET NEEDS
Surveillance Epidemiology and End Results, MDS is the most Over the last decade, the number of transplants per-
common myeloid malignancy in the United States, with most formed for MDS has risen dramatically, particularly in older
patients (88%) older than 60 years at diagnosis [1]. Allogeneic adults, mainly because of increasing numbers of unrelated
hematopoietic cell transplantation (HCT) is the only known donor (URD) and reduced-intensity conditioning (RIC)
treatment modality with curative potential in MDS. Although transplants (Figure 1) [2]. This has been accompanied by
HCT outcomes in MDS have improved over the last decade, significant decreases in overall mortality (by 40%) and im-
several challenges pertaining to transplant-related mortality proved long-term survival because of decreased transplant-
(TRM) and morbidity remain. This review summarizes the related complications, notably infections, organ toxicities,
advances made in the last decade, focusing mainly on more and severe acute graft-versus-host disease (GVHD) [3]. These
nuanced disease risk stratification, transplant implications of results are even more promising considering that trans-
newly identified molecular mutations, the emerging role of planted cohorts in recent years are on average older by
haploidentical and umbilical cord blood (UCB) grafts, the role 2 decades compared with their counterparts from the late
1980s and the proportion of URD recipients increased from
6% to 57% in the same interval (exceeding related donors) [4].
The Center for International Blood and Marrow Trans-
Financial disclosure: See Acknowledgments on page 418. plant Research (CIBMTR) reports MDS as the third most
* Correspondence and reprint requests: Sudipto Mukherjee, MD, PhD, common indication for allogeneic HCT, with 1016 transplants
MPH, Associate Staff, Leukemia Program, Department of Hematologic
Oncology and Blood Disorders, Cleveland Clinic Taussig Cancer Institute,
registered between 2008 and 2010 [2]. A comparable trend
R35, 9500 Euclid Avenue, Cleveland, OH 44195. has been reported in Europe, where the European Group for
E-mail address: mukhers2@ccf.org (S. Mukherjee). Blood and Marrow Transplantation (EBMT) registered a

http://dx.doi.org/10.1016/j.bbmt.2014.07.027
1083-8791/Ó 2015 American Society for Blood and Marrow Transplantation.
S. Mukherjee et al. / Biol Blood Marrow Transplant 21 (2015) 412e420 413

cohort with the highest MDS rates [10]. Figures from 2012
show that a formal search was initiated for only 62% of these
patients of which only 60% actually proceeded to HCT [12].
Although the reasons for low utilization of HCT are
multifactorial, most of the concerns center around the high
risk of TRM or diminution of quality of life due to chronic
GVHD or other delayed effects of HCT, especially so in older
patients with diminished physiologic reserves because of the
combined effect of their advanced age and existing comor-
bidities. Limited understanding of referring physicians re-
garding transplant eligibility, late transplant referrals, delay
in HLA typing, and underuse of URD transplantation is also
responsible. The feasibility of surmounting some of these
barriers was demonstrated in a recent study in which a
combination of pragmatic strategies, including use of alter-
Figure 1. Number of allogeneic HCTs for MDS patients 65 years of age in the native donors, different graft sources, and RIC regimens
United States, 2005-2012. Data compiled by CIBMTR. Figure provided by Dr. (42%), allowed 67% of AML patients in complete remission to
Mary Horowitz and adapted with permission. proceed to HCT [13]. A concerted effort in educating the
clinicians providing care for MDS patients would result in
doubling in the number of allogeneic HCT for MDS/second- more frequent and earlier consideration of this modality.
ary AML (sAML) from 737 to 1636 between the years 2001
and 2010, paralleled by a 10% to 33% increase in HCT use in
patients over age 60 [5]. Although these temporal trends are WHO NEEDS A TRANSPLANT? SEPARATING THE GOOD
impressive, when considered in the context of the entire pool FROM THE BAD AND THE UGLY
of high-risk MDS patients in the United States, statistics from Selection of the appropriate transplant candidate has
various epidemiologic sources suggest a staggeringly low use undergone refinements over the past decade, aided by
of transplantation in this population, as shown in Figure 2 improved understanding of disease risk, use of disease-
[6-10]. A prospective feasibility study of RIC HCT in 259 modifying therapies that improve survival (since 2004),
higher risk MDS/AML patients reported that only 14 patients more sensitive tools to assess transplant vulnerability, and
(5%) harboring unfavorable cytogenetics eventually received increased HCT opportunities in the elderly with the advent of
transplantation [11]. The findings of a cross-sectional survey RIC and nonmyeloablative (NMA) regimens. As shown in
of US physicians from different geographic regions and a Table 1, upfront HCT is currently recommended for fit MDS
wide variety of practice settings are even more sobering, patients stratified as higher risk based on clonal chromo-
with only 4% of recently diagnosed MDS patients referred for somal abnormalities, blast percentage, peripheral cytope-
HCT [8]. With 21 million potential marrow donors world- nias, and additional features depending on the particular
wide, including more than 600,000 UCB units, it is expected prognostic model applied. Originally designed to prognosti-
that a suitable URD or UCB unit can be found to meet the cate for nontransplanted patients, several risk models, such
needs of approximately 12,000 patients in the United States as the International Prognostic Scoring System (IPSS), the
who need URD transplantations every year. Despite this and World Health Organization classification-based Prognostic
results comparable with those with HLA-identical sibling Scoring System (WPSS), and the revised IPSS (IPSS-R), also
donors, the estimated usage of these donor sources is strik- predict survival and relapse in the transplant setting
ingly low, at 10% for 65- to 74-year-olds, the demographic [4,14,15], even in those receiving RIC [14,15]. The newly

Figure 2. Crude estimation of transplant-eligible older MDS patients (age >60-65 years) in the United States with data extrapolated from contemporaneous pop-
ulation registries highlighting the staggeringly low uptake of allogeneic HCTs in this age group. SEER indicates Surveillance Epidemiology and End Results; NAACCR,
North American Association of Central Cancer Registries; NHANES III, National Health and Nutrition Examination Survey III; alloHSCT, allogeneic hematopoietic stem
cell transplantation. * MDS cases equal to or greater than 60 years were extracted from all 18 SEER registries using the R package SEERaBomb available at http://
cran.r-project.org/web/packages/SEERaBomb/index.html (courtesy of Dr. Tomas Radivoyevitch).
414 S. Mukherjee et al. / Biol Blood Marrow Transplant 21 (2015) 412e420

Table 1 ASXL1, the median survival was significantly shorter than


Indications for Transplantation in MDS Based on Different Risk Assessments predicted by IPSS and resembled that of the next highest IPSS
Definite Prognostic modelebased risk score risk group [19]. In lower risk MDS patients (low and inter-
IPSS intermediate-2 mediate risk-1 [IR-1] IPSS risk), presence of EZH2, RUNX1, and
IPSS high ASXL1 independently predicted for higher risk of death
IPSS-R high risk
IPSS-R very high risk
compared with unmutated counterparts even after adjusting
WPSS high risk for IPSS [20]. In addition, an increasing number of oncogenic
WPSS very high risk driver mutations in an individual patient also predicted for
Disease morphology deterioration in leukemia-free survival [21]. None of the risk
RAEB-1, RAEB-2
models in clinical use has yet incorporated these mutations in
RAEB-t
Probable Presence of poor prognostic features (clinical and/or their prognostic algorithm, resulting in the likelihood of a
pathologic) substantial number of high-risk patients being treated
Failure of HMA less aggressively because of inaccurate risk assignment.
Transfusion dependency despite growth factor support Mutation-based models based solely on molecular markers
Severe neutropenia/thrombocytopenia (even with low
IPSS scores)
can identify prognostic risk groups with different survival
High-risk cytogenetic abnormalities (IPSS-R)* probabilities [18,21], and when combined with IPSS-variables
Secondary/therapy-related MDS have been shown to enhance prognostication [18-21].
IPSS indicates International Prognostic Scoring System; WPSS, the World The impact of these mutations on post-transplant out-
Health Organization classification-based Prognostic Scoring System; IPSS-R, come in MDS patients was investigated by Thol et al. [22],
revised IPSS; RAEB-1, refractory anemia with excess blasts-1; RAEB-2, re- who analyzed the impact of the 3 most commonly mutated
fractory anemia with excess blasts-2; RAEB-t, refractory anemia with excess spliceosomal genes, U2AF1, SRSF2, and SF3B1, in 339 patients
blasts in transformation.
* inv(3)/t(3q)/del(3q), 7; double abnormalities including 7/del(7q),
transplanted for MDS/sAML. Only U2AF1 mutations inde-
and complex (3 abnormalities). pendently predicted for shorter OS (P ¼ .011) and higher cu-
mulative incidence of relapse (P ¼ .007). Hamilton et al. [23]
proposed 5-group cytogenetic risk classification has greater reported a survival advantage in patients with U2AF1 or
discriminatory capacity in predicting post-HCT relapse and SRSF2 mutations who underwent upfront HCT compared with
mortality than the 3 IPSS risk categories and reassigns risk of those who received high-intensity chemotherapy or hypo-
65% of “classic” IPSS patients [4]. Applying the 5-group methylating agents (HMAs) (53 versus 17 months, P ¼ .05). No
criteria, the IPSS poor-risk cytogenetics group can be strati- difference in OS, relapse, and nonrelapse mortality (NRM)
fied into 3 additional risk groups, intermediate, poor, and was reported between wild-type and mutated SRSF2 patients,
very poor, with high relapse rates of 30% and 50% for the suggesting HCT can abrogate the adverse effects of SRSF2 [24].
latter two cohorts. Mutations in TP53, TET2, and DNMT3A have been signifi-
In a large series of 519 patients transplanted for MDS or cantly associated with shortened survival following HCT after
oligoblastic AML (<30% marrow blasts), Gruppo Italiano adjusting for known adverse disease and transplant-related
Trapianto di Midollo Osseo (GITMO) confirmed the high-risk predictive variables (Dr. Rafael Bejar, personal communica-
IPSS-R category as an independent predictor of relapse and tion). Patients with TP53 mutations in particular had worst
lower survival [15]. Additionally, the presence of monosomal outcomes, with a median post-transplant survival of only
karyotype, particularly those with chromosome 7 abnor- 4.6 months, raising questions about any benefit of HCT in this
mality (even when present as an isolated abnormality), group. As shown in Figure 3, several mutations predict for
significantly worsens prognosis beyond that of a complex poor survival in a population of lower risk patients, and such
karyotype [4,16]. In one series, no patient with the mono- patients should be treated as high risk and considered for
somal karyotype was alive at 5 years post-transplant early HCT, preferably in the context of clinical trials because
compared with the 5-year overall survival (OS) rate of 37% prospective data are lacking.
in those without the monosomal karyotype [16]. These data
suggest that standard HCT approaches may be futile in some DO ALL THERAPY-RELATED MDS/AML PATIENTS NEED
high-risk patients, and clinical trials might be a more rational TRANSPLANTATION? NOT ALL CURES ARE POISON
approach. Emerging data support early HCT in certain sub- Another question that has garnered increasing attention
sets of lower risk IPSS patients who exhibit an aggressive is whether HCT should be recommended for all therapy-
clinical course characterized by severe thrombocytopenia, related MDS/AML (t-MDS/AML) patients. With overre-
transfusion dependency, or marrow fibrosis [17]. presentation of poor-risk cytogenetic features including,
chromosome 5 and/or 7 abnormalities in 70% of t-MDS/AML
DEFINING RISK BEYOND CYTOGENETICS: WAR OF THE patients and median survival of 6 to 8 months, trans-
CLONES plantation alone has been thought to offer long-term dis-
High-throughput sequencing analysis has revealed the ease-free survival. However, this concept is challenged by
complex genomic landscape of MDS with identification of data that suggest the biology and outcome of t-MDS/AML
recurrent somatic mutations in over 40 genes involved in depends on the treatment modality originally received. The
several pathways, including signal transduction, epigenetic outcomes of transplanted t-MDS/AML patients reported to
regulation, transcriptional regulation, and RNA splicing, CIBMTR and EBMT is reflective of exposure to chemotherapy
among others. These somatic mutations are highly prevalent, or combined modality, considering 82% and 95% of the entire
affecting a staggering 90% of MDS patients, including 50% to cohort in the registries, respectively, had such an exposure
70% of patients with normal cytogenetics [18,19]. Several [25,26]. Nardi et al. [27] reported that MDS developing after
of these mutations have been shown to independently influ- radiation monotherapy behaves like de novo disease, with no
ence survival in nontransplant setting [18-21] and can upstage significant differences in median survival (38 versus 30
IPSS or IPSS-R predicted disease risk. For patients harboring months) and a similar prevalence of chromosome 5 and 7
mutations in any of the 5 genes TP53, EZH2, ETV6, RUNX1, and abnormalities in both groups [27]. When compared with
S. Mukherjee et al. / Biol Blood Marrow Transplant 21 (2015) 412e420 415

considerable controversy exists in older adults. The identi-


fication of a patient as “elderly” in the transplantation liter-
ature varies by study, with most referring to patients aged 50
to 70 years. With advancing patient age, increased rates of
TRM, particularly with myeloablative (MA) conditioning
regimens, have been observed [30]. However, an accumu-
lating body of evidence shows that HCT can be performed
safely in older individuals selected based on limited
comorbidities.
McClune et al. [31] analyzed 1080 patients with MDS
(n ¼ 535) or AML in first complete remission (n ¼ 545),
identified from the CIBMTR, who underwent RIC or NMA
conditioning followed by HCT between 1995 and 2005. No
significant difference in 2-year risk of NRM, relapse, disease-
free survival, OS, or GVHD was observed across 4 age cohorts:
40 to 54 years, 55 to 59 years, 60 to 65 years, and >65 years.
Lim et al. [32] performed a similar analysis of a contempo-
raneous cohort of 1333 MDS patients registered in the EBMT
who received MA (n ¼ 500) or RIC (n ¼ 833) transplantation
between 1998 and 2006. When compared with the 50- to 59-
year age group, patients >60 years had comparable NRM
(36% versus 39%), relapse (32% versus 41%), and OS (34%
versus 27%) at 4 years. Although advanced disease stage at
the time of transplantation was the most important prog-
nostic variable for OS in the EBMT series, in the CIBMTR study
performance status, HLA disparity, and unfavorable cytoge-
netics predicted for poor OS and disease-free survival. Of
note, only 34% and 10% of transplanted MDS patients in the
CIBMTR and EBMT series, respectively, were over age
60 years, likely identifying a selection bias. Two studies in
septuagenarians with MDS observed no increase in adverse
transplant-related outcomes [30,33]. Although chronologic
Figure 3. (A) Comparison of median survival of MDS patients stratified by age should not be an exclusion criterion for transplantation,
IPSS-R cytogenetic risk categories or based on presence of prognostic molec-
it must be considered in decision making because of the
ular mutations. Median survival predicted by the molecular mutations as
shown in the figure were adjusted for the IPSS risk group assigned at the time increased prevalence of comorbidities and age-related
of sample collection. IPSS-R Int, Poor and Very Poor indicate intermediate, decline in organ function.
poor, and very poor cytogenetic categories in revised International Prognostic
Scoring System (IPSS-R). (B) Comparison of overall survival in patients with
IMPACT OF COMORBIDITIES: FULFILLING THE
any of the prognostically adverse somatic mutations (in TP53, EZH2, RUNX1,
ASXL1, or ETV6) to unmutated patients within each of the IPSS-R “lower” risk
DARWINIAN RULE (THE FITTER, THE BETTER)
groups identifies added disease risk in each of the categories. Dotted arrows Several pretransplant scoring systems objectively char-
indicate decrease in survival in the presence of any of the 5 mutations. (Mo- acterize transplant vulnerability and predict outcomes. The
lecular data provided by Dr. Rafael Bejar and adapted with permission. Figures most widely accepted is the HCT Comorbidity Index (HCT-CI),
courtesy of Dr. Aaron Gerds.)
which reliably predicts toxicities, NRM, and OS in MDS or
AML patients receiving MA or RIC transplants [34]. An HCT-CI
patients treated with chemotherapy alone or in combination composite score >3 predicted for an exceedingly high 2-year
with radiation, those who developed MDS after definitive NRM (hazard ratio, 5.35) and inferior 2-year OS (hazard ratio,
radiotherapy had better OS rates and a lower incidence of 5.27). To further refine risk prediction, Sorror et al. [35]
high-risk karyotypes. New data also raise question about proposed a modified HCT-CI that integrated the classic
whether all radiation-related MDS cases are truly t-MDS. A HCT-CI scores with disease risk determined by morphology,
retrospective study of 11,000 prostate cancer patients remission status, and cytogenetics. Low disease risk, mostly
showed no excess risk of MDS in those who underwent refractory anemia and refractory anemia with ring side-
radiation monotherapy (n ¼ 5119) compared with those roblasts with an HCT-CI of 0 to 2, predicted for best outcome,
treated with radical prostatectomy (the control group) and with a low NRM of 4% and 11% and a high 2-year OS of 70%
with population-based registries [28]. Analogous to findings and 78% after NMA and MA conditioning, respectively. Those
from the t-AML literature [29] and taking into account the with intermediate-/high-risk disease and HCT-CI >3 fared
fact that these patients may have transplant vulnerabilities worst, with NRM of 29% and 46% after NMA and MA condi-
from cumulative toxicities of prior therapies, it might be tioning, respectively and corresponding OS of 29% and 24%,
reasonable to treat radiation-related MDS, and perhaps other respectively.
t-MDS patients, without high-risk cytogenetic features like Impaired functional status, as measured by the Karnofsky
de novo lower-risk disease using nontransplant approaches. performance scale (KPS) or the Eastern Cooperative Oncology
Group scale, independently predicts for worse transplant
IMPACT OF RECIPIENT AGE: BIOLOGY TRUMPS outcome. Typically, published studies have used KPS score
CHRONOLOGY <70% or Eastern Cooperative Oncology Group score of 2 or
Although there is broad consensus for recommending less as exclusion criteria for transplantation. However, these
HCT for appropriate higher-risk younger MDS patients, “cutoff” criteria may not entirely capture transplant
416 S. Mukherjee et al. / Biol Blood Marrow Transplant 21 (2015) 412e420

vulnerability, as shown in the CIBMTR registry where 16% of HMAs (36 versus 28 months) with this benefit apparent only
patients 65 years or older fared poorly despite KPS score after 40 months of follow-up.
>70%. To better characterize transplant risk beyond HCT-CI Two contemporaneous European registry analyses, how-
and performance scales, Muffly et al. recently proposed use ever, showed differing results [42,43]. The GITMO-Pavia
of a comprehensive geriatric assessment tool that uncovered study adopted a continuous-time multistate Markov model
a high prevalence of impairments in functional status, frailty, using IPSS and WPSS risk categories as time-dependent in-
disability, and mental health in HCT recipients >50 years old dicators of the natural course of MDS to estimate and
who were assessed as fit by standard risk criteria [36]. compare life expectancy from diagnosis [42]. This analysis
The negative impact of iron overload on transplant failed to show any survival advantage by delaying transplant
outcome in transfusion-dependent MDS patients has been a in the IR-1 group, a significant finding considering that almost
subject of ongoing debate. In contrast to retrospective ana- 50% of the patients in the Cutler study [40] had IR-1 disease.
lyses that evaluated HCT outcomes based on serum ferritin Based on the best WPSS-based transplantation strategy in
levels, a recent prospective study used pretransplant mag- this model, lower-risk IPSS patients with multilineage
netic resonance imagingequantified hepatic iron concen- dysplasia and/or transfusion dependency benefit from early
tration as a marker of hyperferritinemia and reported no HCT, whereas outcomes are better if transplant is delayed for
effect on OS or NRM [37]. Whether pretransplant iron che- IR-1 patients who have favorable cytogenetics or those
lation can improve transplant outcomes has never been without excess blasts. A recent EBMT study used several
prospectively studied. statistical models to compare treatment outcomes of older
MDS patients (>55 years) with refractory anemia with excess
blasts or refractory anemia with excess blasts in trans-
TIMING OF TRANSPLANTATION: BRINGING OUT THE formation who underwent transplantation (EBMT registry) or
BIG GUNS were managed with nontransplant strategies (Düsseldorf
Although early HCT is indicated in patients with higher- Registry) and reported no survival advantage with HCT [43].
risk MDS provided the recipient is fit and a donor available, Notably, this study exposed the limitation of drawing con-
the optimal timing of transplantation remains a clinical clusions about the causal effect of time interval from diag-
challenge in many MDS patients, especially with the intro- nosis to transplant on outcome using observational data, even
duction of HMAs that improve survival and the availability of with the most sophisticated mathematical models.
clinical trials. Although not routinely recommended, there is The unavoidable flaw in these models is the reliance on
evidence of high success rates with modest toxicity when crucial assumptions that do not reflect nonproportional dis-
HCT is performed in early-stage disease or low-risk IPSS tribution of hazards (related to patient and disease-risk pa-
categories [38,39]. Analysis of EBMT registry data showed an rameters) among the comparator groups. The question of
absolute increase in OS of 10% at 4 years in patients with re- whether there is any survival advantage with transplant in
fractory anemia and refractory anemia with ring sideroblasts older adults is being addressed prospectively by a phase III
who underwent earlier transplantation (<12 months) from trial (BMT CTN 1102) that will biologically assign patients aged
the time of diagnosis, mainly because of lower NRM [38]. 50 to 75 years with IPSS IR-2 and high-risk disease to RIC HCT
Similar outcomes were reported by Deeg et al. [39] in their or non-HCT treatment (HMAs or best supportive care) based
cohort of IPSS low-risk patients with no reported relapses and on availability of suitable matched related donor or URD.
80% relapse-free survival compared with corresponding fig- Transplant decisions present additional challenges in
ures of 42% and 29% observed in the high-risk group. These patients on HMA therapy. Clinical responses to HMA are seen
single-arm studies are inherently limited by selection bias, after 4 to 6 months of therapy, and of the roughly 50% of
and in the absence of any prospective data comparing HCT patients who respond, the median duration of response is
with nontransplantation approaches, it remains unclear only 9 to 15 months, with most losing response within 1 to
whether transplantation early in disease course confers any 2 years [44,45]. In those who fail azacitidine, the median
survival advantage. GITMO experience indicates optimal survival is dismal at <6 months [46]. This raises several
benefit of HCT when performed before progression of disease questions. Is it possible to identify patients who have a low
to an advanced stage [14]. probability of responding to HMAs and consider them for
Results of a Markov decision analysis showed a beneficial upfront HCT or other alternative approach? In those who are
role of early HCT in younger patients with IR-2 or high-risk responders, should HMAs be continued until disease pro-
IPSS disease undergoing MA transplantation with HLA- gression or should they be transplanted while they are in
identical sibling grafts, whereas delaying transplant until remission?
leukemic evolution led to longer life expectancy in patients Based on recent evidence, patients harboring TET2 and or
with low and IR-1 disease [40]. However, the study findings DNMT3A mutations are more likely to respond to HMAs, but
are not applicable to most MDS patients typically seen in this has not been validated prospectively [47,48]. Deferring
daily clinical practice because the data pertain to an exclu- transplant until the time of disease progression can jeopar-
sively young cohort (<60 years) and predated the era of dize HCT chances because of several factors, notably un-
HMAs. To address these limitations, Koreth et al. [41] used a controlled disease, acquisition of highly resistant clonal
similar statistical approach to compare the outcome of 2 abnormalities and comorbidities, or treatment-related tox-
different treatment arms, RIC HCT and nontransplantation icities accumulated over the course of disease. Long-term
strategies (best supportive care, hematopoietic growth fac- disease-free survival (median of 19.5 months) with HCT can
tors, and HMAs), in patients >60 years. This analysis similarly be achieved in only a small subset of patients who fail HMA
showed best survival with nontransplantation strategies in [46]. Among the azacitidine-treated cohort, a nonsignificant
older patients with lower-risk MDS with near doubling of improvement in median survival was seen in those who had
median life expectancy compared with HCT (77 versus stable disease at the time of azacitidine discontinuation
38 month). For higher-risk disease, only a modest survival compared with those who underwent transplantation with
advantage was seen with earlier transplant compared with progressive disease (not reached versus 17 months) [46].
S. Mukherjee et al. / Biol Blood Marrow Transplant 21 (2015) 412e420 417

Thus, consultation to discuss HCT should be routinely offered patients who underwent transplantation after chemo-
to higher-risk patients who are transplant eligible shortly therapy alone, HMAs, both, or no prior therapy. Jabbour et al.
after diagnosis. Early transplantation without exposure to [63] did not observe any benefit of HCT after HMA therapy
pretransplant agents in patients with higher-risk disease compared with upfront HCT. Gerds et al. presented data on
who are willing and able is reasonable. 68 MDS and sAML patients pretreated with AZA (n ¼ 35) or
IC (n ¼ 33) prior to allo-HSCT and similarly reported no dif-
DONOR SELECTION: SOMETHING FOR MISMATCHED, ference in 1 year survival, NRM and relapse between the
UNRELATED, AND HAVE NOTS groups [64]. The failure to demonstrate a benefit for therapy
With high-resolution HLA typing, outcomes of 8/8 before transplantation suggests that high-risk MDS patients
matched URD recipients are comparable with HLA-identical should, where possible, proceed directly to transplant
siblings, with the exception of slightly higher rates of acute without delay [62]. Because rates of complete remission are
and chronic GVHD [49]. The question of whether a younger higher with induction chemotherapy than with HMA, this
matched unrelated donor (MUD) rather than an older MSD should be considered as an option to rescue younger fit pa-
improves outcome was addressed in an EBMT series of 719 tients with high disease burden. However, for those with
MDS patients (>50 years), showing a significant survival poor-risk cytogenetic abnormalities, evidence suggests use
advantage with allografts originating from younger MUDs of HMAs over induction chemotherapy is reasonable. In older
(<30 years) compared with older MUDs or MSDs [50]. In individuals with comorbidities awaiting RIC HCT, use of
contrast, CIBMTR analysis reported better outcomes with HMAs as “bridging therapy” to prevent disease progression is
older matched related donor (>50 years) than young URD an acceptable approach.
(<50 years), particularly in recipients with poor pretrans-
plant performance scores [51]. The preferential source of
hematopoietic progenitor cells for URD transplantation was PREPARATIVE REGIMENS: HOW MUCH MARROW
addressed in an international randomized trial that demon- ABLATION IS ENOUGH?
strated comparable survival rates but a higher incidence of Few studies confined to MDS patients have directly
chronic GVHD with peripheral blood stem cells compared compared the effectiveness and toxicity of specific condi-
with bone marrow grafts [52]. We favor the use of sibling tioning regimens. Traditionally, MA preparation using total
donors with peripheral blood stem cells because of earlier body irradiation (TBI)- or busulfan-based regimens have
engraftment and better quality of life. been used. Of adults with MDS who underwent HCT from
In the absence of a suitable MSD or URD, unrelated UCB HLA-MSD or adult URD in the United States between 2002
cells or cells from HLA-haploidentical family donors provide and 2006 reported from the CIBMTR, nearly 60% received MA
reasonable alternatives. Majhail et al. [53] assessed the regimens [49]. RIC, however, has been increasingly used over
feasibility of UCB transplants in 98 older MDS/AML patients the last few years.
(>55 years) by prospectively comparing outcomes between Although TBI-based MA regimens are effective and were
4-6/6 HLA-matched UCB and MSD after RIC HCT and reported predominantly used for patients with MDS until the last
no significant difference in 3-year OS (37% versus 31%), decade [58,65], the use of busulfan-based regimens has
relapse, or TRM. In single UCB transplantation, higher cell progressively increased [39,66]. Advances in busulfan
doses predict for improved survival [54], and in cases of low administration, including i.v. administration [67] and/or
cell doses, alternative approaches include infusion of double targeted plasma levels based on pharmacokinetics, have
cord blood units or a single cord blood unit after in vitro improved results [39]. Deeg et al. [39] combined oral
expansion [55,56]. Outcomes similar to matched related busulfan targeted to predetermined plasma levels in com-
donor and MUD recipients have been reported with hap- bination with cyclophosphamide in 109 patients with MDS
loidentical transplants using T cellereplete grafts and high- receiving hematopoietic cells from related or unrelated do-
dose post-transplantation cyclophosphamide after NMA nors. NRM at 100 days and 3 years were 12% and 28% for
conditioning [57]. The low incidence of chronic GVHD after related and 13% and 30% for unrelated recipients. Relapse-
post-transplant cyclophosphamide is a particular advantage free survival at 3 years was 56% and 59%, respectively, and
of this approach. this regimen appeared to be better tolerated and safer than
traditional TBI regimens, including in patients older than
PRETRANSPLANT CYTOREDUCTION: TAMING THE ROGUE 60 years of age [39].
MARROW Intravenous administration of busulfan is associated with
Pretransplant blast burden (>5% marrow blasts) and decreased incidences of hepatic veno-occlusive disease and
failure to achieve complete remission significantly correlate early mortality [67]. A recently reported large retrospective
with relapse in MDS patients undergoing HCT, particularly analysis from the CIBMTR of AML patients in first remission
with RIC regimens [58,59]. Retrospective studies examining who underwent transplantation between 2000 and 2006
the role of induction chemotherapy before HCT showed no demonstrated significantly lower NRM, relapse-free survival,
convincing benefit [60]. Use of HMAs as a bridging therapy to and OS rates using i.v. busulfan (1-year NRM of 12%)
HCT offers an alternative to induction chemotherapy to compared with TBI in combination with cyclophosphamide
reduce blast burden with fewer treatment-related toxicities. [68]. A subsequent cohort of patients undergoing trans-
The French Society of Bone Marrow Transplantation and Cell plantation between 2008 and 2011, including patients with
Therapy conducted the largest study on the use of azacitidine MDS, also showed better survival in patients receiving i.v.
before HCT and reported no significant differences in out- busulfan compared with TBI [69]. An MA regimen of busulfan
comes among patients who received azacitidine or induction combined with fludarabine has gained popularity because
chemotherapy before transplantation [61]. It also found that of reported decreased toxicity compared with busulfane
patients who required both azacitidine and induction cyclophosphamide; however, a randomized study in AML
chemotherapy had poorer outcomes. Oran et al. [62] found indicated higher relapse rates and lower survival compared
no difference in event-free survival among high-risk MDS with busulfanecyclophosphamide [70].
418 S. Mukherjee et al. / Biol Blood Marrow Transplant 21 (2015) 412e420

RIC regimens have extended the use of allogeneic GVHD [81]. Treatment decision based on detection of mini-
transplantation to selected older patients and those with mal residual disease after transplantation is a growing area
comorbidities that place them at high risk of TRM with of research. Platzbecker et al. [76] reported monitoring
MA preparation. For patients with MDS, lower intensity recipient CD34þ lineage-specific chimerism as a marker of
conditioning is associated with higher risk of relapse [71- minimal residual disease to preemptively start treatment
73], lower NRM, and generally similar survival to MA with azacitidine [76]. With the discovery of prognostic
regimens [73]. Nonablative regimens, however, have been molecular markers in MDS, it is expected that these will
associated with substantially higher relapse rates and increasingly become an integral part of minimal residual
lower relapse-free survival and OS than MA conditioning diseaseebased treatment strategies.
[5,75]. RIC regimens generally including lower than MA
doses of busulfan or melphalan combined with fludar- CONCLUSION
abine can cure a significant proportion of patients with Although the use of HCT in MDS has become safer
MDS who would be at high risk of mortality with MA and more widespread, it remains underused. Clinicians
therapy. Other regimens, including 550-cGy TBI with should be cognizant of the newly identified molecular
cyclophosphamide, have also proven to be well tolerated mutations that can help identify patients who would
and effective [74]. benefit from transplantation but who are outside the
A retrospective study from the EBMT reported that 38% paradigm of current decision models. Future efforts to
of patients older than 50 years with MDS received an MA improve transplant outcomes in MDS should focus on
regimen [32]. No difference in NRM or survival was incorporating molecular data into treatment algorithms
detected in this analysis among patients <60 years of age for accurate and earlier identification of higher-risk MDS
compared with those 60 years. Because relapse remains a patients, initiating earlier donor search, use of alternative
particular concern in patients with 5% marrow blasts, donors, and modifying treatment strategies regarding
adverse cytogenetics, or molecular mutations, MA therapy pretransplant cytoreduction and conditioning regimen
may be appropriate in selected patients up to at least that can abrogate the effects of high risk chromosomal
70 years of age [5,71]. At present, we favor the use of MA abnormalities.
i.v. busulfan-based regimens in patients without significant
comorbidities and RIC regimens in those with significant ACKNOWLEDGMENTS
comorbidities, particularly when accompanied by lower- Financial disclosure: The authors have nothing to disclose.
risk MDS. Well-designed prospective studies to analyze Conflict of interest statement: There are no conflicts of in-
the relative effectiveness and toxicity of specific MA and terest to report.
RIC regimens in patients at varied risk of relapse and TRM
are needed. Two randomized trials were designed to pro- REFERENCES
spectively compare toxicity and efficacy between patients 1. Howlader N, Noone AM, Krapcho M, et al. SEER cancer statistics review.
receiving high-intensity and reduced-intensity regimens: Bethesda, MD: National Cancer Institute; 1975-2010.
2. Pasquini MC, Wang Z. Current use and outcome of hematopoietic stem
EBMT NCT00682396 and BMT CTN 0901 NCT01339910. cell transplantation: CIBMTR Summary Slides, 2013. Available at:
Preliminary review suggesting a benefit of the MA arm http://www.cibmtr.org
over RIC regimens led to the early closure of the BMT CTN 3. Gooley TA, Chien JW, Pergam SA, et al. Reduced mortality after allo-
geneic hematopoietic-cell transplantation. N Engl J Med. 2010;363:
0901 trial. 2091-2101.
4. Deeg HJ, Scott BL, Fang M, et al. Five-group cytogenetic risk classifi-
POST-TRANSPLANTATION RELAPSE cation, monosomal karyotype, and outcome after hematopoietic cell
transplantation for MDS or acute leukemia evolving from MDS. Blood.
After relapse prognosis is extremely poor, especially if 2012;120:1398-1408.
relapse occurred within a year of HCT. In the absence of any 5. Kröger N. Allogeneic stem cell transplantation for elderly patients with
randomized trials, there is no clear consensus about post- myelodysplastic syndrome. Blood. 2012;119:5632-5639.
6. Rollison DE, Howlader N, Smith MT, et al. Epidemiology of myelodys-
transplant treatment strategies. Several efforts to reduce
plastic syndromes and chronic myeloproliferative disorders in the
relapse rates after transplantation include withdrawal of United States, 2001-2004, using data from the NAACCR and SEER
immunosuppression, HMAs [76,77], infusion of selected programs. Blood. 2008;112:45-52.
populations of donor lymphocytes [78], and, occasionally, a 7. Cogle CR, Craig BM, Rollison DE, List AF. Incidence of the myelodys-
plastic syndromes using a novel claims-based algorithm: high number
second HCT. It is postulated that azacitidine can enhance the of uncaptured cases by cancer registries. Blood. 2011;117:7121-7125.
graft-versus-leukemia effect by increased expression of 8. Sekeres MA, Schooen M, Kantarijan H, et al. Characteristics of US pa-
cancer-testis antigens [79] that become potential targets for tients with myelodysplastic syndromes: results of six cross-sectional
physician surveys. J Natl Cancer Inst. 2008;100:1542-1551.
the donor immune system and hence can play a role in dis- 9. Guralnik JM, Eisenstaedt RS, Ferrucci L, et al. Prevalence of anemia in
ease control after transplant. The role of azacitidine for persons 65 years and older in the United States: evidence for a high
treating relapse was studied by de Lima et al. [77] in a phase I rate of unexplained anemia. Blood. 2004;104:2263-2268.
10. Yao S, Hahn T, Zhang Y, et al. Unrelated donor allogeneic hematopoietic
dose escalating trial. Monthly doses of 32 mg/m2 daily for cell transplantation is underused as a curative therapy in eligible pa-
5 days beginning 40 days after HCT resulted in 1-year OS and tients from the United States. Biol Blood Marrow Transplant. 2013;19:
event-free survival rates of 77% and 58%, respectively. 1459-1464.
11. Estey E, de Lima M, Tibes R, et al. Prospective feasibility analysis of
Azacitidine has also shown some activity when used in reduced-intensity conditioning (RIC) regimens for hematopoietic stem
combination with donor lymphocyte infusions in relapsed cell transplantation (HSCT) in elderly patients with acute myeloid
AML/MDS patients [80]. As maintenance therapy after leukemia (AML) and high-risk myelodysplastic syndrome (MDS). Blood.
2007;109:1395-1400.
transplantation, azacitidine appears to be well tolerated with
12. Majhail NS, Lazarus HM. Many are called but few are chosen: under-
no impact on GVHD. utilization of unrelated donor transplantation. Biol Blood Marrow
An ongoing randomized trial (NCT00887068) is com- Transplant. 2013;19:1414-1415.
paring azacitidine versus current standard of care after 13. Mawad R, Gooley TA, Sandhu V, et al. Frequency of allogeneic
hematopoietic cell transplantation among patients with high- or
allogeneic HCT. Lenalidomide has been tried as maintenance intermediate-risk acute myeloid leukemia in first complete remission.
therapy in del(5q) MDS/AML but results in increased rates of J Clin Oncol. 2013;31:3883-3888.
S. Mukherjee et al. / Biol Blood Marrow Transplant 21 (2015) 412e420 419

14. Alessandrino EP, Della Porta MG, et al. WHO classification and WPSS myeloid leukemia or myelodysplasia receiving allogeneic hematopoi-
predict posttransplantation outcome in patients with myelodysplastic etic cell transplantation. J Clin Oncol. 2007;25:4246-4254.
syndrome: a study from the Gruppo Italiano Trapianto di Midollo 36. Muffly LS, Boulukos M, Swanson K, et al. Pilot study of comprehensive
Osseo (GITMO). Blood. 2008;112:895-902. geriatric assessment (CGA) in allogeneic transplant: CGA captures a
15. Della Porta MG, Alessandrino EP, Bacigalupo A, et al., Gruppo Italiano high prevalence of vulnerabilities in older transplant recipients. Biol
Trapianto di Midollo Osseo. Predictive factors for the outcome of Blood Marrow Transplant. 2013;19:429-434.
allogeneic transplantation in patients with MDS stratified according to 37. Trottier BJ, Burns LJ, DeFor TE, et al. Association of iron overload with
the revised IPSS-R. Blood. 2014;123:2333-2342. allogeneic hematopoietic cell transplantation outcomes: a prospective
16. Van Gelder M, Schetelig J, Volin L, et al. Monosomal karyotype predicts cohort study using R2-MRI-measured liver iron content. Blood. 2013;
poor outcome for MDS/sAML patients with chromosome 7 abnormal- 122:1678-1684.
ities after allogeneic stem cell transplantation for MDS/sAML: a study 38. de Witte T, Brand R, van Biezen A, et al. Allogeneic stem cell trans-
of the MDS subcommittee of the Chronic Leukemia Working Party of plantation for patients with refractory anaemia with matched related
the European Group for Blood and Marrow Transplantation (EBMT). and unrelated donors: delay of the transplant is associated with infe-
ASH Annual Meeting Abstracts 2009. Available at: http://abstract- rior survival. Br J Haematol. 2009;146:627-636.
s.hematologylibrary.org/cgi/content/abstract/ashmtg;114/22/293 39. Deeg HJ, Storer B, Slattery JT, et al. Conditioning with targeted busulfan
17. Gyurkocza B, Deeg HJ. Allogeneic hematopoietic cell transplantation for and cyclophosphamide for hemopoietic stem cell transplantation from
MDS: for whom, when and how? Blood Rev. 2012;26:247-254. related and unrelated donors in patients with myelodysplastic syn-
18. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions drome. Blood. 2002;100:1201-1207.
in 944 patients with myelodysplastic syndromes. Leukemia. 2014; 40. Cutler CS, Lee SJ, Greenberg P, et al. A decision analysis of allogeneic
28(2):241-247. bone marrow transplantation for the myelodysplastic syndromes:
19. Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point delayed transplantation for low-risk myelodysplasia is associated with
mutations in myelodysplastic syndromes. N Engl J Med. 2011;364: improved outcome. Blood. 2004;104:579-585.
2496-2506. 41. Koreth J, Pidala J, Perez WS, et al. Role of reduced-intensity condi-
20. Bejar R, Stevenson KE, Caughey BA, et al. Validation of a prognostic tioning allogeneic hematopoietic stem-cell transplantation in older
model and the impact of mutations in patients with lower-risk mye- patients with de novo myelodysplastic syndromes: an international
lodysplastic syndromes. J Clin Oncol. 2012;30:3376-3382. collaborative decision analysis. J Clin Oncol. 2013;31:2662-2670.
21. Papaemmanuil E, Gerstung M, Malcovati L, et al. Clinical and biological 42. Alessandrino EP, Porta MG, Malcovati L, et al. Optimal timing of allo-
implications of driver mutations in myelodysplastic syndromes. Blood. geneic hematopoietic stem cell transplantation in patients with mye-
2013;122:3616-3627. lodysplastic syndrome. Am J Hematol. 2013;88(7):581-588.
22. Thol F, Koenecke C, Kade S, et al. Prognostic effect of mutations in the 43. Brand R, Putter H, van Biezen A, et al. Comparison of allogeneic stem cell
splicing gene machinery in 339 patients with MDS or secondary AML transplantation and non-transplant approaches in elderly patients with
following MDS after allogeneic hematopoietic stem cell transplantation. advanced myelodysplastic syndrome: optimal statistical approaches
ASH Annual Meeting Abstracts 2012. Available at: http://abstracts. and a critical appraisal of clinical results using non-randomized data.
hematologylibrary.org/cgi/content/abstract/ashmtg;120/21/357 PLoS One. 2013 Oct 7;8(10):e74368. http://dx.doi.org/10.1371/journal.-
23. Hamilton BK, Tabarroki A, Visconte V, et al. Impact of mutations in the pone.0074368. eCollection 2013.
spliceosome machinery and ring sideroblasts in patients with myeloid 44. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine
malignancies who received conventional chemotherapy or allogeneic compared with that of conventional care regimens in the treatment of
hematopoietic cell transplantation. ASH Annual Meeting Abstracts higher-risk myelodysplastic syndromes: A randomised, open-label,
2012. Available at: http://abstracts.hematologylibrary.org/cgi/content/ phase III study. Lancet Oncol. 2009;10:223-232.
abstract/ashmtg;120/21/1973 45. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient
24. Hamilton B, Tabarroki A, Elson P, et al. Impact of allogeneic hemato- outcomes in myelodysplastic syndromes: results of a phase III ran-
poietic stem cell transplant (HSCT) on patients harboring the spliceo- domized study. Cancer. 2006;106:1794-1803.
some mutation SRSF2. ASCO Annual Meeting Abstracts 2013. Available 46. Prebet T, Gore SD, Esterni B, et al. Outcome of high-risk myelodys-
at: http://meetinglibrary.asco.org/content/111577-132 plastic syndrome after azacitidine treatment failure. J Clin Oncol. 2011;
25. Litzow MR, Tarima S, Perez WS, et al. Allogeneic transplantation for 29:3322-3327.
therapy related myelodysplastic syndrome and acute myeloid leuke- 47. Itzykson R, Kosmider O, Cluzeau T, et al. Impact of TET2 mutations on
mia. Blood. 2010;115:1850-1857. response rate to azacitidine in myelodysplastic syndromes and low
26. Kröger N, Brand R, van Biezen A, et al. Risk factors for therapy-related blast count acute myeloid leukemias. Leukemia. 2011;25:1147-1152.
myelodysplastic syndrome and acute myeloid leukemia treated 48. Traina F, Visconte V, Elson P, et al. Impact of molecular mutations on
with allogeneic stem cell transplantation. Haematologica. 2009;94: treatment response to DNMT inhibitors in myelodysplasia and related
542-549. neoplasms. Leukemia. 2014;28:78-87.
27. Nardi V, Winkfield KM, Ok CY, et al. Acute myeloid leukemia and 49. Saber W, Cutler CS, Nakamura R, et al. Impact of donor source on he-
myelodysplastic syndromes after radiation therapy are similar to de matopoietic cell transplantation outcomes for patients with myelo-
novo disease and differ from other therapy-related myeloid neoplasms. dysplastic syndromes (MDS). Blood. 2013;122:1974-1982.
J Clin Oncol. 2012;30:2340-2347. 50. Kröger N, Zabelina T, Guardiola P, et al. Allogeneic stem cell trans-
28. Mukherjee S, Reddy CA, Ciezki JP, et al. Risk of developing myelodys- plantation for older advanced MDS patients: improved survival with
plastic syndromes (MDS) in prostate cancer patients definitively young unrelated donor in comparison with HLA-identical siblings.
treated with radiation. J Natl Cancer Inst. 2014 Mar;106(3):djt462. http: Leukemia. 2013;27:604-609.
//dx.doi.org/10.1093/jnci/djt462. Epub 2014 Feb 27. 51. Alousi AM, Le-Rademacher J, Saliba RM, et al. Who is the better donor
29. Larson RA, Le Beau MM. Prognosis and therapy when acute promye- for older hematopoietic transplant recipients: an older-aged sibling or
locytic leukemia and other “good risk” acute myeloid leukemias occur a young, matched unrelated volunteer? Blood. 2013;121:2567-2573.
as a therapy-related myeloid neoplasm. Mediterr J Hematol Infect Dis. 52. Anasetti C, Logan BR, Lee SJ, et al. Peripheral-blood stem cells versus
2012;3:e2011032. bone marrow from unrelated donors. N Engl J Med. 2012;367:1487-1496.
30. Sorror ML, Sandmaier BM, Storer BE, et al. Long-term outcomes among 53. Majhail NS, Brunstein CG, Shanley R, et al. Reduced-intensity he-
older patients following nonmyeloablative conditioning and allogeneic matopoietic cell transplantation in older patients with AML/MDS:
hematopoietic cell transplantation for advanced hematologic malig- umbilical cord blood is a feasible option for patients without HLA-
nancies. JAMA. 2011;306:1874-1883. matched sibling donors. Bone Marrow Transplant. 2012;47:494-498.
31. McClune BL, Weisdorf DJ, Pedersen TL, et al. Effect of age on outcome of 54. Ballen KK, Klein JP, Pedersen TL, et al. Relationship of race/ethnicity and
reduced-intensity hematopoietic cell transplantation for older patients survival after single umbilical cord blood transplantation for adults and
with acute myeloid leukemia in first complete remission or with children with leukemia and myelodysplastic syndromes. Biol Blood
myelodysplastic syndrome. J Clin Oncol. 2010;28:1878-1887. Marrow Transplant. 2012;18:903-912.
32. Lim Z, Brand R, Martino R, et al. Allogeneic hematopoietic stem cell 55. Kelly SS, Sola CB, de Lima M, Shapll E. Ex vivo expansion of cord blood.
transplantation for patients 50 years or older with myelodysplastic Bone Marrow Transplant. 2009;44:673-681.
syndromes or secondary acute myeloid leukemia. J Clin Oncol. 2010;28: 56. Barker JN, Weisdorf DJ, Defor TE, et al. Transplantation of 2 partially
405-411. HLA-matched umbilical cord blood units to enhance engraftment in
33. Brunner AM, Kim HT, Coughlin E, et al. Outcomes in patients age 70 or adults with hematologic malignancy. Blood. 2005;105:1343-1347.
older undergoing allogeneic hematopoietic stem cell transplantation 57. Bashey A, Zhang X, Sizemore CA, et al. T-cell-replete HLA-haploidentical
for hematologic malignancies. Biol Blood Marrow Transplant. 2013;19: hematopoietic transplantation for hematologic malignancies using
1374-1380. post-transplantation cyclophosphamide results in outcomes equivalent
34. Sorror ML, Maris MB, Storb R, et al. Hematopoietic cell transplantation to those of contemporaneous HLA-matched related and unrelated donor
(HCT)-specific comorbidity index: a new tool for risk assessment transplantation. J Clin Oncol. 2013;31:1310-1316.
before allogeneic HCT. Blood. 2005;106:2912-2919. 58. Warlick ED, Cioc A, DeFor T, et al. Allogeneic stem cell transplantation
35. Sorror ML, Sandmaier BM, Storer BE, et al. Comorbidity and disease for adults with myelodysplastic syndromes: importance of pretrans-
status based risk stratification of outcomes among patients with acute plant disease burden. Biol Blood Marrow Transplant. 2009;15:30-38.
420 S. Mukherjee et al. / Biol Blood Marrow Transplant 21 (2015) 412e420

59. Martino R, Iacobelli S, Brand R, et al. Retrospective comparison of 70. Lee JH, Joo YD, Kim H, et al. Randomized trial of myeloablative con-
reduced-intensity conditioning and conventional high-dose condi- ditioning regimens: busulfan plus cyclophosphamide versus busulfan
tioning for allogeneic hematopoietic stem cell transplantation using plus fludarabine. J Clin Oncol. 2013;31:701-709.
HLA-identical sibling donors in myelodyspl myelodysplastic syn- 71. Alyea EP, Kim HT, Ho V, et al. Impact of conditioning regimen intensity
dromes. Blood. 2006;108:836-846. on outcome of allogeneic hematopoietic cell transplantation for
60. Scott BL, Storer B, Loken MR, et al. Pretransplantation induction advanced acute myelogenous leukemia and myelodysplastic syn-
chemotherapy and posttransplantation relapse in patients with drome. Biol Blood Marrow Transplant. 2006;12:1047-1055.
advanced myelodysplastic syndrome. Biol Blood Marrow Transplant. 72. Shimoni A, Hardan I, Shem-Tov N, et al. Allogeneic hematopoietic
2005;11:65-73. stem-cell transplantation in AML and MDS using myeloablative versus
61. Damaj G, Duhamel A, Robin M, et al. Impact of azacitidine before reduced-intensity conditioning: the role of dose intensity. Leukemia.
allogeneic stem-cell transplantation for myelodysplastic syndromes: a 2006;20:322-328.
study by the Société Française de Greffe de Moelle et de Thérapie- 73. Luger SM, Ringdén O, Zhang MJ, et al. Similar outcomes using mye-
Cellulaire and the Groupe-Francophone des Myélodysplasies. J Clin loablative vs reduced-intensity allogeneic transplant preparative regi-
Oncol. 2012;30:4533-4540. mens for AML or MDS. Bone Marrow Transplant. 2012;47:203-211.
62. Oran B, Kongtim P, de Lima M, et al. Prior hypomethylating agents or 74. Hallemeier CL, Girgis MD, Blum WG, et al. Long-term remissions in pa-
chemotherapy does not improve the outcome of allogeneic hemato- tients with myelodysplastic syndrome and secondary acute myeloge-
poietic transplantation for high risk MDS. ASH Annual Meeting Ab- nous leukemia undergoing allogeneic transplantation following a
stracts 2013. Available at: http://bloodjournal.hematologylibrary. reduced intensity conditioning regimen of 550 cGy total body irradiation
org/content/122/21/305.abstract and cyclophosphamide. Biol Blood Marrow Transplant. 2006;12:749-757.
63. Jabbour E, Mathisen MS, Garcia-Manero G, et al. Allogeneic hemato- 75. Alatrash G, de Lima M, Hamerschlak N, et al. Myeloablative reduced-
poietic stem cell transplantation versus hypomethylating agents in toxicity i.v. busulfan-fludarabine and allogeneic hematopoietic stem
patients with myelodysplastic syndrome: a retrospective case-control cell transplant for patients with acute myeloid leukemia or myelo-
study. Am J Hematol. 2013;88:198-200. dysplastic syndrome in the sixth through eighth decades of life. Biol
64. Gerds AT, Gooley TA, Estey EH, et al. Pretransplantation therapy with Blood Marrow Transplant. 2011;17:1490-1496.
azacitidine vs induction chemotherapy and posttransplantation 76. Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treatment of
outcome in patients with MDS. Biol Blood Marrow Transplant. 2012; imminent relapse in MDS or AML patients after allogeneic HSCT:
18(8):1211-1218. results of the RELAZA trial. Leukemia. 2012;26:381-389.
65. Runde V, de Witte T, Arnold R, et al. Bone marrow transplantation from 77. de Lima M, Giralt S, Thall PF, et al. Maintenance therapy with low-dose
HLA-identical siblings as first-line treatment in patients with myelo- azacitidine after allogeneic hematopoietic stem cell transplantation for
dysplastic syndromes: early transplantation is associated with recurrent acute myelogenous leukemia or myelodysplastic syndrome.
improved outcome. Chronic Leukemia Working Party of the European Cancer. 2010;116:5420-5431.
Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 78. Schneidawind D, Pierini A, Negrin RS. Regulatory T cells and natural
1998;21:255-261. killer T cells for modulation of GVHD following allogeneic hemato-
66. Khabori MA, El-Emary M, Xu W, et al. Impact of intensity of condi- poietic cell transplantation. Blood. 2013;122:3116-3121.
tioning therapy in patients aged 40-60 years with AML/myelodys- 79. Goodyear O, Agathanggelou A, Novitzky-Basso I, et al. Induction of a
plastic syndrome undergoing allogeneic transplantation. Bone Marrow CD81 T-cell response to the MAGE cancer testis antigen by combined
Transplant. 2011;46:516-522. treatment with azacitidine and sodium valproate in patients with acute
67. Kashyap A, Wingard J, Cagnoni P, et al. Intravenous versus oral myeloid leukemia and myelodysplasia. Blood. 2010;116:1908-1918.
busulfan as part of a busulfan/cyclophosphamide preparative regimen 80. Schroeder T, Czibere A, Kroger N, et al. Phase II study of azacitidine
for allogeneic hematopoietic stem cell transplantation: decreased (Vidaza (R), Aza) and donor lymphocyte infusions (DLI) as first salvage
incidence of hepatic venoocclusive disease (HVOD), HVOD-related therapy in patients with acute myeloid leukemia (AML) or myelodys-
mortality, and overall 100-day mortality. Biol Blood Marrow Trans- plastic syndromes (MDS) relapsing after allogeneic hematopoietic stem
plant. 2002;8:493-500. cell transplantation (allo-SCT): final results from the AZARELA trial
68. Copelan EA, Hamilton BK, Avalos B, et al. Better leukemia-free and overall (NCT-00795548). Leukemia. 2013;27:1910-1913.
survival in AML in first remission following cyclophosphamide in com- 81. Sockel K, Bornhaeuser M, Mischak-Weissinger E, et al. Lenalidomide
bination with busulfan compared to TBI. Blood. 2013;122:3863-3870. maintenance after allogeneic HSCT seems to trigger acute graft-versus-
69. Bredeson C, Lerademacher J, Kato K, et al. Prospective cohort study host disease in patients with high-risk myelodysplastic syndromes or
comparing intravenous busulfan to total body irradiation in hemato- acute myeloid leukemia and del(5q): results of the LENAMAINT trial.
poietic cell transplantation. Blood. 2013;122:3871-3878. Haematologica. 2012;97:e34-e35.

You might also like